Fact-checked by Grok 2 weeks ago

Intestinal permeability

Intestinal permeability is the functional property of the that regulates the selective passage of ions, water, nutrients, and other molecules from the gut lumen into the bloodstream, while acting as a barrier to prevent harmful substances such as pathogens, toxins, and undigested particles from entering systemic circulation. This process occurs across a vast surface area of approximately 32 square meters, primarily in the , requiring a substantial portion of the body's energy expenditure to maintain barrier integrity and . Under normal conditions, it facilitates essential physiological functions like nutrient absorption (e.g., glucose and electrolytes via transcellular routes) and immune surveillance through the , supported by a layer and secretory immunoglobulins such as IgA. The intestinal barrier's permeability is primarily controlled by tight junctions (TJs), protein complexes including claudins, , and zonula occludens-1 (ZO-1) that seal the paracellular space between epithelial cells, alongside transcellular mechanisms involving transporters and for larger or specific molecules. Regulation involves multiple factors, including the , which modulates TJ expression and barrier function through and like Escherichia coli Nissle 1917; dietary components such as high-fat diets or that can disrupt TJs; and humoral modulators like , a protein that reversibly opens TJs to increase permeability in response to physiological stimuli. Stress, inflammation, and environmental factors (e.g., or nanoparticles) further influence this dynamic equilibrium, with vitamins A and D enhancing barrier integrity by promoting TJ assembly. Pathological increases in intestinal permeability, often termed "leaky gut," occur when integrity is compromised, allowing translocation of luminal contents that trigger local and systemic inflammation via immune activation and bacterial products like lipopolysaccharides. This dysfunction is implicated in a range of gastrointestinal disorders, including (e.g., and , where elevated permeability precedes clinical relapse), disease (driven by gluten-induced release), and . Beyond the gut, it contributes to systemic conditions such as , , , non-alcoholic , and even neurological disorders like , through the gut-liver axis or microbiota-brain interactions that amplify chronic low-grade inflammation; as of 2025, elevated permeability biomarkers are also prospectively linked to incident (e.g., and ) and . Emerging therapies target permeability restoration, including antagonists, , and dietary interventions like supplementation, with recent 2024 studies highlighting exercise's role in improving microbiota composition and .

Overview and Physiology

Definition and Importance

Intestinal permeability refers to the capacity of the to selectively permit the passage of essential substances, such as nutrients, ions, and , while forming a robust barrier against the translocation of harmful luminal contents, including pathogens and toxins. This regulated process occurs primarily through transcellular and paracellular routes, ensuring efficient absorption without compromising barrier integrity. The importance of intestinal permeability lies in its central role in physiological , where it balances uptake with protection against microbial invasion, thereby preventing systemic immune activation from inappropriate exposure to gut contents. Disruptions in this barrier function, leading to increased permeability, have been implicated in the initiation and progression of chronic inflammatory conditions by allowing the leakage of bacterial products like lipopolysaccharides into the bloodstream. Historically, investigations into intestinal absorption began in the through physiological studies on across the gut wall, laying foundational insights into barrier dynamics. Modern understanding advanced significantly in the mid-20th century with the electron microscopic discovery of tight junctions in 1963, followed by 1980s research elucidating their role in modulating permeability. In healthy individuals, baseline intestinal permeability maintains low paracellular , restricting the of macromolecules to negligible levels and thereby minimizing endotoxemia ; in contrast, hyperpermeability states elevate this , facilitating harmful substance translocation and contributing to immune dysregulation.

