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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline

Search Results (207)

Search Parameters:
Keywords = paracellular permeability

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 19724 KB  
Article
Endothelial Cell Transition: Preliminary Data on Cross-Organ Shift from Brain to Liver
by Alexey Larionov, Luis Filgueira and Christian M. Hammer
Cells 2025, 14(19), 1538; https://doi.org/10.3390/cells14191538 - 1 Oct 2025
Viewed by 628
Abstract
Background: Endothelial cells (EC), crucial components of the vascular system, are adaptable cells that maintain homeostasis and respond to pathological events through structural and functional plasticity. Hepatocyte growth factor (HGF) is a multifunctional cytokine that has been demonstrated to have protective and [...] Read more.
Background: Endothelial cells (EC), crucial components of the vascular system, are adaptable cells that maintain homeostasis and respond to pathological events through structural and functional plasticity. Hepatocyte growth factor (HGF) is a multifunctional cytokine that has been demonstrated to have protective and disruptive influence on the blood barrier function. In endothelial biology, its role is also poorly characterized. The present study explores the impact of supraphysiological concentrations of HGF on mouse brain endothelial cells (MBECs), scrutinizing how it alters their integrity and morphology. Methods: Two groups of MBECs—control (CTR) and experimental (EXP)—were analyzed at two time points: early passage (p5) and late passage (p41). The EXP-groups (p5 and p41) were treated with HGF at a concentration of 4 µL/mL. Cellular morphology was assessed with brightfield microscopy; protein expression and localization of the tight junction marker (ZO-1) and the endothelial marker (Factor VII related antigen/von Willebrand factor, vWf) were analyzed using Western blotting, immunocytochemistry, and confocal microscopy. Intercellular barrier function was estimated via Transendothelial Electric Resistance (TEER) and Transendothelial Dextran Permeability (TEDP) assays. Results: Microscopical analysis demonstrated a change in the morphology of the MBECs from a longitudinal, spindle-like shape to a rounded, more spheroid, cobblestone-like morphology under high-dose HGF treatment. Western blotting revealed a progressive decrease of ZO-1 expression in the EXP-groups. The expression of vWf did not show significant differences. Qualitative immunocytochemical staining: vWf showed consistent expression across all groups. ZO-1 displayed a punctate, well-defined membrane and cytoplasmic localization pattern in the CTR-groups at p5 and p41. In contrast, the p5 EXP-group demonstrated a shift to a more diffuse cytoplasmic pattern. At p41, the EXP-group displayed a markedly reduced ZO-1 signal with no clear-cut membrane localization. Confocal analysis: ZO-1: punctate membrane-associated localization in CTR-groups at p5 and 41. The EXP-groups at p5 and p41 confirmed the diffuse cytoplasmic ZO-1 distribution. Phalloidin: well-organized actin cytoskeleton in CTR-groups, but rearrangement and stress fiber disorganization in the EXP-groups, especially at p41. The merged images confirmed reduced co-localization of ZO-1 with actin structures. Barrier function: TEER values dropped significantly in HGF-treated cells. TEDP to small and medium molecular weight dextran increased markedly under HGF treatment. Conclusions: Our data demonstrate that supraphysiological doses of HGF in an in vitro MBEC-barrier-like model disrupt TJ organization, leading to morphological changes and functional weakening of the MBEC-barrier-like structure, as shown by uncoupling between ZO-1/F-actin cytoskeleton, reduced TEER, and increased size-selective paracellular permeability (TEDP). Full article
Show Figures

Figure 1

23 pages, 18324 KB  
Article
Tissue Regression-Related Alterations in the Expression of Adherens and Tight Junction Proteins in the Hen Oviduct
by Karolina Frydrych and Anna Hrabia
Int. J. Mol. Sci. 2025, 26(19), 9451; https://doi.org/10.3390/ijms26199451 - 27 Sep 2025
Viewed by 388
Abstract
Intercellular junctions are involved in the regulation of epithelial function and remodeling in the female reproductive system; however, their importance in the avian oviduct is poorly known. The aim of this study was: first, to provide information on the expression and localization of [...] Read more.
Intercellular junctions are involved in the regulation of epithelial function and remodeling in the female reproductive system; however, their importance in the avian oviduct is poorly known. The aim of this study was: first, to provide information on the expression and localization of key tight (occludin, claudin 1, 4, 5, junctional adhesion molecule [JAM] 2, 3) and adherens (E-cadherin, β-catenin) junction proteins in the hen oviduct, and second, to compare expression and localization of these molecules between laying and subjected to fasting-induced pause in laying hens. Tissue samples from all oviductal segments, i.e., infundibulum, magnum, isthmus, shell gland, and vagina were collected on the sixth day of the experiment from the control hens and hens that had been fasted for five consecutive days. Specific oviductal part-dependent expression patterns of examined genes (by quantitative real-time polymerase chain reaction [qRT-PCR]) and/or proteins (by Western blotting) were found, with the highest mRNA transcript and protein abundances in the infundibulum, shell gland, and vagina, and the lowest in the magnum. Fasting-induced partial regression of the oviduct was accompanied by alterations in mRNA transcript and protein abundances of examined molecules. Reduced staining intensity of immunoreaction (analyzed by immunofluorescence) for occludin, E-cadherin, and β-catenin proteins was observed in the oviduct of non-laying hens. Our results indicate the potential involvement of these proteins in controlling intercellular communication, cell signaling, paracellular permeability, and mucosal barrier functionality, which impact the functioning of the hen oviduct. Furthermore, our observations provide novel insights into the molecular composition of tight and adherens junctions and its contribution to the remodeling of the oviduct during its regression induced by fasting. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

