Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (148)

Search Parameters:
Keywords = oral–gut interactions

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
30 pages, 1428 KiB  
Review
The Oral–Gut Microbiota Axis Across the Lifespan: New Insights on a Forgotten Interaction
by Domenico Azzolino, Margherita Carnevale-Schianca, Luigi Santacroce, Marica Colella, Alessia Felicetti, Leonardo Terranova, Roberto Carlos Castrejón-Pérez, Franklin Garcia-Godoy, Tiziano Lucchi and Pier Carmine Passarelli
Nutrients 2025, 17(15), 2538; https://doi.org/10.3390/nu17152538 - 1 Aug 2025
Viewed by 243
Abstract
The oral–gut microbiota axis is a relatively new field of research. Although most studies have focused separately on the oral and gut microbiota, emerging evidence has highlighted that the two microbiota are interconnected and may influence each other through various mechanisms shaping systemic [...] Read more.
The oral–gut microbiota axis is a relatively new field of research. Although most studies have focused separately on the oral and gut microbiota, emerging evidence has highlighted that the two microbiota are interconnected and may influence each other through various mechanisms shaping systemic health. The aim of this review is therefore to provide an overview of the interactions between oral and gut microbiota, and the influence of diet and related metabolites on this axis. Pathogenic oral bacteria, such as Porphyromonas gingivalis and Fusobacterium nucleatum, can migrate to the gut through the enteral route, particularly in individuals with weakened gastrointestinal defenses or conditions like gastroesophageal reflux disease, contributing to disorders like inflammatory bowel disease and colorectal cancer. Bile acids, altered by gut microbes, also play a significant role in modulating these microbiota interactions and inflammatory responses. Oral bacteria can also spread via the bloodstream, promoting systemic inflammation and worsening some conditions like cardiovascular disease. Translocation of microorganisms can also take place from the gut to the oral cavity through fecal–oral transmission, especially within poor sanitary conditions. Some metabolites including short-chain fatty acids, trimethylamine N-oxide, indole and its derivatives, bile acids, and lipopolysaccharides produced by both oral and gut microbes seem to play central roles in mediating oral–gut interactions. The complex interplay between oral and gut microbiota underscores their crucial role in maintaining systemic health and highlights the potential consequences of dysbiosis at both the oral and gastrointestinal level. Some dietary patterns and nutritional compounds including probiotics and prebiotics seem to exert beneficial effects both on oral and gut microbiota eubiosis. A better understanding of these microbial interactions could therefore pave the way for the prevention and management of systemic conditions, improving overall health outcomes. Full article
(This article belongs to the Special Issue Exploring the Lifespan Dynamics of Oral–Gut Microbiota Interactions)
Show Figures

Figure 1

29 pages, 1209 KiB  
Review
The Oral–Gut Microbiota Axis as a Mediator of Frailty and Sarcopenia
by Domenico Azzolino, Margherita Carnevale-Schianca, Lucrezia Bottalico, Marica Colella, Alessia Felicetti, Simone Perna, Leonardo Terranova, Franklin Garcia-Godoy, Mariangela Rondanelli, Pier Carmine Passarelli and Tiziano Lucchi
Nutrients 2025, 17(15), 2408; https://doi.org/10.3390/nu17152408 - 23 Jul 2025
Viewed by 622
Abstract
Traditionally studied in isolation, the oral and gut microbiota are now being recognized as interconnected through anatomical and physiological pathways forming a dynamic “oral–gut microbiota axis”. Both oral and gut microbiota undergo changes with aging, characterized by a decline in microbial diversity and [...] Read more.
Traditionally studied in isolation, the oral and gut microbiota are now being recognized as interconnected through anatomical and physiological pathways forming a dynamic “oral–gut microbiota axis”. Both oral and gut microbiota undergo changes with aging, characterized by a decline in microbial diversity and a shift toward potentially harmful species. The aim of this review is, therefore, to provide an overview of oral–gut communications in mediating frailty and sarcopenia. PubMed, EMBASE and Scopus databases were searched for relevant articles. We limited our search to manuscripts published in the English language. Interactions between oral and gut microbiota occur mainly through three pathways namely the enteral, the bloodstream and the fecal-oral routes. Alterations in the oral–gut microbiota axis contribute to chronic low-grade inflammation (i.e., “inflamm-ageing”) and mitochondrial dysfunction, key mechanisms underlying frailty and sarcopenia. Microbial metabolites, such as short-chain fatty acids and modified bile acids, appear to play an emerging role in influencing microbial homeostasis and muscle metabolism. Furthermore, poor oral health associated with microbial dysbiosis may contribute to altered eating patterns that negatively impact gut microbiota eubiosis, further exacerbating muscle decline and the degree of frailty. Strategies aimed at modulating the microbiota, such as healthy dietary patterns with reduced consumption of ultra-processed foods, refined carbohydrates and alcohol, ensuring an adequate protein intake combined with physical exercise, as well as supplementation with prebiotics, probiotics, and omega-3 polyunsaturated fatty acids, are increasingly recognized as promising interventions to improve both oral and gut microbiota health, with beneficial effects on frailty and sarcopenia. A better understanding of the oral–gut microbiota axis offers promising insights into nutritional interventions and therapeutic strategies for the age-related muscle decline, frailty and systemic health maintenance. Full article
(This article belongs to the Special Issue Addressing Malnutrition in the Aging Population)
Show Figures