Structure of the Intestinal Barrier

The intestinal barrier consists of multiple layered components that collectively prevent the of harmful substances while allowing selective . The outermost layer is the , produced by goblet cells, which forms a protective over the epithelial surface. Beneath this lies a single monolayer of epithelial cells, including enterocytes for and Paneth cells for defense, sealed by tight junctions to regulate paracellular permeability. The underlying contains immune cells that contribute to barrier surveillance and response. The mucus layer is a gel-like structure primarily composed of glycoproteins, such as secreted by goblet cells, which expands upon release to create a stratified barrier. In the colon, it features an inner adherent layer that remains largely sterile and an outer loose layer harboring commensal microbes, while the has a single, thinner, non-stratified mucus layer. This hierarchical organization, with trefoil factor family peptides like TFF3 and FCGBP enhancing stability through disulfide bonds, physically separates luminal contents from the . The epithelial monolayer is a contiguous sheet of polarized cells, dominated by enterocytes that form the absorptive surface, alongside goblet cells for production and Paneth cells located in crypts that secrete . These cells maintain structural integrity through apical microvilli for increased surface area and basolateral connections to the . The , a layer beneath the , houses immune cells such as dendritic cells, macrophages, and lymphocytes, which monitor and reinforce the barrier without direct luminal exposure. Tight junctions form intricate protein complexes at the apical-lateral borders of epithelial cells, sealing paracellular spaces to restrict and solute diffusion. Core transmembrane proteins include claudins (a family of over 20 members forming selective strands), (a tetraspan protein stabilizing the junction), and junctional adhesion molecules (JAMs) that mediate cell-cell . Scaffolding proteins like zonula occludens-1 (ZO-1) link these to the actin cytoskeleton, while adherens junctions involving E-cadherin provide additional mechanical stability. Transcellular permeability is facilitated by elements within epithelial cells, including endocytic vesicles that enable vesicular transport of macromolecules via and transporters embedded in the . For instance, PEPT1, a proton-coupled transporter on the apical of enterocytes, structurally consists of 12 transmembrane helices and facilitates the uptake of di- and tripeptides into the cell interior. These components allow controlled passage through the cell without compromising the overall barrier. Regional variations in barrier structure adapt to functional demands, with the featuring a looser mucus layer and more permeable optimized for , whereas the colon exhibits a thicker, stratified and tighter junctions (e.g., higher expression of sealing claudins) to handle byproducts and denser . These differences arise from variations in cell composition, such as greater density in the colon and prominence in the . The intestinal barrier undergoes significant maturation during infancy, driven by microbial that begins at birth and shapes structural development. Early establishment promotes epithelial proliferation, assembly, and production, with commensal bacteria inducing antimicrobial peptide secretion from Paneth cells and enhancing function. Without timely , such as in germ-free models, the barrier remains immature with increased permeability, but restoration via transfer can normalize these features within critical postnatal windows.

Mechanisms of Permeability

Intestinal permeability is primarily governed by two distinct pathways: the paracellular route, which facilitates the of small ions and hydrophilic molecules between adjacent epithelial cells via s, and the transcellular route, which enables the transport of solutes and larger entities across the interior of the cells. The paracellular pathway is a passive process regulated by the dynamic assembly and disassembly of strands, allowing selective passage based on molecular size and charge while maintaining barrier integrity under normal physiological conditions. This route is crucial for the absorption of ions like sodium and small nutrients, with permeability tightly controlled to prevent uncontrolled leakage. In contrast, the transcellular pathway involves energy-dependent mechanisms that exploit the apical-basolateral polarity of intestinal epithelial cells. Active transport carriers, such as the sodium-glucose linked transporter 1 (SGLT1), mediate the uptake of glucose from the luminal side coupled with sodium influx, followed by facilitated diffusion across the basolateral membrane. For larger molecules, including proteins and lipids, endocytosis allows internalization at the apical membrane and subsequent transcytosis or exocytosis at the basolateral side, ensuring directed vectorial transport. This pathway supports the absorption of essential macronutrients and maintains cellular homeostasis by coupling transport to metabolic energy. The intestinal barrier exhibits selective permeability, distinguishing between paracellular and transcellular routes based on molecular properties. Paracellular transport is generally limited to hydrophilic molecules smaller than approximately 500 , with charge selectivity influenced by proteins that form cation- or anion-preferring pores. Transcellular mechanisms, however, are often active and energy-dependent for polar solutes, while passive predominates for lipophilic compounds that partition into the . This selectivity ensures efficient nutrient uptake without compromising barrier function against pathogens or toxins. Homeostatic balance in intestinal permeability is maintained through feedback mechanisms involving signaling, which fine-tunes epithelial flux in response to luminal contents and immune cues. For instance, the anti-inflammatory interleukin-10 (IL-10) reinforces barrier integrity by suppressing pro-inflammatory signals that could widen junctions, thereby adjusting permeability to preserve physiological equilibrium. Such regulatory loops prevent excessive paracellular leakage while supporting adaptive transport needs. Quantitative assessment of permeability often employs the apparent permeability coefficient (P_app) in models to characterize across epithelial monolayers. This metric is calculated as P_{\text{app}} = \frac{dQ/dt}{A \cdot \Delta C}, where dQ/dt represents the steady-state rate, A is the surface area, and \Delta C is the concentration difference across the barrier. This formula provides a standardized measure of efficiency, aiding in the distinction between passive and carrier-mediated processes under controlled conditions.