13 pages, 12457 KB  
Article
Goblet Cell-Mediated Pathway: A Major Contributor to Increased Intestinal Permeability in Streptozotocin-Induced Type 1 Diabetic Mice
by Ming-Hsun Wu, Lee-Wei Chen, Jiann-Hwa Chen and Chieh-Wen Lai
Int. J. Mol. Sci. 2025, 26(18), 8890; https://doi.org/10.3390/ijms26188890 - 12 Sep 2025
Viewed by 502
Abstract
Gut barrier dysfunction and increased intestinal permeability are closely linked to the pathogenesis of type 1 diabetes and its complications. Streptozotocin (STZ)-induced diabetic mice, which mimic β-cell destruction and insulin deficiency, provide a widely used model for studying type 1 diabetes-associated intestinal barrier [...] Read more.
Gut barrier dysfunction and increased intestinal permeability are closely linked to the pathogenesis of type 1 diabetes and its complications. Streptozotocin (STZ)-induced diabetic mice, which mimic β-cell destruction and insulin deficiency, provide a widely used model for studying type 1 diabetes-associated intestinal barrier impairment. However, the cellular pathways mediating this dysfunction, particularly the role of goblet cells, remain incompletely elucidated. This study aimed to investigate the association between the gut barrier function and diabetes. Using real-time intravital multiphoton microscopy, we investigated intestinal barrier integrity in STZ-induced type 1 diabetic mice. Three groups were analysed: the control, STZ-diabetic, and STZ-diabetic mice treated with fructooligosaccharide (FOS) for 1 week. Intestinal permeability was assessed by measuring fluorescein isothiocyanate (FITC)-dextran concentrations in the portal vein and visualising translocation into villi. Epithelial morphology was examined, focusing on goblet cell density and leakage pathways. STZ-diabetic mice demonstrated a significant increase in intestinal permeability, evidenced by elevated FITC-dextran levels in the portal vein and villi. Multiphoton imaging revealed a notable rise in the goblet cell-to-enterocyte ratio in diabetic mice, while the gap density remained unchanged. The predominant route of macromolecular leakage in STZ-diabetic mice was via goblet cells rather than by paracellular gaps. One-week FOS supplementation significantly reduced goblet cell density and partially restored barrier function without altering the distribution of leakage pathways. These findings highlight goblet cell-mediated transcellular leakage as a major mechanism of gut barrier dysfunction in type 1 diabetic mice. Short-term FOS treatment partially reverses these alterations. Targeting goblet cell function may offer a promising therapeutic strategy to restore gut barrier integrity in diabetes. Full article
(This article belongs to the Special Issue Targeted Therapy for Immune Diseases)
Show Figures

Figure 1

18 pages, 4379 KB  
Article
Botulinum Toxin Type A Inhibits Submandibular Secretion via the ERK/miR-124-3p/Specificity Protein 1/Claudin-1 Axis
by Qian-Ying Mao, Yan Huang, Zhuo Chen, Xiao-Feng Shan, Shang Xie, Li-Ling Wu, Ruo-Lan Xiang and Zhi-Gang Cai
Cells 2025, 14(17), 1366; https://doi.org/10.3390/cells14171366 - 2 Sep 2025
Viewed by 676
Abstract
Botulinum toxin type A (BTXA) is widely used for the treatment of sialorrhea; however, its mechanism remains unclear. Tight junctions (TJs) are limiting factors for salivary secretion through the paracellular pathway in the salivary gland, among which claudin-1 (Cldn1) is a TJ protein [...] Read more.
Botulinum toxin type A (BTXA) is widely used for the treatment of sialorrhea; however, its mechanism remains unclear. Tight junctions (TJs) are limiting factors for salivary secretion through the paracellular pathway in the salivary gland, among which claudin-1 (Cldn1) is a TJ protein that mainly plays a barrier role. This study observed that Cldn1 was upregulated in BTXA-treated rats’ submandibular glands and SMG-C6 cells. Knockdown of Cldn1 reversed the BTXA-induced reduction in paracellular permeability. The transcription factor specificity protein-1 (Sp1), which binds to the Cldn1 promoter, was also upregulated by BTXA, and its expression was linked to the ERK1/2 pathway. Inhibition of ERK1/2 by U0126 reversed the BTXA-induced upregulation of Sp1 and Cldn1, as well as the reduction in paracellular permeability. MiR-124-3p, which directly targets Sp1, was downregulated by BTXA, but its overexpression counteracted Sp1 and Cldn1 upregulation. Although miR-124-3p did not affect ERK1/2 phosphorylation, ERK1/2 inhibition reversed the BTXA-induced decrease in miR-124-3p expression. These findings reveal a regulatory pathway through which BTXA reduces paracellular permeability in SMG-C6 cells via the ERK1/2/miR-124-3p/Sp1/Cldn1 axis. Full article
(This article belongs to the Section Cell Signaling)
Show Figures