Figure 1

34 pages, 6295 KiB  
Article
ROS/Enzyme Dual-Responsive Drug Delivery System for Targeted Colorectal Cancer Therapy: Synergistic Chemotherapy, Anti-Inflammatory, and Gut Microbiota Modulation
by Xin Zhang, Ruonan Lian, Bingbing Fan, Lei Meng, Pengxia Zhang, Yu Zhang and Weitong Sun
Pharmaceutics 2025, 17(7), 940; https://doi.org/10.3390/pharmaceutics17070940 - 21 Jul 2025
Viewed by 434
Abstract
Objectives: Colorectal cancer (CRC) is a leading cause of cancer-related mortality, driven by chronic inflammation, gut microbiota dysbiosis, and complex tumor microenvironment interactions. Current therapies are limited by systemic toxicity and poor tumor accumulation. This study aimed to develop a ROS/enzyme dual-responsive oral [...] Read more.
Objectives: Colorectal cancer (CRC) is a leading cause of cancer-related mortality, driven by chronic inflammation, gut microbiota dysbiosis, and complex tumor microenvironment interactions. Current therapies are limited by systemic toxicity and poor tumor accumulation. This study aimed to develop a ROS/enzyme dual-responsive oral drug delivery system, KGM-CUR/PSM microspheres, to achieve precise drug release in CRC and enhance tumor-specific drug accumulation, which leverages high ROS levels in CRC and the β-mannanase overexpression in colorectal tissues. Methods: In this study, we synthesized a ROS-responsive prodrug polymer (PSM) by conjugating polyethylene glycol monomethyl ether (mPEG) and mesalazine (MSL) via a thioether bond. CUR was then encapsulated into PSM using thin-film hydration to form tumor microenvironment-responsive micelles (CUR/PSM). Subsequently, konjac glucomannan (KGM) was employed to fabricate KGM-CUR/PSM microspheres, enabling targeted delivery for colorectal cancer therapy. The ROS/enzyme dual-response properties were confirmed through in vitro drug release studies. Cytotoxicity, cellular uptake, and cell migration were assessed in SW480 cells. In vivo efficacy was evaluated in AOM/DSS-induced CRC mice, monitoring tumor growth, inflammatory markers (TNF-α, IL-1β, IL-6, MPO), and gut microbiota composition. Results: In vitro drug release studies demonstrated that KGM-CUR/PSM microspheres exhibited ROS/enzyme-responsive release profiles. CUR/PSM micelles demonstrated significant anti-CRC efficacy in cytotoxicity assays, cellular uptake studies, and cell migration assays. In AOM/DSS-induced CRC mice, KGM-CUR/PSM microspheres significantly improved survival and inhibited CRC tumor growth, and effectively reduced the expression of inflammatory cytokines (TNF-α, IL-1β, IL-6) and myeloperoxidase (MPO). Histopathological and microbiological analyses revealed near-normal colon architecture and microbial diversity in the KGM-CUR/PSM group, confirming the system’s ability to disrupt the “inflammation-microbiota-tumor” axis. Conclusions: The KGM-CUR/PSM microspheres demonstrated a synergistic enhancement of anti-tumor efficacy by inducing apoptosis, alleviating inflammation, and modulating the intestinal microbiota, which offers a promising stimuli-responsive drug delivery system for future clinical treatment of CRC. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

30 pages, 2043 KiB  
Review
Berberine as a Bioactive Alkaloid: Multi-Omics Perspectives on Its Role in Obesity Management
by Bartłomiej Zieniuk and Magdalena Pawełkowicz
Metabolites 2025, 15(7), 467; https://doi.org/10.3390/metabo15070467 - 9 Jul 2025
Viewed by 888
Abstract
Berberine, a bioactive isoquinoline alkaloid derived from medicinal plants such as Berberis and Coptis species, shows significant promise for managing obesity and associated metabolic disorders. This review synthesizes evidence on its modulation of AMP-activated protein kinase (AMPK) signaling, gut microbiota composition, lipid metabolism, [...] Read more.
Berberine, a bioactive isoquinoline alkaloid derived from medicinal plants such as Berberis and Coptis species, shows significant promise for managing obesity and associated metabolic disorders. This review synthesizes evidence on its modulation of AMP-activated protein kinase (AMPK) signaling, gut microbiota composition, lipid metabolism, and adipokine networks, elucidating how these actions converge to suppress adipogenesis and improve insulin sensitivity. Metabolomic profiling reveals critical shifts in bile acid metabolism, short-chain fatty acid production, and mitochondrial function. Recent studies also highlight berberine’s anti-inflammatory effects and regulatory influence on glucose homeostasis. Despite its promise, challenges in oral bioavailability and drug interactions necessitate the development of advanced delivery strategies. We further discuss nanoformulations and multi-omics approaches, which integrate data from genomics, transcriptomics, proteomics, and metabolomics, provide new insights into berberine’s mechanisms, and may guide personalized therapeutic applications. While promising, further studies are needed to validate these findings in humans and translate them into effective clinical strategies. Full article
Show Figures