Regulation and Measurement

Physiological and Pathological Modulators

Intestinal permeability is tightly regulated by various physiological factors that maintain barrier integrity under normal conditions. Microbiota-derived (SCFAs), such as butyrate, enhance assembly and function in intestinal epithelial cells, thereby strengthening the paracellular barrier and reducing permeability. These metabolites, produced through bacterial fermentation of dietary fibers, promote the expression of proteins like zonula occludens-1 and , contributing to overall gut . Similarly, the glucagon-like peptide-2 (GLP-2), secreted by enteroendocrine L cells in response to nutrient intake, promotes intestinal barrier integrity by increasing transepithelial electrical resistance and enhancing both paracellular and pathways. GLP-2 achieves this through upregulation of proteins and cytoskeletal elements, preventing excessive leakage while supporting nutrient absorption. Recent studies as of 2024 have further highlighted the role of enteroendocrine cells in regulating intestinal barrier permeability through hormone secretion and direct epithelial interactions. Nutritional components also play a critical role in modulating permeability, with certain nutrients supporting barrier maintenance and others potentially disrupting it in susceptible individuals. serves as the primary metabolic fuel for , the absorptive cells of the , where it supports cellular proliferation and repair, thereby preserving integrity and reducing permeability. Supplementation with has been shown to decrease intestinal permeability in stressed or inflamed states by maintaining enterocyte energy supply and mitigating epithelial damage. In contrast, gluten-derived peptides like can trigger release from intestinal epithelial cells in genetically predisposed individuals, leading to reversible opening of s and increased paracellular permeability. This -mediated effect occurs independently of overt but is amplified in conditions like celiac disease, where binding to chemokine receptor initiates the signaling cascade. Pathological conditions often involve modulators that compromise the intestinal barrier, primarily through inflammatory and oxidative mechanisms. Proinflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ), disrupt tight junction structure and function during intestinal inflammation by downregulating key proteins like claudins and occludins, resulting in elevated paracellular permeability. These cytokines act synergistically to alter the actin cytoskeleton and increase myosin light chain phosphorylation, further impairing barrier selectivity. Nonsteroidal anti-inflammatory drugs (NSAIDs) exacerbate this by inducing oxidative stress in the intestinal mucosa, which damages enterocytes and elevates permeability through lipid peroxidation and mitochondrial dysfunction. Chronic alcohol exposure similarly promotes hyperpermeability via oxidative stress, where ethanol metabolism generates reactive oxygen species that degrade tight junction complexes and facilitate bacterial translocation. External stressors, including psychological and infectious challenges, can acutely alter permeability through neuroendocrine and microbial pathways. activates the hypothalamic-pituitary-adrenal axis, releasing corticotropin-releasing factor (), which increases intestinal permeability in a mast cell-dependent manner by promoting release and protease activity that loosen s. This CRF-mediated effect mimics acute stress responses and can be blocked by mast cell stabilizers, highlighting the gut-brain axis's role in barrier regulation. Bacterial infections, such as those caused by species, further increase permeability by upregulating "leaky" proteins like claudin-2 and disrupting epithelial integrity through effector proteins and toxins. In this context, enteric pathogens trigger secretion as part of the host's innate response, which transiently enhances paracellular to expel microbes but can lead to prolonged barrier defects if unresolved. Genetic factors underlie certain hereditary defects in intestinal permeability by impairing formation. Mutations in the CLDN1 gene, which encodes claudin-1—a major component of tight junctions—disrupt epithelial sealing and are linked to hereditary conditions featuring increased paracellular leakage, such as neonatal sclerosing cholangitis associated with . These mutations reduce claudin-1 polymerization in the junctional strand, compromising barrier selectivity and allowing inappropriate passage of luminal contents. Claudin-1 deficiency thus exemplifies how genetic variations can predispose to baseline permeability alterations, independent of environmental triggers. As of 2025, emerging research identifies (MLCK) as a key regulator of permeability, representing a promising therapeutic target for restoring in inflammatory conditions.