Graphical abstract

34 pages, 2426 KB  
Review
Claudin 5 Across the Vascular Landscape: From Blood–Tissue Barrier Regulation to Disease Mechanisms
by Mohamed S. Selim, Bayan R. Matani, Harry O. Henry-Ojo, S. Priya Narayanan and Payaningal R. Somanath
Cells 2025, 14(17), 1346; https://doi.org/10.3390/cells14171346 - 29 Aug 2025
Viewed by 2903
Abstract
Claudin 5 (Cldn5) is a critical tight junction protein essential for maintaining paracellular barrier integrity across endothelial and epithelial cells in barrier-forming tissues, including the blood–brain barrier and blood–retinal barrier. Cldn5 plays a central role in regulating vascular permeability, immune responses, and tissue [...] Read more.
Claudin 5 (Cldn5) is a critical tight junction protein essential for maintaining paracellular barrier integrity across endothelial and epithelial cells in barrier-forming tissues, including the blood–brain barrier and blood–retinal barrier. Cldn5 plays a central role in regulating vascular permeability, immune responses, and tissue homeostasis. The complex distribution and organ-specific regulation of Cldn5 underscore its potential as a promising therapeutic target. This review comprehensively analyzes the role of Cldn5 in endothelial and epithelial barrier function, its regulation of vascular permeability, and the discrepancies in the literature regarding its expression, regulation, and function in both physiological and pathological conditions across multiple organ systems, including the retina, brain, lung, heart, gut, kidney, liver, skin, and peripheral nerves, while emphasizing its tissue-specific expression patterns. We discuss how both reduced and excessive expressions of Cldn5 can disrupt barrier integrity and contribute to the pathogenesis of ischemic retinopathies, neuroinflammation, cardiovascular injury, and other forms of barrier dysfunction. Furthermore, we explore the dual role of Cldn5 as both a biomarker and a therapeutic target, highlighting emerging strategies such as RNA silencing, pharmacological stabilizers, and transcriptional modulators in controlling barrier leakage in disease conditions. Full article
Show Figures

Figure 1

19 pages, 2653 KB  
Article
Fine Particulate Matter (PM2.5) Disrupts Intestinal Barrier Function by Inducing Oxidative Stress and PI3K/AKT-Mediated Inflammation in Caco-2 Cells
by Ruiwei Liao, Qianwen Zhang, Yao Lu, Feifei Huang, Wenjuan Cao, Ming Li, Lin Zhou and Yan Li
Int. J. Mol. Sci. 2025, 26(17), 8271; https://doi.org/10.3390/ijms26178271 - 26 Aug 2025
Viewed by 995
Abstract
Fine particulate matter (PM2.5) is an environmental factor that triggers gastrointestinal diseases. However, the effects of PM2.5 on intestinal function are not fully understood. This study established an environmental exposure cell model to explore PM2.5-induced intestinal permeability alteration and its mechanisms. Intestinal barrier [...] Read more.
Fine particulate matter (PM2.5) is an environmental factor that triggers gastrointestinal diseases. However, the effects of PM2.5 on intestinal function are not fully understood. This study established an environmental exposure cell model to explore PM2.5-induced intestinal permeability alteration and its mechanisms. Intestinal barrier permeability was evaluated via trans-epithelial electrical resistance (TEER) measurement and FITC–dextran paracellular penetration analysis, followed by detection of intercellular junction protein β-catenin and its coding gene CTNNB1. Expression of inflammatory cytokines (TNF-α, IL-6) and phosphorylation of PI3K and AKT were assessed using quantitative real-time polymerase chain reaction and Western blot, respectively. Reactive oxygen species (ROS) and malondialdehyde were measured using commercial kits to observe cellular oxidative stress. The results showed that PM2.5 impaired the intestinal barrier, as indicated by reduced TEER, increased FITC–dextran penetration, down-regulated expression of β-catenin and CTNNB1. Additionally, compared with the control, inflammatory cytokines and oxidative stress markers were significantly elevated after PM2.5 exposure. The ratio of p-PI3K/PI3K and p-AKT/AKT was also up-regulated in PM2.5-exposed Caco-2 cells. Pretreatment with PI3K inhibitor LY294002 and ROS scavenger NAC modulated β-catenin expression, reduced inflammation/ROS, and alleviated the hyperpermeability of Caco-2 cells. Thus, our results reveal that PM2.5 induces PI3K/AKT-mediated inflammation and ROS generation in Caco-2 cells, leading to intestinal barrier impairment. Full article
(This article belongs to the Section Molecular Toxicology)
Show Figures