Figure 1

13 pages, 2419 KiB  
Article
Modulation of Gut Microbial Composition by Lactobacillus delbrueckii subsp. lactis CKDB001 Supplementation in a High-Fat-Diet-Induced Obese Mice
by Jaeryang Chu, Chae-Won No, Hyunchae Joung, Kyung Hwan Kim, Chang Hun Shin, Jisu Lee and Jung-Heun Ha
Nutrients 2025, 17(13), 2251; https://doi.org/10.3390/nu17132251 - 7 Jul 2025
Viewed by 424
Abstract
Background/Objectives: Lactobacillus delbrueckii subsp. lactis CKDB001 (LL) has demonstrated anti-inflammatory, antioxidant, and lipid-regulatory effects in vitro and in vivo, including attenuation of hepatic steatosis and modulation of lipid metabolism. Given the known interactions between host metabolism and gut microbiota, these findings suggest [...] Read more.
Background/Objectives: Lactobacillus delbrueckii subsp. lactis CKDB001 (LL) has demonstrated anti-inflammatory, antioxidant, and lipid-regulatory effects in vitro and in vivo, including attenuation of hepatic steatosis and modulation of lipid metabolism. Given the known interactions between host metabolism and gut microbiota, these findings suggest a potential role for LL in modulating microbial composition under conditions of diet-induced obesity. This study aimed to investigate the microbiome-related effects of LL using an established murine model. To evaluate the effect of LL supplementation on gut microbial composition and predict microbial metabolic functions in mice with high-fat diet-induced obesity. Methods: Male C57BL/6J mice were fed a high-fat diet and administered LL orally for 12 weeks. Fecal samples were collected and analyzed using 16S rRNA gene sequencing. Microbial taxonomic profiles were assessed using linear discriminant analysis effect size, and functional predictions were performed using PICRUSt2. Results: LL supplementation significantly altered the gut microbiota by increasing the relative abundance of Lactobacillus and other commensal taxa while reducing the prevalence of pro-inflammatory genera such as Alistipes and Bilophila. Functional prediction analysis revealed a downregulation of lipopolysaccharide and ADP-L-glycero-β-D-manno-heptose biosynthesis pathways. Microbial functions associated with carbohydrate metabolism and short-chain fatty acid production were enriched in the LL-treated group. Conclusions: LL modulated gut microbial composition and suppressed pro-inflammatory microbial pathways while enhancing beneficial metabolic functions in high-fat diet-fed mice. These findings support the potential of LL as a safe and effective microbiota-targeted probiotic for managing obesity-related metabolic disorders. Full article
(This article belongs to the Special Issue Prebiotics and Probiotics in Metabolism Disorder—2nd Edition)
Show Figures

Figure 1

21 pages, 506 KiB  
Article
Fermented Milk Supplemented with Sodium Butyrate and Inulin: Physicochemical Characterization and Probiotic Viability Under In Vitro Simulated Gastrointestinal Digestion
by Katarzyna Szajnar, Małgorzata Pawlos, Magdalena Kowalczyk, Julita Drobniak and Agata Znamirowska-Piotrowska
Nutrients 2025, 17(13), 2249; https://doi.org/10.3390/nu17132249 - 7 Jul 2025
Viewed by 614
Abstract
Background/Objectives: Probiotics are increasingly recognized for their role in managing gastrointestinal disorders through modulation of gut microbiota. Restoring microbial balance remains a therapeutic challenge. Recent strategies combine probiotics, inulin, and sodium butyrate as synergistic agents for gut health. This study aimed to evaluate [...] Read more.
Background/Objectives: Probiotics are increasingly recognized for their role in managing gastrointestinal disorders through modulation of gut microbiota. Restoring microbial balance remains a therapeutic challenge. Recent strategies combine probiotics, inulin, and sodium butyrate as synergistic agents for gut health. This study aimed to evaluate the effects of milk supplementation with inulin and sodium butyrate on physicochemical properties, sensory characteristics, and the survival of selected probiotic strains during in vitro simulated gastrointestinal digestion. Methods: Fermented milk samples were analyzed for color, pH, titratable acidity, and syneresis. A trained sensory panel evaluated aroma, texture, and acceptability. Samples underwent a standardized in vitro digestion simulating oral, gastric, and intestinal phases. Viable probiotic cells were counted before digestion and at each stage, and survival rates were calculated. Results: Physicochemical and sensory attributes varied depending on probiotic strain and supplementation. Inulin and the inulin–sodium butyrate combination influenced syneresis and acidity. Lacticaseibacillus casei 431 and Lactobacillus johnsonii LJ samples showed the highest viable counts before digestion. Two-way ANOVA confirmed that probiotic strain, supplementation type, and their interactions significantly affected bacterial survival during digestion (p < 0.05). Conclusions: The addition of inulin and sodium butyrate did not impair probiotic viability under simulated gastrointestinal conditions. The effects on product characteristics were strain-dependent (Bifidobacterium animalis subsp. lactis BB-12, L. casei 431, L. paracasei L26, L. acidophilus LA-5, L. johnsonii LJ). These findings support the use of inulin–butyrate fortification in dairy matrices to enhance the functional potential of probiotic foods targeting gut health. Full article
(This article belongs to the Special Issue Probiotics, Postbiotics, Gut Microbiota and Gastrointestinal Health)
Show Figures

