Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,920)

Search Parameters:
Keywords = mucosal cell

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
29 pages, 2139 KB  
Review
Overcoming Oral Cavity Barriers for Peptide Delivery Using Advanced Pharmaceutical Techniques and Nano-Formulation Platforms
by Ali A. Amer, Lewis Bingle, Amal Ali Elkordy and Cheng Shu Chaw
Biomedicines 2025, 13(11), 2735; https://doi.org/10.3390/biomedicines13112735 (registering DOI) - 8 Nov 2025
Abstract
Therapeutic peptides have gained significant attention due to their high specificity, potency, and safety profiles in treating various diseases. However, their clinical application via the oral route remains challenging. Peptides are inherently unstable in the gastrointestinal environment, where they are rapidly degraded by [...] Read more.
Therapeutic peptides have gained significant attention due to their high specificity, potency, and safety profiles in treating various diseases. However, their clinical application via the oral route remains challenging. Peptides are inherently unstable in the gastrointestinal environment, where they are rapidly degraded by proteolytic enzymes and acidic pH, leading to poor bioavailability. Additionally, their large molecular size and hydrophilicity restrict passive diffusion across the epithelial barriers of the gastrointestinal tract. These limitations have traditionally necessitated parenteral administration, which reduces patient compliance and convenience. The oral cavity, comprising the buccal and sublingual mucosa, offers a promising alternative for peptide delivery. Its rich vascularization allows for rapid systemic absorption while bypassing hepatic first-pass metabolism. Furthermore, the mucosal surface provides a relatively permeable and accessible site for drug administration. However, the oral cavities also present significant barriers: the mucosal epithelium limits permeability, the presence of saliva causes rapid clearance, and enzymes in saliva contribute to peptide degradation. Therefore, innovative strategies are essential to enhance peptide stability, retention, and permeation in this environment. Nanoparticle-based delivery systems, including lipid-based carriers such as liposomes and niosomes, as well as polymeric nanoparticles like chitosan and PLGA, offer promising solutions. These nanocarriers protect peptides from enzymatic degradation, enhance mucoadhesion to prolong residence time, and facilitate controlled release. Their size and surface properties can be engineered to improve mucosal penetration, including through receptor-mediated endocytosis or by transiently opening tight junctions. Among these, niosomes have shown high encapsulation efficiency and sustained release potential, making them particularly suitable for oral peptide delivery. Despite advances, challenges remain in translating these technologies clinically, including ensuring biocompatibility, scalable manufacturing, and patient acceptance. Nevertheless, the oral cavity’s accessibility, combined with nanotechnological innovations, offers a compelling platform for personalized, non-invasive peptide therapies that could significantly improve treatment outcomes and patient quality of life. Full article
(This article belongs to the Special Issue Advances Research on Nanomedicine)
Show Figures

Figure 1

24 pages, 9023 KB  
Article
Pentachroma O-H: A Five-Color Histological Staining Method for Enhanced Intestinal Tissue Analysis
by Emanuel-Ciprian Onica, Cristina-Stefania Dumitru, Flavia Zara, Marius Raica, Cristian Silviu Suciu, Alina Cristina Barb, Oana-Alexia Ene, Cristi Tarta and Dorin Novacescu
Int. J. Mol. Sci. 2025, 26(22), 10811; https://doi.org/10.3390/ijms262210811 - 7 Nov 2025
Abstract
Current histological staining methods for intestinal tissue analysis face limitations in simultaneously visualizing multiple tissue components, often requiring multiple sequential stains that increase processing time and tissue consumption. This proof-of-concept study aimed to define and develop a pentachromatic staining method for enhanced visualization [...] Read more.
Current histological staining methods for intestinal tissue analysis face limitations in simultaneously visualizing multiple tissue components, often requiring multiple sequential stains that increase processing time and tissue consumption. This proof-of-concept study aimed to define and develop a pentachromatic staining method for enhanced visualization of gastrointestinal tissue architecture. We developed the Pentachroma O-H method, an original protocol using readily available histological reagents (Alcian Blue pH 2.5, Weigert’s resorcin–fuchsin, Mayer’s hematoxylin, and Van Gieson’s solution) applied in an optimized sequence. The protocol was tested on healthy human ileum tissue obtained from surgical specimens as proof of concept. Thirty serial sections were stained with Pentachroma O-H and compared to adjacent sections stained with conventional hematoxylin–eosin (H&E) to document the emerging morphological characteristics of this original stain. Pentachroma O-H achieved distinct five-color differentiation in approximately 45 min: acidic mucins appeared turquoise–blue, collagen fibers red, elastic fibers black–purple, smooth muscle and erythrocyte cytoplasm yellow, and nuclei blue–black. The method clearly delineated intestinal architecture, including mucosal goblet cells, muscularis mucosae, connective tissue vasculature (parietal smooth muscle and elastic laminae), fibers, and cellular components, as well as lymphoid tissue aggregates and infiltrates, with improved contrast compared to H&E. All tissue components were simultaneously visualized in single sections with excellent morphological preservation. This first description of Pentachroma O-H demonstrates its capability to provide comprehensive ileum tissue visualization equivalent to multiple traditional special stains in a single, efficient protocol, offering significant potential advantages for gastrointestinal pathology assessment and warranting future validation studies across diverse tissue types and pathological conditions. Full article
(This article belongs to the Special Issue Molecular Research of Gastrointestinal Disease 2.0)
Show Figures