Figure 1

17 pages, 2063 KB  
Article
Comprehensive UPLC-MS/MS Method for Quantifying Four Key Intestinal Permeability Markers in Caco-2 Models
by Luciana Silva de Araújo, Eduardo José Crevelin, Luiz Alberto Beraldo de Moraes and Niege Araçari Jacometti Cardoso Furtado
Molecules 2025, 30(17), 3477; https://doi.org/10.3390/molecules30173477 - 24 Aug 2025
Viewed by 1080
Abstract
A comprehensive UPLC-MS/MS method was developed and validated for the simultaneous separation and quantification of atenolol, propranolol, quinidine, and verapamil, using established intestinal permeability standards in the Caco-2 cell monolayer model. This in vitro model is widely accepted for predicting intestinal drug permeability [...] Read more.
A comprehensive UPLC-MS/MS method was developed and validated for the simultaneous separation and quantification of atenolol, propranolol, quinidine, and verapamil, using established intestinal permeability standards in the Caco-2 cell monolayer model. This in vitro model is widely accepted for predicting intestinal drug permeability and is formally recognized by global regulatory agencies, including the FDA, EMA, and WHO, as a surrogate for assessing drug permeability in biowaiver applications under the Biopharmaceutics Classification System (BCS) framework. Despite its regulatory importance, standardized methods for the simultaneous quantification of key permeability markers remain scarce. The selected compounds represent distinct transport pathways: paracellular (atenolol), passive transcellular (propranolol, verapamil), and P-glycoprotein-mediated efflux (quinidine). Method validation followed FDA guidelines and demonstrated high selectivity, linearity (r2 > 0.998), precision, and accuracy. Solid-phase extraction enhanced recovery and reduced matrix effects. Application to Caco-2 permeability assays confirmed expected transport profiles, including P-gp inhibition effects with verapamil. By integrating multiple analytes in a single workflow, the method improves analytical throughput, supports mechanistic interpretation, and ensures consistency across assays. This advanced separation strategy, combined with sensitive mass spectrometric detection, supports regulatory and BCS-based classification studies, contributing to the standardization of permeability assessments in drug development. Full article
Show Figures

Graphical abstract

15 pages, 1034 KB  
Article
In Vitro Oral Cavity Permeability Assessment to Enable Simulation of Drug Absorption
by Pankaj Dwivedi, Priyata Kalra, Haiying Zhou, Khondoker Alam, Eleftheria Tsakalozou, Manar Al-Ghabeish, Megan Kelchen and Giovanni M. Pauletti
Pharmaceutics 2025, 17(7), 924; https://doi.org/10.3390/pharmaceutics17070924 - 17 Jul 2025
Viewed by 1307
Abstract
Background/Objectives: The oral cavity represents a convenient route of administration for drugs that exhibit significant hepatic first-pass extraction. In this study, the mucosal permeation properties of selected active pharmaceutical ingredients (APIs) incorporated into oral cavity drug products that are approved by the U.S. [...] Read more.
Background/Objectives: The oral cavity represents a convenient route of administration for drugs that exhibit significant hepatic first-pass extraction. In this study, the mucosal permeation properties of selected active pharmaceutical ingredients (APIs) incorporated into oral cavity drug products that are approved by the U.S. Food and Drug Administration were quantified using the human-derived sublingual HO-1-u-1 and buccal EpiOral™ in vitro tissue models. Methods: Epithelial barrier properties were monitored using propranolol and Lucifer Yellow as prototypic transcellular and paracellular markers. APIs were dissolved in artificial saliva, pH 6.7, and transepithelial flux from the apical to the basolateral compartment was quantified using HPLC. Results: Apparent permeability coefficients (Papp) calculated for these APIs in the sublingual HO-1-u-1 tissue model varied from Papp = 2.72 ± 0.06 × 10−5 cm/s for asenapine to Papp = 6.21 ± 2.60 × 10−5 cm/s for naloxone. In contrast, the buccal EpiOral™ tissue model demonstrated greater discrimination power in terms of permeation properties for the same APIs, with values ranging from Papp = 3.31 ± 0.83 × 10−7 cm/s for acyclovir to Papp = 2.56 ± 0.68 × 10−5 cm/s for sufentanil. The tissue-associated dose fraction recovered at the end of the transport experiment was significantly increased in the buccal EpiOral™ tissue model, reaching up to 8.5% for sufentanil. Conclusions: Experimental permeation data collected for selected APIs in FDA-approved oral cavity products will serve as a training set to aid the development of predictive computational models for improving algorithms that describe drug absorption from the oral cavity. Following a robust in vitro–in vivo correlation analysis, it is expected that such innovative in silico modeling strategies will the accelerate development of generic oral cavity products by facilitating the utility of model-integrated evidence to support decision making in generic drug development and regulatory approval. Full article
Show Figures