Figure 1

35 pages, 5123 KiB  
Review
Prebiotic Oligosaccharides in Skin Health: Benefits, Mechanisms, and Cosmetic Applications
by Meijun Zeng, Yang Li, Jie Cheng, Jingyu Wang and Qiyu Liu
Antioxidants 2025, 14(6), 754; https://doi.org/10.3390/antiox14060754 - 18 Jun 2025
Cited by 2 | Viewed by 1409
Abstract
Prebiotic oligosaccharides have attracted significant interest in dermatology and skin health due to their ability to modulate the skin microbiome and microbiota–host interactions. This review offers a novel dual perspective, systematically examining the benefits of both oral intake and topical application of prebiotic [...] Read more.
Prebiotic oligosaccharides have attracted significant interest in dermatology and skin health due to their ability to modulate the skin microbiome and microbiota–host interactions. This review offers a novel dual perspective, systematically examining the benefits of both oral intake and topical application of prebiotic oligosaccharides, including well-established prebiotics (e.g., human milk oligosaccharides, galacto- and fructo-oligosaccharides) and emerging prebiotic candidates (e.g., gluco-oligosaccharides, chitosan-oligosaccharides, agaro-oligosaccharides). First, cutting-edge synthetic processes for producing diverse oligosaccharides and their structural chemistry are introduced. Then, we discuss in vitro studies demonstrating their efficacy in promoting skin commensals, inhibiting pathogens, and conferring protective effects, such as antioxidant, anti-inflammatory, anti-melanogenic, and wound-healing properties. Furthermore, we emphasize in vivo animal studies and clinical trials revealing that prebiotic oligosaccharides, administered orally or topically, alleviate atopic dermatitis, enhance skin hydration, attenuate acne, and protect against photo-aging by modulating skin–gut microbiota and immune responses. Mechanistically, we integrate genetic and molecular insights to elucidate how oligosaccharides mediate these benefits, including gut–skin axis crosstalk, immune regulation, and microbial metabolite signaling. Finally, we highlight current commercial applications of oligosaccharides in cosmetic formulations while addressing scientific and practical challenges, such as structure–function relationships, clinical scalability, and regulatory considerations. This review bridges mechanistic understanding with practical applications, offering a comprehensive resource for advancing prebiotic oligosaccharides-based skincare therapies. Full article
Show Figures

Figure 1

16 pages, 803 KiB  
Review
The Role of Microbiota in Upper Gastrointestinal Cancers
by Giovanni Marasco, Luigi Colecchia, Daniele Salvi, Angelo Bruni, Cecilia Capelli, Elton Dajti, Maria Raffaella Barbaro, Cesare Cremon, Vincenzo Stanghellini and Giovanni Barbara
Cancers 2025, 17(10), 1719; https://doi.org/10.3390/cancers17101719 - 21 May 2025
Viewed by 875
Abstract
The gut microbiota significantly impacts the development and progression of upper gastrointestinal (GI) cancers, including esophageal and gastric cancers. Microbial dysbiosis contributes to carcinogenesis through mechanisms such as inflammation, immune modulation, and direct DNA damage. Techniques for sampling oral, esophageal, and gastric microbiota [...] Read more.
The gut microbiota significantly impacts the development and progression of upper gastrointestinal (GI) cancers, including esophageal and gastric cancers. Microbial dysbiosis contributes to carcinogenesis through mechanisms such as inflammation, immune modulation, and direct DNA damage. Techniques for sampling oral, esophageal, and gastric microbiota vary, with standardization being essential for reliable results. Barrett’s esophagus (BE) and esophageal adenocarcinoma (EAC) are associated with an enrichment of Gram-negative bacteria, promoting inflammation and cancer progression. Esophageal squamous cell carcinoma (ESCC) also shows distinct microbial patterns, with reduced diversity and increased harmful bacteria like Porphyromonas gingivalis and Fusobacterium nucleatum. In gastric cancer (GC), Helicobacter pylori (HP) and non-HP gastric microbiota play significant roles, with diverse microbial communities contributing to cancer development through nitrate reduction, immune modulation, and inflammation. Emerging evidence highlights the role of non-HP bacteria in promoting carcinogenesis, with specific taxa like Fusobacterium nucleatum and Lactobacillus influencing tumor growth and immune evasion. Further research is needed to elucidate the complex interactions between gut microbiota and upper GI cancers, paving the way for novel diagnostic and therapeutic approaches. Understanding these microbial dynamics offers potential for microbiota-based interventions, improving the early detection, prognosis, and treatment of upper GI cancers. This comprehensive review summarizes the available evidence on the role of microbiota in upper GI oncology and the need for continued exploration in this field. Full article
(This article belongs to the Special Issue Developments in the Management of Gastrointestinal Malignancies)
Show Figures