Figure 1

21 pages, 14815 KB  
Article
In Vitro Testing of Botanical Extracts as Safe and Effective Alternatives for Oral Care: A Two-Pronged Model Integrating Pathogen Control and Host Compatibility
by Nicole Beatrice Ponce, Nathalie B. Milbrandt, Md. Masud Alam, Carlene Rome M. Ledesma, Minseon Ju, Sylesh Venkataraman, Elena Draganoiu, Liliana Miinea, Yafan Li and Anna Cristina S. Samia
Oral 2025, 5(4), 89; https://doi.org/10.3390/oral5040089 - 6 Nov 2025
Abstract
Background: Commercial oral care products commonly incorporate synthetic antimicrobials such as cetylpyridinium chloride (Cetyl Cl.), L-Arginine (L-arg.), and stannous fluoride (SnF2). Although effective against oral pathogens, these agents are often associated with adverse effects including mucosal irritation, taste alteration, and disruption [...] Read more.
Background: Commercial oral care products commonly incorporate synthetic antimicrobials such as cetylpyridinium chloride (Cetyl Cl.), L-Arginine (L-arg.), and stannous fluoride (SnF2). Although effective against oral pathogens, these agents are often associated with adverse effects including mucosal irritation, taste alteration, and disruption of the oral microbiome. These limitations have spurred growing interest in safer, plant-based alternatives. In this study, we present a two-pronged in vitro oral care testing model that integrates cell assays with machine-guided quantitative microscopy analyses to assess both antibacterial efficacy and host biocompatibility of botanical extracts. Methods: Using Miswak (Salvadora persica) and Neem (Azadirachta indica) as representative natural products, we conducted antibacterial and antibiofilm testing including the evaluation of the minimum inhibitory concentration (MIC), minimum biofilm inhibitory concentration (MBIC), and minimum biofilm eradication concentration (MBEC), alongside biocompatibility assessments via MTT cell viability assays on probiotic bacteria and mammalian oral cells. To evaluate biofilm structure and disruption, we employed scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM), augmented with machine-guided Weka segmentation and automated image analysis. Results: Our findings show that Miswak and Neem extracts exhibited 75–100% antibacterial and antibiofilm efficacy against all tested bacteria, as demonstrated by cell assays and microscopy analyses, comparable to synthetic oral care agents. They also maintained ~100% viability toward commensal microbes and mammalian oral cells, whereas Cetyl Cl. and SnF2 showed dose-dependent cytotoxicity. Conclusions: This dual-assessment oral care testing model provides a comprehensive and biologically relevant framework for the discovery and screening of safe and effective natural herbal extracts in oral care applications. Full article
Show Figures