Graphical abstract

19 pages, 3360 KB  
Article
PTEN Inactivation in Mouse Colonic Epithelial Cells Curtails DSS-Induced Colitis and Accelerates Recovery
by Larissa Kotelevets, Francine Walker, Godefroy Mamadou, Bruno Eto, Thérèse Lehy and Eric Chastre
Cancers 2025, 17(14), 2346; https://doi.org/10.3390/cancers17142346 - 15 Jul 2025
Viewed by 814
Abstract
Background: PTEN is a tumor suppressor that controls many pathophysiological pathways, including cell proliferation, differentiation, apoptosis and invasiveness. Although PTEN down-modulation is a critical event in neoplastic progression, it becomes apparent that transient and local inhibition of PTEN activity might be beneficial [...] Read more.
Background: PTEN is a tumor suppressor that controls many pathophysiological pathways, including cell proliferation, differentiation, apoptosis and invasiveness. Although PTEN down-modulation is a critical event in neoplastic progression, it becomes apparent that transient and local inhibition of PTEN activity might be beneficial for the healing process. Methods: In the present study, we investigated the impact of PTEN invalidation in mouse intestinal epithelium under a physiological condition and after dextran sulfate sodium (DSS) treatment to induce experimental colitis. PTEN conditional knockout was induced in intestinal epithelial cells after crossing villin-Cre and PTENflox/flox mice. Results: PTEN invalidation alleviates experimental colitis induced by DSS, as evidenced by decreased weight loss during the acute phase, the lower expression of inflammation markers, including the proinflammatory cytokines IFN-γ, CXCL1 and CXCL2, reduced mucosal lesions, and faster recovery after resolution of inflammation. This protective effect might result in part from the sustained proliferation of colonic epithelium, leading to hyperplasia and increased colonic crypt depth under physiological conditions, which was further exacerbated in the vicinity of mucosal injury induced by DSS treatment. Furthermore, PTEN knockout decreased paracellular permeability, thereby enhancing the intestinal barrier function. This process was associated with the reinforcement of claudin-3 immunostaining, especially on the surface epithelium of villin-Cre PTENflox/flox mice. Conclusions: PTEN inactivation exerts a protective effect on the onset of colitis, and the transient and local down-modulation of PTEN might constitute an approach to drive recovery following acute intestinal inflammation. Full article
(This article belongs to the Special Issue PTEN: Regulation, Signalling and Targeting in Cancer)
Show Figures

Graphical abstract

19 pages, 3876 KB  
Article
Improving Ex Vivo Nasal Mucosa Experimental Design for Drug Permeability Assessments: Correcting Mucosal Thickness Interference and Reevaluating Fluorescein Sodium as an Integrity Marker for Chemically Induced Mucosal Injury
by Shengnan Zhao, Jieyu Zuo, Marlon C. Mallillin, Ruikun Tang, Michael R. Doschak, Neal M. Davies and Raimar Löbenberg
Pharmaceuticals 2025, 18(6), 889; https://doi.org/10.3390/ph18060889 - 13 Jun 2025
Cited by 1 | Viewed by 2298
Abstract
Objectives: Ex vivo nasal mucosa models provide physiologically relevant platforms for evaluating nasal drug permeability; however, their application is often limited by high experimental variability and the absence of standardized methodologies. This study aimed to improve experimental design by addressing two major [...] Read more.
Objectives: Ex vivo nasal mucosa models provide physiologically relevant platforms for evaluating nasal drug permeability; however, their application is often limited by high experimental variability and the absence of standardized methodologies. This study aimed to improve experimental design by addressing two major limitations: the confounding effects of mucosal thickness and the questionable reliability of fluorescein sodium (Flu-Na) as an integrity marker for chemically induced mucosal injury. Methods: Permeability experiments were conducted using porcine nasal tissues mounted in Franz diffusion cells, with melatonin and Flu-Na as model compounds. Tissues of varying thickness were collected from both intra- and inter-individual sources, and a numerical simulation-based method was employed to normalize apparent permeability coefficients (Papp) to a standardized mucosal thickness of 0.80 mm. The effects of thickness normalization and chemically induced damage were systematically evaluated. Results: Thickness normalization substantially reduced variability in melatonin Papp, particularly within same-animal comparisons, thereby improving statistical power and data reliability. In contrast, Flu-Na exhibited inconsistent correlations across different pigs and failed to reflect the expected increase in permeability following isopropyl alcohol (IPA)-induced epithelial damage. These results suggest that the relationship between epithelial injury and paracellular transport may be non-linear and not universally applicable under ex vivo conditions, limiting the suitability of Flu-Na as a standalone marker of mucosal integrity. Conclusions: The findings highlight the importance of integrating mucosal thickness correction into standardized experimental protocols and call for a critical reassessment of Flu-Na in nasal drug delivery research. Full article
(This article belongs to the Section Pharmaceutical Technology)
Show Figures