Figure 1

19 pages, 16029 KiB  
Article
Antibiotic Cocktail Exacerbates Esomeprazole-Induced Intestinal Dysmotility While Ameliorating Gastric Dyspepsia in Mice
by Jing-Hua Wang, Song-Yi Han, Kyungjae Lee, Uijeong Han, Si-Kyung Cho and Hojun Kim
Antibiotics 2025, 14(5), 442; https://doi.org/10.3390/antibiotics14050442 - 27 Apr 2025
Viewed by 652
Abstract
Background/Objectives: Esomeprazole, a proton pump inhibitor (PPI), is commonly prescribed for gastric-acid-related disorders but has been associated with impaired gastrointestinal (GI) motility with long-term use. However, the effect of concurrent antibiotic administration on this dysfunction remains unclear. Therefore, this study aimed to investigate [...] Read more.
Background/Objectives: Esomeprazole, a proton pump inhibitor (PPI), is commonly prescribed for gastric-acid-related disorders but has been associated with impaired gastrointestinal (GI) motility with long-term use. However, the effect of concurrent antibiotic administration on this dysfunction remains unclear. Therefore, this study aimed to investigate the effects of antibiotics on esomeprazole-induced GI motility dysfunction and explore the underlying mechanisms in a mouse model. Methods: Male C57BL/6 mice were orally administered esomeprazole (160 mg/kg) five times per week for 4 weeks. Three days before initiating esomeprazole treatment, a broad-spectrum antibiotic cocktail (ABX) consisting of ampicillin (1 g/kg), neomycin (1 g/kg), metronidazole (1 g/kg), and vancomycin (0.5 g/kg) was provided in drinking water and maintained throughout the experimental period. Mosapride (3 mg/kg), a prokinetic agent, was used as a positive control. Results: Neither esomeprazole alone nor in combination with ABX affected body weight or food intake. Compared to normal controls, esomeprazole treatment significantly delayed both intestinal transit and gastric emptying. However, ABX co-administration further pronounced intestinal transit time and improved gastric motility. The potential mechanisms may involve interactions among gastric H+/K+-ATPase, CYP3A11, gastrointestinal hormones (secretin and motilin), and the gut microbiome. Conclusions: Long-term esomeprazole use can impair both gastric and intestinal motility, and ABX co-treatment further exacerbates intestinal transit delay while paradoxically enhancing gastric emptying. These findings highlight the critical role of the gut microbiota in esomeprazole-induced GI motility dysfunction and suggest that antibiotic use should be approached with caution, particularly when combined with PPI therapy. Full article
Show Figures