Graphical abstract

24 pages, 2296 KB  
Review
Regenerative Strategies for Vocal Fold Repair Using Injectable Materials
by Se Hyun Yeou and Yoo Seob Shin
Biomimetics 2025, 10(11), 748; https://doi.org/10.3390/biomimetics10110748 - 6 Nov 2025
Viewed by 64
Abstract
Injectable biomaterials for vocal fold disorders are being developed to provide not only mechanical reinforcement but also a regenerative microenvironment. Recent hydrogels based on hyaluronic acid (HA) derivatives, calcium hydroxylapatite and decellularized matrix scaffolds are designed to approximate the viscoelastic behavior of native [...] Read more.
Injectable biomaterials for vocal fold disorders are being developed to provide not only mechanical reinforcement but also a regenerative microenvironment. Recent hydrogels based on hyaluronic acid (HA) derivatives, calcium hydroxylapatite and decellularized matrix scaffolds are designed to approximate the viscoelastic behavior of native tissue, allow controlled degradation, and modulate local immune responses. Rather than serving merely as space-filling agents, several of these materials deliver extracellular matrix (ECM)-like biochemical signals that help maintain pliability and overcome some limitations of conventional augmentation. Experimental and early clinical studies involving growth factor delivery, stem cell-based injections, and ECM-mimetic hydrogels have demonstrated improved mucosal wave vibration and reduced fibrosis in cases of scarring. In clinical series, benefits from basic fibroblast growth factor can persist for up to 12 months. Further progress will depend on correlating material properties with objective vibratory performance to achieve lasting restoration of phonation and advance true tissue-regenerative therapy. Full article
(This article belongs to the Special Issue Biomimetic Application on Applied Bioengineering)
Show Figures

Graphical abstract

16 pages, 3586 KB  
Article
Active Factors in the Adult Pig Colon: Microbial Transplantation Versus Supplementation with Metabolites in Weaned Piglets
by Jianhao Cui, Liefa Tang, Zixuan Li, Shuang Wang, Jiayi Zhou, Huichao Yan and Xiaofan Wang
Microorganisms 2025, 13(11), 2533; https://doi.org/10.3390/microorganisms13112533 - 5 Nov 2025
Viewed by 95
Abstract
The adult pig intestinal microbiota boosts piglet intestinal and microbiome development, thereby improving growth. However, the functional bacteria, metabolites, and their region-specific intestinal roles remain to be characterized. Administration of adult colon microbiota (CM; devoid of metabolites) to piglets promoted intestinal development post-weaning, [...] Read more.
The adult pig intestinal microbiota boosts piglet intestinal and microbiome development, thereby improving growth. However, the functional bacteria, metabolites, and their region-specific intestinal roles remain to be characterized. Administration of adult colon microbiota (CM; devoid of metabolites) to piglets promoted intestinal development post-weaning, as indicated by increased intestinal mucosal weight, villus-to-crypt ratio of the ileum (p < 0.05), and stimulated mucin secretion (p < 0.05). This effect was potentially mediated by modulating beneficial microbiota, including ASV50_Prevotella 7, ASV52_Prevotella 1, and ASV81_Coprococcus 1. Adult colon-derived microbiota was found to preferentially colonize the piglet colon, supported by significantly higher bacterial loads in colonic contents. Piglets receiving adult colon supernatant (CS; without bacterial cells) showed improved feed efficiency (FE; p < 0.05), with numerically higher body weight (BW) and average daily gain (ADG) compared to the control (CON) group. Additionally, CS transplantation (CST) promoted intestinal development, potentially by modulating abundances of beneficial bacteria species, including ASV95_Turicibacter, and ASV109_Ruminococcaceae, which correlated with increased production of antioxidant and anti-inflammatory chemicals, including protocatechuic acid (PCA, p < 0.01). Adult colon-derived microbiota and metabolites enhanced intestinal development in piglets. CS supplementation improved growth and immunity, mitigating post-weaning stress potentially through enriching growth-linked bacteria (e.g., Turicibacter and Ruminococcaceae) and metabolites production (e.g., prephenate and PCA). These findings highlight these functional microbiota and metabolites as promising direct-fed microbial or metabolite additives for piglet growth and intestinal health post-weaning. Full article
(This article belongs to the Section Gut Microbiota)
Show Figures