Assessment Methods

Intestinal permeability is assessed using a variety of , , and biomarker-based techniques to evaluate the integrity of the intestinal barrier. These methods aim to quantify paracellular and transcellular pathways through which solutes and macromolecules may pass, providing insights into barrier dysfunction. In vivo approaches often involve non-invasive probe administration, while ex vivo and imaging techniques offer more direct measurements but may require invasive sampling. Limitations across methods include inter-individual variability in probe absorption, metabolism, and renal clearance, which can confound results. Probe-based tests represent a cornerstone of assessment, particularly the lactulose-mannitol (L:M) dual-sugar test, which differentiates paracellular from transcellular permeability. In this procedure, participants ingest a solution containing 5 g (a marker for larger paracellular pores, 342 ) and 2 g (a for smaller transcellular pores, 182 ) after overnight , followed by collection of all urine over 5 hours. Urinary concentrations are measured via , and the L:M excretion ratio is calculated; a normal ratio is approximately 0.03 (range 0.003-0.25 in healthy individuals), with elevated ratios (>0.03) indicating increased paracellular leakage, as recovery exceeds that of in barrier-compromised states. This test is widely used due to its non-invasiveness and ability to reflect small intestinal permeability, though gastric emptying and renal function can influence outcomes. Other probes, such as glycols (/1000) or 51Cr-EDTA, provide complementary site-specific data but share similar variability issues. Biomarker assays offer indirect, non-invasive evaluation through circulating or fecal indicators of barrier integrity. , a 47 kDa protein that modulates tight junctions, is commonly measured via (); levels above 30-34 ng/mL suggest hyperpermeability, correlating with conditions like celiac disease where zonulin upregulation increases intestinal leakiness. However, assay reliability is debated, as some commercial kits may detect unrelated proteins like , leading to calls for standardized validation. -binding protein (LBP), an acute-phase reactant, assesses microbial translocation and endotoxemia by binding bacterial (); elevated LBP levels (>10 μg/mL in some cohorts) indicate barrier breach allowing luminal bacteria-derived products into circulation, though and can elevate it independently. These biomarkers provide accessible screening but lack specificity for permeability alone. In vivo imaging techniques enable real-time visualization of barrier dynamics. Confocal laser (CLE), performed during , uses fluorescein sodium (100 mg IV) to highlight epithelial gaps and cell shedding; increased fluorescein leakage into the (quantified as cell junctions extruded or fluorescein leak score) signifies local permeability defects, as validated in where it predicts relapse with high sensitivity. In animal models, FITC-dextran gavage (e.g., 4 kDa FITC-dextran at 60 mg/100 g body weight) measures fluorescence 1-4 hours post-oral administration via fluorimetry; elevated levels (>0.5 μg/mL) denote small intestinal permeability, offering a translational tool for preclinical studies though limited by species-specific gut transit. These methods provide but require specialized equipment and expertise. Ex vivo assessments, such as Ussing chambers, allow precise quantification of and solute in isolated tissue. Biopsies or mucosal sheets from are mounted between chambers filled with oxygenated at 37°C, enabling measurement of transepithelial electrical resistance (TEER, normal >50 Ω·cm² for human ) via Ag/AgCl electrodes and of probes like 4 kDa FITC-dextran or 51Cr-EDTA across the tissue (apparent permeability coefficient, Papp <10^{-6} cm/s in intact barriers). Short-circuit current recordings further assess ; reduced TEER or increased indicates permeability alterations, distinguishing paracellular (e.g., EDTA) from transcellular routes. This gold-standard technique is ideal for mechanistic studies but is invasive and reflects only the sampled region. Emerging multi-omics approaches integrate permeability markers with broader profiling for holistic insights. For instance, combining fecal calprotectin (a neutrophil-derived marker, normal <50 μg/g) with serum or probe-derived data via , , and reveals microbial-host interactions driving barrier dysfunction, as seen in cohorts where elevated calprotectin correlates with dysbiosis-linked permeability. These high-throughput methods, using samples, overcome single-marker limitations but face challenges in and due to probe variability and confounding factors like composition. As of 2025, advancements include systematic evaluations of intestinal permeability assays like models and multi-sugar assays for non-invasive whole-gut assessment, enhancing precision in clinical and preclinical research.