Figure 1

13 pages, 3805 KiB  
Article
Intestinal Microbiota Composition in Patients with Type 2 Diabetes and Effects of Oral Antidiabetics
by Ahmet Toygar Kalkan, Goknur Yorulmaz, Aysen Akalin and Ener Cagri Dinleyici
J. Clin. Med. 2025, 14(8), 2786; https://doi.org/10.3390/jcm14082786 - 17 Apr 2025
Viewed by 564
Abstract
Introduction: The cause–effect relationships between microbiota composition changes and type 2 diabetes (T2D) are complex, likely involving two-way interactions, and require further elucidation. Few studies have examined the interactions of antidiabetic drugs with the gut microbiota. This study’s goal was to evaluate [...] Read more.
Introduction: The cause–effect relationships between microbiota composition changes and type 2 diabetes (T2D) are complex, likely involving two-way interactions, and require further elucidation. Few studies have examined the interactions of antidiabetic drugs with the gut microbiota. This study’s goal was to evaluate the gut microbiota of patients with type 2 diabetes at first diagnosis and again after 12 weeks of taking oral antidiabetic drugs. Methods: We performed a fecal microbiota analysis of adult patients who recently received a T2D diagnosis and healthy adults. We compared the microbiota compositions between the T2D patients and healthy controls; we also evaluated changes from baseline to 12 weeks of treatment in the total group receiving oral antidiabetics, as well as in the subgroups receiving metformin and linagliptin. Results: The alpha diversity and beta diversity indices were different at baseline between patients with type 2 diabetes and healthy controls. The LEfSe analysis showed that, at the genus level, the Lactobacillus, Rothia, Collinsella, and Eubacterium genera increased in relative abundance in the T2D group while, at the species level, the Rothia mucilaginosa, Collinsella aerofaciens, and Eubacterium bioforme strains were found to be dominant in the T2D group. Faecalibacterium at the genus level and Faecalibacterium prausnitzii at the strain level increased in relative abundance in the T2D group after 12 weeks. After 12 weeks of intervention, the alpha diversity indices were significantly lower in the T2D group compared to the control group. At the end of the 12th week, the Gemmiger and Collinsella genera were dominant in the T2D group, with Gemmiger formicilis and Collinsella aerofaciens being dominant at the species level; in the control group, Bacteroides and Alistipes were dominant at the genus level, and Prevotella stercorea and Alistipes finegoldii were dominant. There was no difference in the LEfSe analysis results between baseline and 12 weeks of linagliptin treatment. At the strain level, Gemmiger formicilis, Ruminococcus bromii, Rothia mucilaginosa, and Lactobacillus ruminis were predominant at the start of metformin treatment; however, after 12 weeks, Collinsella aerofaciens became predominant. Conclusions: We report that there is a substantial change in the composition of the gut microbiota in patients with T2D. Oral antidiabetic treatments, especially metformin, have some beneficial effects on microbiota composition. Few studies have explored the interaction of antidiabetic drugs with the gut microbiota; further research will elucidate the clinical impact of these changes in gut microbiota composition in diabetes. Full article
(This article belongs to the Section Endocrinology & Metabolism)
Show Figures

Figure 1

44 pages, 2568 KiB  
Review
Matrix Dynamics and Microbiome Crosstalk: Matrix Metalloproteinases as Key Players in Disease and Therapy
by Paraskevi Ioannou, Elias Katsoulieris and Nikolaos A. Afratis
Int. J. Mol. Sci. 2025, 26(8), 3621; https://doi.org/10.3390/ijms26083621 - 11 Apr 2025
Viewed by 1199
Abstract
Matrix metalloproteinases (MMPs) are key enzymes involved in extracellular matrix (ECM) remodeling, regulating a wide range of cellular and immune processes in both homeostatic and pathological conditions. Host–microbiota interactions play a critical role in maintaining ECM balance; however, during dysbiosis, this regulation is [...] Read more.
Matrix metalloproteinases (MMPs) are key enzymes involved in extracellular matrix (ECM) remodeling, regulating a wide range of cellular and immune processes in both homeostatic and pathological conditions. Host–microbiota interactions play a critical role in maintaining ECM balance; however, during dysbiosis, this regulation is disrupted, leading to compromised barrier integrity, pathogen translocation into circulation, and the development of systemic diseases and cancer. This review highlights the bidirectional relationship between MMP expression/activity and microbiota dysbiosis, emphasizing tissue-specific alterations in MMP activity that contribute to disease progression. In addition, it integrates interdisciplinary evidence to illustrate the MMP-dependent mechanisms underlying various pathologies associated with oral and gut microbiome dysbiosis, including long-range effects through the gut–skin and gut–brain axes. Thus, this review introduces the emerging field of MatrixBiome, which explores the complex interactions between the ECM, microbiota, and host tissues. Finally, it also outlines therapeutic strategies to modulate MMP levels, either indirectly through microbiome-targeted approaches (e.g., prebiotics, probiotics, and postbiotics) or directly using MMP inhibitors, offering promising avenues for future clinical interventions. Full article
(This article belongs to the Special Issue The Biology and Therapeutic Potential of Metalloproteases)
Show Figures