Figure 1

16 pages, 931 KB  
Review
Gut–Liver Axis, Microbiota, Bile Acids, and Immune Response in Pathogenesis of Primary Sclerosing Cholangitis: An Overview
by Fotios S. Fousekis, Konstantinos Mpakogiannis, Georgios D. Lianos, Elisabetta Antonelli, Gabrio Bassotti and Konstantinos H. Katsanos
J. Clin. Med. 2025, 14(21), 7817; https://doi.org/10.3390/jcm14217817 - 3 Nov 2025
Viewed by 289
Abstract
Primary sclerosing cholangitis (PSC) is a chronic, immune-mediated cholestatic liver disease characterized by progressive bile duct inflammation and fibrosis. Its strong association with inflammatory bowel disease (IBD) highlights the possible role of the gut–liver axis in disease pathogenesis. Here, we review the mechanisms [...] Read more.
Primary sclerosing cholangitis (PSC) is a chronic, immune-mediated cholestatic liver disease characterized by progressive bile duct inflammation and fibrosis. Its strong association with inflammatory bowel disease (IBD) highlights the possible role of the gut–liver axis in disease pathogenesis. Here, we review the mechanisms that may contribute to the disruption of the gut–liver axis, leading to liver injury and the development of PSC. In particular, disruption of the intestinal barrier allows microbial products to enter the portal circulation, stimulating hepatic immune cells and triggering biliary inflammation. Concurrently, gut-primed lymphocytes expressing mucosal homing receptors migrate aberrantly to the liver, where they may contribute to biliary epithelial cell injury. Dysbiosis, characterized by reduced microbial diversity and the expansion of bile-tolerant and pro-inflammatory taxa, amplifies this immune activation and disturbs gut–liver homeostasis. Moreover, bile acids act as signaling molecules, regulating metabolism and immune responses through receptors such as FXR and TGR5. Dysregulation of these pathways may promote cholestasis, inflammation, and fibrosis. By understanding these interactions, we may identify novel therapeutic targets for PSC. Full article
Show Figures

Figure 1

15 pages, 3483 KB  
Article
In Vivo Iterative Adjuvant Screening Identifies an Intranasal Vaccine Formulation for Elicitation of Protective Mucosal Immune Responses Against SARS-CoV-2
by Yang Jiao, Sara H. Mahmoud, Chengjin Ye, Yuan Luo, Wei-Chiao Huang, Qinzhe Li, Shiqi Zhou, Yiting Song, Moriya Tsuji, Luis Martinez-Sobrido and Jonathan F. Lovell
Pharmaceutics 2025, 17(11), 1422; https://doi.org/10.3390/pharmaceutics17111422 - 3 Nov 2025
Viewed by 190
Abstract
Background: Intranasal (I.N.) vaccination holds promise to elicit mucosal immunity that counters respiratory pathogens at the site of infection. For subunit protein vaccines, immunostimulatory adjuvants are typically required. Methods: We screened a panel of 22 lipid-phase adjuvants to identify which ones elicited antigen-specific [...] Read more.
Background: Intranasal (I.N.) vaccination holds promise to elicit mucosal immunity that counters respiratory pathogens at the site of infection. For subunit protein vaccines, immunostimulatory adjuvants are typically required. Methods: We screened a panel of 22 lipid-phase adjuvants to identify which ones elicited antigen-specific IgA with I.N. immunization of liposome-displayed SARS-CoV-2 receptor-binding domain (RBD). Results: Initial screening showed the TLR-4 agonist Kdo2-Lipid A (KLA) effectively elicited RBD-specific IgA. A second round of screening identified further inclusion of the invariant NKT cell ligands α-Galactosylceramide (α-GalCer) and its synthetic analog 7DW8-5 as complementary adjuvants for I.N. immunization, resulting in orders-of-magnitude-greater mucosal IgA response relative to intramuscular (I.M.) immunization. The inclusion of cationic lipids conferred capacity for mucosal adhesion and maintained immune responses. In K18 hACE2 transgenic mice, vaccination significantly reduced viral replication and prevented mortality from SARS-CoV-2 challenge. Conclusions: These results point towards the potential for the use of KLA and α-GalCer for I.N. subunit vaccines. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Figure 1