Graphical abstract

17 pages, 6693 KB  
Article
Covalent Grafting of Inorganic Selenium to the Water-Soluble and Nondigestive Chinese Yam Polysaccharides Causes Greater Protection of IEC-6 Cells with Acrylamide Injury
by Zhen-Xing Wang, Li-Li Zhang and Xin-Huai Zhao
Foods 2025, 14(9), 1560; https://doi.org/10.3390/foods14091560 - 29 Apr 2025
Viewed by 598
Abstract
Acrylamide, a harmful substance generated during the normal thermal treatment of foods, has been shown to adversely affect human health, particularly the vital intestinal barrier function. Meanwhile, natural polysaccharides are recognized to exert an important biofunction in the intestine by protecting barrier integrity. [...] Read more.
Acrylamide, a harmful substance generated during the normal thermal treatment of foods, has been shown to adversely affect human health, particularly the vital intestinal barrier function. Meanwhile, natural polysaccharides are recognized to exert an important biofunction in the intestine by protecting barrier integrity. In this study, the non-starch, water-soluble, and nondigestive yam polysaccharide (YP) was extracted from fresh Chinese yam, while two selenylated derivatives with different extents of selenylation were prepared via the HNO3-Na2SeO3 reaction system, and designated as YPSe-I and YPSe-II, respectively. Their protective activities and the associated molecular mechanisms of these substances against acrylamide-induced damage in rat intestinal epithelial (IEC-6) cells were thereby investigated. The experimental results demonstrated that the selenium contents of YPSe-I and YPSe-II were 0.80 and 1.48 g/kg, respectively, whereas that of the original YP was merely 0.04 g/kg. In IEC-6 cells, in comparison with YP, both YPSe-I and YPSe-II showed higher efficacy than YP in alleviating acrylamide-induced cell toxicity through promoting cell viability, suppressing the release of lactate dehydrogenase, and decreasing the generation of intracellular reactive oxygen species. Both YPSe-I and YPSe-II could also manifest higher effectiveness than YP in maintaining cell barrier integrity against the acrylamide-induced barrier disruption. The mentioned barrier protection was achieved by increasing transepithelial electrical resistance, reducing paracellular permeability, facilitating the distribution and expression of F-actin between the cells, and up-regulating the production of three tight junctions, namely ZO-1, occludin, and claudin-1. Additionally, acrylamide was observed to trigger the activation of the MAPK signaling pathway, thereby leading to cell barrier dysfunction. In contrast, YPSe-I and particularly YPSe-II were capable of down-regulating two MAPK-related proteins, namely p-p38 and p-JNK, and thereby inhibiting the acrylamide-induced activation of the MAPK signaling pathway. Moreover, YPSe-II in the cells was consistently shown to provide greater barrier protection than YPSe-I. In conclusion, chemical selenylation of YP could cause higher activity in mitigating acrylamide-induced cytotoxicity and intestinal barrier dysfunction, while the efficacy of activity enhancement was positively affected by the selenylation extent. Full article
(This article belongs to the Section Food Toxicology)
Show Figures