Figure 1

30 pages, 3002 KiB  
Review
Autism Spectrum Disorder, Oral Implications, and Oral Microbiota
by Emiliana D’Angelo, Fausto Fiori, Giuseppe A. Ferraro, Assunta Tessitore, Luca Nazzaro, Rosario Serpico and Maria Contaldo
Children 2025, 12(3), 368; https://doi.org/10.3390/children12030368 - 15 Mar 2025
Cited by 3 | Viewed by 2207
Abstract
Background/Objectives: Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by difficulties in social interaction, communication, and repetitive behaviors. Recent evidence indicates a significant relationship between ASD and imbalances in microbiota, particularly in the oral and gastrointestinal areas. This review examines [...] Read more.
Background/Objectives: Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by difficulties in social interaction, communication, and repetitive behaviors. Recent evidence indicates a significant relationship between ASD and imbalances in microbiota, particularly in the oral and gastrointestinal areas. This review examines the impact of oral microbiota, self-injurious behaviors (SIB), sensory sensitivity, and dietary choices on the comorbidities associated with ASD. Methods: An extensive literature review was conducted using PubMed and Scopus. The focus was on human studies with full-text availability, utilizing search terms related to ASD, oral health, oral microbiota, and neurodevelopmental disorders. The research was evaluated for methodological quality and its relevance to the connections between microbiota, oral health, and ASD. Results: Individuals with ASD face unique oral health challenges, including injuries from self-injurious behaviors and increased sensory sensitivity, which complicate oral hygiene and care. Selective eating can lead to nutritional deficiencies and worsen oral health issues. Dysbiosis in oral and gut microbiota, marked by altered levels of acetate, propionate, and butyrate, interferes with gut-brain and oral-brain connections, contributing to behavioral and neurological symptoms. Treatment options such as probiotics, fecal microbiota transfer, and sensory integration therapies can potentially alleviate symptoms and improve quality of life. Conclusions: The relationship between ASD, oral health, and microbiota suggests a bidirectional influence through neuroinflammatory mechanisms and metabolic disturbances. Proactive strategies focusing on microbiota and dental health may help reduce comorbidities and enhance the overall management of ASD, underscoring the need for further research into microbiota–host interactions and their therapeutic potential. Full article
Show Figures

Figure 1

19 pages, 11997 KiB  
Article
Visualizing Oral Infection Dynamics of Beauveria bassiana in the Gut of Tribolium castaneum
by Lautaro Preisegger, Juan Cruz Flecha, Fiorella Ghilini, Daysi Espin-Sánchez, Eduardo Prieto, Héctor Oberti, Eduardo Abreo, Carla Huarte-Bonnet, Nicolás Pedrini and Maria Constanza Mannino
J. Fungi 2025, 11(2), 101; https://doi.org/10.3390/jof11020101 - 28 Jan 2025
Cited by 1 | Viewed by 1530 | Correction
Abstract
The ability of entomopathogenic fungi, such as Beauveria bassiana, to infect insects by penetrating their cuticle is well documented. However, some insects have evolved mechanisms to combat fungal infections. The red flour beetle (Tribolium castaneum), a major pest causing significant [...] Read more.
The ability of entomopathogenic fungi, such as Beauveria bassiana, to infect insects by penetrating their cuticle is well documented. However, some insects have evolved mechanisms to combat fungal infections. The red flour beetle (Tribolium castaneum), a major pest causing significant economic losses in stored product environments globally, embeds antifungal compounds within its cuticle as a protective barrier. Previous reports have addressed the contributions of non-cuticular infection routes, noting an increase in mortality in beetles fed with conidia. In this study, we further explore the progression and dynamics of oral exposure in the gut of T. castaneum after feeding with an encapsulated B. bassiana conidia formulation. First, we characterized the formulation surface using atomic force microscopy, observing no significant topological differences between capsules containing and not containing conidia. Confocal microscopy confirmed uniform conidia distribution within the hydrogel matrix. Then, larvae and adult insects fed with the conidia-encapsulated formulation exhibited B. bassiana distributed throughout the alimentary canal, with a higher presence of conidia before the pyloric chamber. More conidia were found in the larval midgut and hindgut compared to adults, but no germinated conidia were observed in the epithelium. These results suggest that the presence of conidia obstructs the gut, impairing the insect’s ability to ingest, process, and absorb nutrients. This disruption may weaken the host, increasing its susceptibility to infections and, ultimately, leading to death. By providing the first direct observation of fungal conidia within the alimentary canal of T. castaneum, this study highlights a novel aspect of fungal–host interaction and opens new avenues for advancing fungal-based pest control strategies by exploiting stage-specific vulnerabilities. Full article
Show Figures

Figure 1

36 pages, 2305 KiB  
Review
Dysbiosis–NK Cell Crosstalk in Pancreatic Cancer: Toward a Unified Biomarker Signature for Improved Clinical Outcomes
by Sara Fanijavadi and Lars Henrik Jensen
Int. J. Mol. Sci. 2025, 26(2), 730; https://doi.org/10.3390/ijms26020730 - 16 Jan 2025
Cited by 2 | Viewed by 1908
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with poor prognosis, primarily due to its immunosuppressive tumor microenvironment (TME), which contributes to treatment resistance. Recent research shows that the microbiome, including microbial communities in the oral cavity, gut, bile duct, and intratumoral environments, [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with poor prognosis, primarily due to its immunosuppressive tumor microenvironment (TME), which contributes to treatment resistance. Recent research shows that the microbiome, including microbial communities in the oral cavity, gut, bile duct, and intratumoral environments, plays a key role in PDAC development, with microbial imbalances (dysbiosis) promoting inflammation, cancer progression, therapy resistance, and treatment side effects. Microbial metabolites can also affect immune cells, especially natural killer (NK) cells, which are vital for tumor surveillance, therapy response and treatment-related side effects. Dysbiosis can affect NK cell function, leading to resistance and side effects. We propose that a combined biomarker approach, integrating microbiome composition and NK cell profiles, can help predict treatment resistance and side effects, enabling more personalized therapies. This review examines how dysbiosis contributes to NK cell dysfunction in PDAC and discusses strategies (e.g., antibiotics, probiotics, vaccines) to modulate the microbiome and enhance NK cell function. Targeting dysbiosis could modulate NK cell activity, improve the effectiveness of PDAC treatments, and reduce side effects. However, further research is needed to develop unified NK cell–microbiome interaction-based biomarkers for more precise and effective patient outcomes. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapies of Pancreatic Cancer: 2nd Edition)
Show Figures