16 pages, 788 KB  
Perspective
The Nallan–Nickel Effect: A Mechanistic Perspective on Burning Sensations and Lichenoid Reactions in Long-Serving Porcelain-Fused-to-Metal Restorations
by Nallan C. S. K. Chaitanya, Nada Tawfig Hashim, Vivek Padmanabhan, Md Sofiqul Islam, Rasha Babiker, Riham Mohammed and Muhammed Mustahsen Rahman
Dent. J. 2025, 13(11), 507; https://doi.org/10.3390/dj13110507 - 3 Nov 2025
Viewed by 242
Abstract
Porcelain-fused-to-metal (PFM) crowns continue to serve as a cornerstone of restorative dentistry owing to their strength, affordability, and esthetics. However, late-onset complications such as oral burning and lichenoid reactions have been observed in long-serving PFMs, suggesting complex host–material interactions that extend beyond simple [...] Read more.
Porcelain-fused-to-metal (PFM) crowns continue to serve as a cornerstone of restorative dentistry owing to their strength, affordability, and esthetics. However, late-onset complications such as oral burning and lichenoid reactions have been observed in long-serving PFMs, suggesting complex host–material interactions that extend beyond simple mechanical wear. This Perspective introduces the Nallan–Nickel Effect, a theoretical model proposing that a host- and environment-dependent threshold of bioavailable nickel ions (Ni2+), once exceeded, may trigger a neuro-immune cascade culminating in a burning phenotype. Within this framework, slow corrosion at exposed PFM interfaces releases Ni2+ into saliva and crevicular fluid, facilitating epithelial uptake and activation of innate immune sensors such as TLR4 and NLRP3. The resulting cytokine milieu (IL-1β, IL-6, TNF-α) drives NF-κB, mediated inflammation and T-cell activation, while neurogenic mediators—including nerve growth factor (NGF), substance P, and CGRP—sensitize TRPV1/TRPA1 nociceptors, establishing feedback loops of persistent burning and neurogenic inflammation. Modifying factors such as low salivary flow, acidic oral pH, mixed-metal galvanic coupling, and parafunctional stress can lower this threshold, whereas replacement with high-noble or all-ceramic materials may restore tolerance. The model generates testable predictions: elevated local free Ni2+ levels and increased expression of TLR4 and TRPV1 in symptomatic mucosa, along with clinical improvement following substitution of nickel-containing restorations. Conceptually, the Nallan–Nickel Effect reframes PFM-associated burning and lichenoid lesions as threshold-governed, neuro-immune phenomena rather than nonspecific irritations. By integrating corrosion chemistry, mucosal immunology, and sensory neurobiology, this hypothesis offers a coherent, testable framework for future translational research and patient-centered management of PFM-related complications. Full article
(This article belongs to the Section Dental Materials)
Show Figures

Graphical abstract

22 pages, 1414 KB  
Review
Distinct Molecular Mechanisms in Oral Mucosal Wound Healing: Translational Insights and Future Directions
by Priscila Chuhuaicura, Cynthia Rodríguez-Niklitschek, Gonzalo H. Oporto and Luis A. Salazar
Int. J. Mol. Sci. 2025, 26(21), 10660; https://doi.org/10.3390/ijms262110660 - 1 Nov 2025
Viewed by 433
Abstract
Oral mucosal wound healing is a rapid, precisely regulated process distinct from cutaneous repair due to the specialized anatomical, microbial, and physiological features of the oral cavity. This review outlines the sequential healing phases—hemostasis, inflammation, proliferation, and remodeling—and examines the coordinated roles of [...] Read more.
Oral mucosal wound healing is a rapid, precisely regulated process distinct from cutaneous repair due to the specialized anatomical, microbial, and physiological features of the oral cavity. This review outlines the sequential healing phases—hemostasis, inflammation, proliferation, and remodeling—and examines the coordinated roles of keratinocytes, fibroblasts, endothelial cells, and immune cell subsets in tissue restoration. Central molecular pathways, including PI3K/Akt, JAK/STAT, Ras/MAPK, TGF-β/SMAD, and Wnt/β-catenin, along with growth factors such as TGF-β, FGF, EGF, and VEGF, are discussed in relation to their regulatory influence on cell behavior and extracellular matrix dynamics. Unique intraoral factors—namely saliva-derived histatins and a distinct resident microbiota—promote accelerated re-epithelialization and attenuated fibrosis. Systemic conditions such as diabetes, aging, and tobacco exposure are identified as key modulators that compromise repair efficiency. Emerging therapeutic strategies, including stem-cell-based interventions, microbiota modulation, bioengineered scaffolds, and photobiomodulation, offer translational potential to enhance clinical outcomes in oral tissue regeneration. Full article
(This article belongs to the Special Issue Molecular Advances in Burn and Wound Healing)
Show Figures