Graphical abstract

26 pages, 5853 KB  
Article
Kinin B1 Receptor Agonist Enhances Blood-Brain Barrier Permeability in Healthy and Glioblastoma Environments
by Carolina Batista, João Victor Roza Cruz, Michele Siqueira, João Bosco Pesquero, Joice Stipursky and Fabio de Almeida Mendes
Pharmaceuticals 2025, 18(4), 591; https://doi.org/10.3390/ph18040591 - 18 Apr 2025
Cited by 1 | Viewed by 1199
Abstract
Background/Objectives: The low permeability of the blood-brain barrier (BBB) represents a significant challenge to effective systemic chemotherapy for primary and metastatic brain cancers. Kinin receptors play a crucial role in modulating BBB permeability, and their agonist analogs have been explored in preclinical [...] Read more.
Background/Objectives: The low permeability of the blood-brain barrier (BBB) represents a significant challenge to effective systemic chemotherapy for primary and metastatic brain cancers. Kinin receptors play a crucial role in modulating BBB permeability, and their agonist analogs have been explored in preclinical animal models to enhance drug delivery to the brain. In this study, we investigated whether des-Arg9-bradykinin (DBK), a physiological agonist of kinin B1 receptor (B1R), acts as a brain drug delivery adjuvant by promoting the transient opening of the BBB. Methods: Human brain microvascular endothelial cells (HBMECs) were treated with DBK in the culture medium and in conditioned media from glioblastoma cell lines, namely T98G (CMT98G) and U87MG (CMU87). Immunofluorescence, RT-qPCR, in-cell Western assay, and proximity ligation assay (PLA) were performed to analyze BBB components, kinin receptors and TLR4, a receptor associated with the kinin pathway and inflammation. The effect of DBK on enhancing paracellular molecule transport was evaluated using Evans blue dye (EB) quantification in a cell culture insert assay and in an in vivo model, where mice with and without brain tumors were treated with DBK. To assess the functional impact of the transient BBB opening induced by DBK, the chemotherapeutic drug doxorubicin (DOX) was administered. Results: Treatment with DBK facilitates the presence of EB in the brain parenchyma by transiently disrupting the BBB, as further evidenced by the increased paracellular passage of the dye in an in vitro assay. B1R activation by DBK induces transient BBB opening lasting less than 48 h, enhancing the bioavailability of the DOX within the brain parenchyma and glioma tumor mass. The interaction between B1R and TLR4 is disrupted by the secreted factors released by glioblastoma cells, as conditioned media from T98G and U87 reduce TLR4 staining in endothelial cells without affecting B1R expression. Conclusions: These results further support the potential of B1R activation as a strategy to enhance targeted drug delivery to the brain. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Graphical abstract

13 pages, 1993 KB  
Article
A Probiotic Mixture of Lactobacillus rhamnosus LR 32, Bifidobacterium lactis BL 04, and Bifidobacterium longum BB 536 Counteracts the Increase in Permeability Induced by the Mucosal Mediators of Irritable Bowel Syndrome by Acting on Zonula Occludens 1
by Maria Raffaella Barbaro, Francesca Bianco, Cesare Cremon, Giovanni Marasco, Vincenzo Stanghellini and Giovanni Barbara
Int. J. Mol. Sci. 2025, 26(6), 2656; https://doi.org/10.3390/ijms26062656 - 15 Mar 2025
Cited by 2 | Viewed by 3642
Abstract
Irritable Bowel Syndrome (IBS) is a disorder of gut- brain interaction characterized by recurrent abdominal pain associated with altered bowel habits. The therapeutic options for IBS patients include the use of probiotics. The aim of this study was to assess the effect of [...] Read more.
Irritable Bowel Syndrome (IBS) is a disorder of gut- brain interaction characterized by recurrent abdominal pain associated with altered bowel habits. The therapeutic options for IBS patients include the use of probiotics. The aim of this study was to assess the effect of a multi-strain probiotic made up by Lactobacillus rhamnosus LR 32, Bifidobacterium lactis BL 04, and Bifidobacterium longum BB 536 (Serobioma, Bromatech s.r.l., Milano, Italy) on an in vitro model of the intestinal epithelial barrier in the presence of mucosal mediators that are released by IBS patients. IBS (n = 28; IBS with predominant diarrhea, IBS-D = 10; IBS with predominant constipation, IBS-C = 9; and IBS with mixed bowel habits, IBS-M = 9) patients, diagnosed according to the Rome IV criteria, and asymptomatic controls (ACs, n = 7) were enrolled. Mucosal mediators that were spontaneously released by colonic biopsies were collected (supernatants). Two doses of Serobioma were tested with/without IBS/AC mediators. RNA was extracted from Caco-2 cells to evaluate the tight junction (TJ) expression. Serobioma (106 CFU/mL) significantly reinforced the Caco-2 monolayer compared to growth medium alone (p < 0.05). IBS supernatants significantly increased Caco-2 paracellular permeability compared to the AC supernatants. The co-incubation of Caco-2 cells with IBS supernatants and Serobioma (106 CFU/mL) avoided the paracellular permeability alterations that were induced by IBS supernatants alone (p < 0.001), and, in particular, IBS-D and IBS-M ones. The co-incubation of Serobioma (106 CFU/mL) and IBS-D supernatants significantly increased ZO-1 expression compared to Caco-2 cells incubated with supernatants alone (p < 0.05), as confirmed via qPCR analyses. Serobioma (106 CFU/mL) counteracts the paracellular permeability changes that are induced by IBS supernatants, in particular IBS-D and IBS-M supernatants, likely modulating ZO-1 expression. Full article
Show Figures