Clinical and Pathophysiological Implications

Associations with Diseases

Altered intestinal permeability, often termed "leaky gut," has been implicated in the pathogenesis of various gastrointestinal (GI) disorders. In celiac disease, ingestion triggers a rapid spike in release, a key regulator of s, leading to increased epithelial permeability that precedes the onset of and villous atrophy. This early permeability allows peptides to translocate across the barrier, initiating immune responses. Similarly, in (IBD), particularly , hyperpermeability is observed in a substantial proportion of patients, with tumor necrosis factor-alpha (TNF-α) playing a central role by disrupting proteins like and claudins, thereby exacerbating mucosal . Antitumor necrosis factor-α therapies have been shown to restore barrier in these cases. Beyond GI conditions, increased intestinal permeability correlates with metabolic diseases. In type 1 and , leaky gut facilitates bacterial translocation and endotoxemia, contributing to and β-cell dysfunction; studies indicate that itself drives barrier impairment, with associations noted in patients exhibiting poor glycemic control. In , particularly among individuals with a (BMI) greater than 30 kg/m², the lactulose:mannitol (L:M) ratio—a marker of paracellular permeability—is elevated, linking barrier dysfunction to systemic low-grade and components. Recent studies from 2023 to 2025 highlight further associations. A 2024 found that 70% of investigations in with diarrhea (IBS-D) reported elevated intestinal permeability compared to controls, suggesting it as a contributing factor to symptom severity. In liver , particularly decompensated cases, serum levels are elevated, indicating heightened permeability that promotes bacterial translocation and complications like . Mechanistically, antigen translocation due to compromised permeability triggers Th17 cell responses, amplifying ; in disease, this occurs pre-symptomatically, with gliadin-induced release enabling access to immune cells. Epidemiological data from 2025 studies suggest that hyperpermeability increases in aging populations, potentially driven by age-related declines in integrity, increasing susceptibility to these diseases.