Figure 1

18 pages, 4759 KB  
Article
Polystyrene Nanoplastics Exposure Alters Gut Microbiota and Correlates with Egg Quality Parameters in Chickens
by Xuan Hu, Yinyin Liu, Wanqiang Chen, Yinping Ma, Yanfeng Fan, Qian Zhou, Mengmeng Lei, Hongsheng Song, Min Zhao, Xiaoxu Jia, Guodong Cai, Jianchun Bian and Yushi Gao
Animals 2025, 15(21), 3154; https://doi.org/10.3390/ani15213154 - 30 Oct 2025
Viewed by 162
Abstract
NPs have become a concerning global environmental problem. Dietary exposure to NPs can cause microbial dysbiosis. However, the risks of NPs to animals, particularly poultry species such as chickens, remain poorly understood. In this study, chickens were continuously exposed to 100 nm NPs [...] Read more.
NPs have become a concerning global environmental problem. Dietary exposure to NPs can cause microbial dysbiosis. However, the risks of NPs to animals, particularly poultry species such as chickens, remain poorly understood. In this study, chickens were continuously exposed to 100 nm NPs via dietary inclusion from 18 weeks of age for 120 days to evaluate the effects of NPs on intestinal health. We found that NPs accumulated in chicken intestinal tissues, leading to adverse alterations in the intestinal mucosal structure, such as villus atrophy and goblet cell depletion, and significantly altering intestinal length. The 16S rRNA sequencing revealed significant gut microbiota dysbiosis, characterized by a loss of diversity and shifts in key bacterial groups. Functional predictions of the microbiota revealed impairments in metabolic pathways, especially carbohydrate and amino acid metabolism. Furthermore, network analysis showed that microbial interactions were disrupted and key functional hubs were lost. Most importantly, NPs exposure led to a significant decline in egg quality parameters, including eggshell thickness and strength, yolk color, weight, shape index, and Haugh units. Correlation analyses connected specific taxa, such as Methanobrevibacter, Rikenellaceae_RC9_gut_group, and Prevotellaceae_UCG-001, to intestinal damage and declines in egg quality. These findings provide a scientific basis for assessing the health risks of NPs in animals and offer insights into the development of gut health interventions. Full article
Show Figures

Figure 1

23 pages, 6518 KB  
Article
Hyocholic Acid Species as the Key Modulator for Cecal Epithelial Homeostasis in Low-Birth-Weight Piglets
by Chang Yin, Xuan Liu, Wei Fang, Qingshi Meng, Xiaohui Feng, Weidong Zhang, Guoqi Dang, Ruqing Zhong, Liang Chen, Zirong Wang and Hongfu Zhang
Nutrients 2025, 17(21), 3415; https://doi.org/10.3390/nu17213415 - 30 Oct 2025
Viewed by 259
Abstract
Background: Low birth weight (LBW) is correlated with gut microbiota dysbiosis and intestinal barrier function disruption, increasing susceptibility to enteric diseases. These alterations underscore the critical need to identify key regulators of gut homeostasis, among which bile acids are increasingly recognized as [...] Read more.
Background: Low birth weight (LBW) is correlated with gut microbiota dysbiosis and intestinal barrier function disruption, increasing susceptibility to enteric diseases. These alterations underscore the critical need to identify key regulators of gut homeostasis, among which bile acids are increasingly recognized as pivotal for barrier integrity, microbial ecology, and host metabolism. Methods: Eight pairs of LBW (the initial BW was 0.850 ± 0.053 kg) and normal-birth-weight (NBW; 1.488 ± 0.083 kg) piglets were compared to evaluate cecal morphology and bile acid profiles. Subsequently, sixteen LBW piglets and eight NBW piglets were allocated into three groups: NBW (1.563 ± 0.052 kg), LBW control (LBW-CON; 0.950 ± 0.120 kg), and LBW with bile acid supplementation (LBW-bile powder; 0.925 ± 0.116 kg). Piglets in the LBW-bile powder group received 25 mg/kg BW of bile powder (hyodeoxycholic acid-enriched) by daily oral gavage for 14 days. Results: LBW piglets exhibited retarded cecal development and lower abundance of hyocholic acid species (p = 0.006). Importantly, bile powder supplementation significantly improved cecal length (p = 0.009) and mucosal thickness (p = 0.020) compared with LBW-CON piglets. Microbial analysis showed that the microbial dysbiosis index was restored to near-normal levels. Transcriptomic analysis revealed impaired extracellular matrix structure and mucus secretion in LBW piglets. Notably, bile powder supplementation markedly upregulated the protein expression of WNT8B (p < 0.001) and the bile acid receptors (i.e., GPBAR1 and FXR), alongside enhanced tight junctions and the goblet cell marker mucin-2 expression (p < 0.05). Conclusions: These findings suggest that specific bile acid supplementation improves gut barrier function and partially supports cecal development in LBW piglets. Full article
(This article belongs to the Special Issue Metabolomics and Nutrition: From Bench to Bedside)
Show Figures