Figure 1

15 pages, 3447 KiB  
Article
Therapeutic Potential of Cajanus cajan (L.) Millsp. Leaf Extract in Modulating Gut Microbiota and Immune Response for the Treatment of Inflammatory Bowel Disease
by Mingzhang Lin, Linghua Piao, Zhendong Zhao, Li Liao, Dayong Wang, Haiwen Zhang and Xiande Liu
Pharmaceuticals 2025, 18(1), 67; https://doi.org/10.3390/ph18010067 - 9 Jan 2025
Cited by 5 | Viewed by 1557
Abstract
Background/Objectives: Inflammatory bowel disease (IBD) is a persistent inflammatory condition affecting the gastrointestinal tract, distinguished by the impairment of the intestinal epithelial barrier, dysregulation of the gut microbiota, and abnormal immune responses. Cajanus cajan (L.) Millsp., traditionally used in Chinese herbal medicine [...] Read more.
Background/Objectives: Inflammatory bowel disease (IBD) is a persistent inflammatory condition affecting the gastrointestinal tract, distinguished by the impairment of the intestinal epithelial barrier, dysregulation of the gut microbiota, and abnormal immune responses. Cajanus cajan (L.) Millsp., traditionally used in Chinese herbal medicine for gastrointestinal issues such as bleeding and dysentery, has garnered attention for its potential therapeutic benefits. However, its effects on IBD remain largely unexplored. Methods: In this study, the major compounds from Cajanus cajan leaf extract (CCLE) were initially characterized by LCMS-IT-TOF. The IBD model was developed in C57BL/6 mice by administering continuous 4% (w/v) dextran sodium sulfate (DSS) aqueous solution over a period of seven days. The body weight, colon length, disease activity index (DAI), and histopathological examination using hematoxylin and eosin (H&E) staining were performed in the IBD model. The levels of the main inflammatory factors, specifically TNF-α, IL-1β, IL-6, and myeloperoxidase (MPO), were quantified by employing enzyme-linked immunosorbent assay (ELISA) kits. Additionally, the levels of tight junction proteins (ZO-1, Occludin) and oxidative stress enzymes (iNOS, SOD1, CAT) were investigated by qPCR. Subsequently, flow cytometry was employed to analyze the populations of various immune cells within the spleen, thereby assessing the impact of the CCLE on the systemic immune homeostasis of IBD mice. Finally, 16S rDNA sequencing was conducted to examine the composition and relative abundance of gut microbiota across different experimental groups. In addition, molecular docking analysis was performed to assess the interaction between the principal components of CCLE and the aryl hydrocarbon receptor (AHR). Results: We identified seven bioactive compounds in CCLE: catechin, cajachalcone, 2-hydroxy-4-methoxy-6-(2-phenylcinyl)-benzoic acid, longistylin A, longistylin C, pinostrobin, amorfrutin A, and cajaninstilbene acid. Our results demonstrated that oral administration of CCLE significantly alleviates gastrointestinal symptoms in DSS-induced IBD mice by modulating the balance of gut-derived pro- and anti-inflammatory cytokines. This modulation is associated with a functional correction in M1/M2 macrophage polarization and the Th17/Treg cell balance in splenic immune cells, as well as shifts in the populations of harmful bacteria (Erysipelatoclostridium and Staphylococcus) and beneficial bacteria (Odoribacter, unidentified Oscillospiraceae, Lachnoclostridium, and Oscillibacter) in the gut. Furthermore, cajaninstilbene acid, longistylin A, and longistylin C were identified as potential AhR agonists. Conclusions: The present results suggested that CCLE, comprising stilbenes like cajaninstilbene acid, longistylin A, and longistylin C, protects the epithelial barrier’s structure and function against DSS-induced acute IBD by restoring gut microbiota balance and systemic immune response as AhR agonists. Overall, CCLE represents a promising natural product-based therapeutic strategy for treating IBD by restoring gut microbiota balance and modulating systemic immune responses. Full article
(This article belongs to the Section Natural Products)
Show Figures

Figure 1

Back to TopTop