Figure 1

16 pages, 3219 KB  
Article
Caco2/HT-29 In Vitro Cell Co-Culture: Barrier Integrity, Permeability, and Tight Junctions’ Composition During Progressive Passages of Parental Cells
by Elena Donetti, Paola Bendinelli, Margherita Correnti, Elena Gammella, Stefania Recalcati and Anita Ferraretto
Biology 2025, 14(3), 267; https://doi.org/10.3390/biology14030267 - 6 Mar 2025
Cited by 1 | Viewed by 3385
Abstract
Epithelial linings are crucial for the maintenance of physiological barriers. The intestinal epithelial barrier (IEB) consists of enterocytes through tight junctions and mucus-secreting cells and can undergo physiological modifications throughout life. To reproduce as closely as possible the IEB main features over time, [...] Read more.
Epithelial linings are crucial for the maintenance of physiological barriers. The intestinal epithelial barrier (IEB) consists of enterocytes through tight junctions and mucus-secreting cells and can undergo physiological modifications throughout life. To reproduce as closely as possible the IEB main features over time, in vitro co-cultures of Caco2/HT-29 70/30 formed by parental Caco2 and HT-29 cells sub-cultivated for more than 40 passages were set up. The measurements of the transepithelial electrical resistance (TEER) identified two populations: physiological TEER co-cultures (PC) with values > 50 Ωcm2 formed by parental cells with fewer than 40 passages, and leaky TEER co-cultures (LC) with values < 50 Ωcm2 formed by parental cells with more than 40 passages. In LC, paracellular permeability increased in parallel. By immunofluorescence and Western blot analysis, an increase in claudin 2 was observed in LC vs. PC, with no differences in occludin expression. MUC-2 immunoreactivity was stronger in PC than in LC. LC also showed an enhanced vulnerability to TNFα+IFN-γ. These results reproduce the main morpho-functional modifications reported in the human leaky/aged gut and support the usefulness of our in vitro cell model for studying the molecular processes underlying these modifications and testing drug/nutraceutical treatments to ameliorate leaky gut aging. Full article
Show Figures

Figure 1

31 pages, 2670 KB  
Review
Molecular Motors in Blood–Brain Barrier Maintenance by Astrocytes
by Ana Filipa Sobral, Inês Costa, Vanessa Teixeira, Renata Silva and Daniel José Barbosa
Brain Sci. 2025, 15(3), 279; https://doi.org/10.3390/brainsci15030279 - 6 Mar 2025
Cited by 3 | Viewed by 3340
Abstract
The blood–brain barrier (BBB) comprises distinct cell types, including endothelial cells, pericytes, and astrocytes, and is essential for central nervous system (CNS) homeostasis by selectively regulating molecular transport and maintaining integrity. In particular, astrocytes are essential for BBB function, as they maintain BBB [...] Read more.
The blood–brain barrier (BBB) comprises distinct cell types, including endothelial cells, pericytes, and astrocytes, and is essential for central nervous system (CNS) homeostasis by selectively regulating molecular transport and maintaining integrity. In particular, astrocytes are essential for BBB function, as they maintain BBB integrity through their end-feet, which form a physical and biochemical interface that enhances endothelial cell function and barrier selectivity. Moreover, they secrete growth factors like vascular endothelial growth factor (VEGF) and transforming growth factor-beta (TGF-β), which regulate tight junction (TJ) proteins (e.g., claudins and occludins) crucial for limiting paracellular permeability. Molecular motors like kinesins, dynein, and myosins are essential for these astrocyte functions. By facilitating vesicular trafficking and protein transport, they are essential for various functions, including trafficking of junctional proteins to support BBB integrity, the proper mitochondria localization within astrocyte processes for efficient energy supply, the polarized distribution of aquaporin (AQP)-4 at astrocyte end-feet for regulating water homeostasis across the BBB, and the modulation of neuroinflammatory responses. Moreover, myosin motors modulate actomyosin dynamics to regulate astrocyte process outgrowth, adhesion, migration, and morphology, facilitating their functional roles. Thus, motor protein dysregulation in astrocytes can compromise BBB function and integrity, increasing the risk of neurodegeneration. This review explores the complex interplay between astrocytes and molecular motors in regulating BBB homeostasis, which represents an attractive but poorly explored area of research. Full article
Show Figures

Figure 1

Back to TopTop