Graphical abstract

18 pages, 5963 KB  
Article
In Vitro Investigation of the Effects of Octenidine Dihydrochloride on Nasal Septum Squamous Carcinoma Cells
by Ihsan Hakki Ciftci, Asuman Deveci Ozkan, Gulay Erman, Elmas Pinar Kahraman Kilbas and Mehmet Koroglu
Biomedicines 2025, 13(11), 2668; https://doi.org/10.3390/biomedicines13112668 - 30 Oct 2025
Viewed by 297
Abstract
Background/Objectives: The aim of this study was to investigate the cytotoxic, genotoxic, apoptotic, and anti-inflammatory effects of the antiseptic agent octenidine dihydrochloride (OCT-D) on the RPMI-2650 cell line derived from human nasal mucosa in vitro. Methods: RPMI-2650 cells and Human Umbilical [...] Read more.
Background/Objectives: The aim of this study was to investigate the cytotoxic, genotoxic, apoptotic, and anti-inflammatory effects of the antiseptic agent octenidine dihydrochloride (OCT-D) on the RPMI-2650 cell line derived from human nasal mucosa in vitro. Methods: RPMI-2650 cells and Human Umbilical Cord Endothelial Cells (HUVECs) were treated with various concentrations of OCT-D (0.00625–0.4%) for 12 and 24 h. Cell viability was assessed using the WST-1 assay, while DNA damage was assessed using the comet and micronucleus (MN) assays. Apoptotic activity was determined using Annexin V flow cytometry and fluorescence microscopy. Intracellular reactive oxygen species (ROS) levels were measured, and inflammatory cytokines (IL-1β, IL-6, TNF-α, and IFN-γ) were measured by Enzyme-Linked Immunosorbent Assay (ELISA). The mRNA expression of genes associated with apoptosis, oxidative stress, and inflammation was analyzed using RT-PCR. Results: OCT-D caused dose- and time-dependent cytotoxicity, and RPMI-2650 cells showed greater resistance compared to HUVECs. While a strong apoptotic response was observed in HUVECs, RPMI-2650 cells exhibited limited apoptosis. OCT-D was found to cause dose-dependent DNA damage and an increase in MN in both cell lines. OCT-D significantly reduced cytokine levels and ROS production in both cell types. RT-PCR results supported its anti-inflammatory and antioxidant effects at the molecular level. Conclusions: In conclusion, this study demonstrated that OCT-D exhibited minimal cytotoxic and apoptotic effects in RPMI-2650 cells, but affected vascular structure by inducing apoptosis in endothelial cells. These findings provide important evidence that OCT-D can be used as a potential adjunctive agent in nasal treatments, and these data need to be supported by preclinical and clinical studies. Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Figure 1

12 pages, 2579 KB  
Article
Effect of Poly (Lactic Acid/ε-Caprolactone) Bilayer Membrane on Tooth Extraction Socket Wound Healing in a Rat Model
by Bin Ji, Tingyu Xie, Ikiru Atsuta, Ikue Narimatsu, Yohei Jinno, Akira Takahashi, Mikio Imai, Kiyoshi Koyano and Yasunori Ayukawa
Materials 2025, 18(21), 4956; https://doi.org/10.3390/ma18214956 - 30 Oct 2025
Viewed by 351
Abstract
Guided bone regeneration membranes are essential for bone formation. While non-resorbable membranes require removal surgery, resorbable membranes such as poly (lactic-co-glycolic acid) PLGA are widely used; however, issues with animal-derived components and degradation control have been identified. A novel bilayer membrane composed of [...] Read more.
Guided bone regeneration membranes are essential for bone formation. While non-resorbable membranes require removal surgery, resorbable membranes such as poly (lactic-co-glycolic acid) PLGA are widely used; however, issues with animal-derived components and degradation control have been identified. A novel bilayer membrane composed of synthetic poly (L-lactic acid-co-ε-caprolactone) (PBM) was developed, offering prolonged degradability and elasticity. This study compared the wound-healing effects of PBM and PLGA membranes in vivo and in vitro experiments. In vivo, maxillary molars were extracted from rats, and membranes were placed over the sockets. Healing was evaluated histologically at 1, 2, 3, 4 and 8 weeks. In vitro, oral epithelial cells and fibroblasts were seeded on both sides of PBM. Adhesion and permeability of the membranes were assessed. In vivo, both groups displayed similar mucosal healing. However, PBM preserved a clear bone-soft tissue boundary. In vitro, the surface of PBM supported significantly greater oral epithelial cell adhesion than the reverse side, with no differences for fibroblasts. Both sides of PBM exhibited better protein permeability compared to PLGA. PBM maintained distinct bone-soft tissue separation in rat extraction sockets, suggesting its potential as an effective space maintainer in guided bone regeneration. Further studies are warranted to investigate the mechanisms underlying these favorable properties. Full article
Show Figures

Figure 1

16 pages, 356 KB  
Review
Mycotoxins and the Intestinal Epithelium: From Barrier Injury to Stem Cell Dysfunction
by Wenying Huo, Yingying Qiao, Xiangru He, Cailing Wang, Ruiqing Li, Long Che and Enkai Li
Toxins 2025, 17(11), 534; https://doi.org/10.3390/toxins17110534 - 30 Oct 2025
Viewed by 445
Abstract
Mycotoxins are toxic secondary metabolites produced by filamentous fungi that contaminate agricultural commodities, posing risks to food safety, animal productivity, and human health. The gastrointestinal tract is the first and most critical site of exposure, where the intestinal epithelium functions as both a [...] Read more.
Mycotoxins are toxic secondary metabolites produced by filamentous fungi that contaminate agricultural commodities, posing risks to food safety, animal productivity, and human health. The gastrointestinal tract is the first and most critical site of exposure, where the intestinal epithelium functions as both a physical and immunological barrier against luminal toxins and pathogens. While extensive research has demonstrated that mycotoxins disrupt epithelial integrity through tight junction impairment, oxidative stress, apoptosis, and inflammation, their effects on the intestinal stem cell (ISC) compartment and epithelial regeneration remain insufficiently understood. This review integrates recent findings from in vivo, cell culture, and advanced 3D intestinal organoid and gut-on-chip models to elucidate how mycotoxins such as deoxynivalenol and zearalenone impair ISC proliferation, alter Wnt/Notch signaling, and compromise mucosal repair. We also discuss dose relevance, species differences, and the modulatory roles of the microbiome and short-chain fatty acids, as well as emerging evidence of additive or synergistic toxicity under co-exposure conditions. By bridging well-established mechanisms of barrier disruption with the emerging concept of ISC-driven regenerative failure, this review identifies a critical knowledge gap in mycotoxin toxicology and highlights the need for integrative models that link epithelial damage to impaired regeneration. Collectively, these insights advance understanding of mycotoxin-induced intestinal dysfunction and provide a foundation for developing nutritional, microbial, and pharmacological strategies to preserve gut integrity and repair. Full article
29 pages, 1603 KB  
Review
The Role and Application of MAdCAM-1/α4β7-Induced Lymphocyte Migration in Inflammatory Enterohepatic Diseases
by Chuchu Yu, Yuqing Pan, Aojie Mao, Yu Zhao, Qiaohong Liu and Yiyang Hu
Biomedicines 2025, 13(11), 2659; https://doi.org/10.3390/biomedicines13112659 - 29 Oct 2025
Viewed by 361
Abstract
Inflammation drives the development of multiple inflammatory enterohepatic diseases. The recruitment of immune cells to inflammatory tissues is essential for maintaining immune homeostasis, mediating immune responses and regulating inflammation. MAdCAM-1/α4β7 is a pair of homing ligand and receptor that plays important roles in [...] Read more.
Inflammation drives the development of multiple inflammatory enterohepatic diseases. The recruitment of immune cells to inflammatory tissues is essential for maintaining immune homeostasis, mediating immune responses and regulating inflammation. MAdCAM-1/α4β7 is a pair of homing ligand and receptor that plays important roles in lymphocyte migration. Their binding induces lymphocytes to cross endothelial structures into corresponding lymphoid tissues, contributing to the inflammatory response. Aberrant lymphocyte migration due to excessive binding is closely related to the occurrence and development of inflammatory bowel disease and liver inflammation. In this review, we focus on the activation of α4β7 and binding to MAdCAM-1 how to induce the migration of multiple kinds of lymphocytes. Additionally, we describe the intestinal microbiota and its metabolites associated with MAdCAM-1/α4β7 in inflammatory enterohepatic diseases. We also discuss the current status of the development of monoclonal antibodies and small molecule drugs targeting MAdCAM-1/α4β7 for the remission and treatment of inflammatory enterohepatic disease. Future research should focus on enhancing hepatic specificity and conducting well-designed clinical trials for inflammatory liver diseases to confirm therapeutic efficacy. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Graphical abstract

Back to TopTop