Role in Systemic Inflammation and Comorbidities

Intestinal hyperpermeability facilitates the translocation of bacterial lipopolysaccharides (LPS) from the gut lumen into the systemic circulation, a process known as metabolic endotoxemia, which triggers low-grade chronic inflammation. This occurs primarily through the activation of Toll-like receptor 4 (TLR4) on immune cells, leading to the release of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). Elevated serum LPS levels, often exceeding 0.5 EU/mL in affected individuals, serve as a biomarker for this endotoxemia and correlate with sustained systemic inflammatory responses that extend beyond the gastrointestinal tract. Gut dysbiosis exacerbates this by increasing the production of pro-inflammatory microbial components, further compromising barrier integrity and perpetuating the inflammatory cycle. This links intestinal hyperpermeability to various comorbidities, notably (CVD). A 2024 meta-analysis of clinical studies demonstrated that patients with CVD exhibit significantly elevated markers of intestinal permeability, such as and LPS-binding protein (LBP), indicating barrier dysfunction as a contributing factor to and endothelial . In , circulating levels positively correlate with symptom severity, as measured by scores above 20, suggesting that leaky gut contributes to neuroinflammatory processes via the gut-brain axis. These associations highlight how intestinal barrier impairment drives extra-intestinal pathologies through immune activation and microbial metabolite dissemination. Along the gut-brain axis, hyperpermeability promotes neuroinflammation in conditions like by allowing neurotoxic metabolites and inflammatory mediators to reach the . Studies from 2024 indicate that increased intestinal permeability in Parkinson's patients correlates with α-synuclein aggregation and dopaminergic neuron loss, exacerbated by microbiota-derived and LPS that amplify microglial activation. This bidirectional interplay underscores the role of gut barrier dysfunction in neurodegenerative progression. In aging populations, intestinal hyperpermeability contributes to chronic low-grade , with 2025 cohort data revealing elevated LBP levels in elderly individuals, linking gut leakage to frailty and . Similarly, in (ICU) patients, initiates a vicious loop where microbial shifts increase permeability, leading to endotoxemia, , and multi-organ dysfunction; this permeability-dysbiosis cycle is a common feature in critically ill cases. Regarding cancer, intestinal hyperpermeability may influence tumor progression through and immune dysregulation. Leaky gut thus facilitates a pro-metastatic microenvironment by enabling the systemic spread of oncogenic signals and inflammatory factors.

Therapeutic and Modulatory Strategies

Dietary and Interventions

Dietary interventions play a significant role in modulating intestinal permeability by influencing the and epithelial . High-fiber intake, typically recommended at 25-30 grams per day, promotes the production of (SCFAs) such as butyrate through fermentation by , which in turn enhances integrity and reduces paracellular permeability. The , rich in fruits, vegetables, whole grains, and healthy fats, has been shown to improve intestinal , with recent , such as the LIBRE trial, demonstrating improved intestinal associated with increased plasma n-3 polyunsaturated fatty acids in participants adhering to this pattern. Incorporating fermented foods, such as , , and , supports restoration and helps maintain epithelial barrier by increasing microbial diversity and reducing inflammation-associated permeability. Regarding specific supplements, randomized controlled trials (RCTs) on at 10 grams per day have yielded inconsistent results for improving permeability, with some showing no overall effect despite benefits in select populations. In contrast, omega-3 fatty acids, particularly 2 grams of combined (EPA) and (DHA) daily, have demonstrated benefits in (IBD), reducing inflammation and supporting barrier repair as evidenced by RCTs. Lifestyle modifications further contribute to permeability regulation. Moderate aerobic exercise, such as brisk walking for 150 minutes per week, prevents stress-induced increases in permeability by mitigating inflammatory responses, though high-intensity sessions exceeding 80% of VO2 max can exacerbate leaks due to splanchnic hypoperfusion. Stress reduction techniques, including mindfulness-based practices, lower corticotropin-releasing factor (CRF) levels, which otherwise promote mast cell degranulation and barrier disruption. Alcohol abstinence rapidly improves permeability markers; for instance, one week of withdrawal in patients with alcohol-related liver disease significantly increases zonulin levels toward normal (p < 0.05) and reduces intestinal fatty acid-binding protein (I-FABP) levels, indicating restored tight junction function. In individuals with metabolic dysfunction-associated steatohepatitis (MASH), achieving 5-10% body weight reduction through diet and exercise enhances permeability, as shown in 2024 studies linking weight loss to decreased microbial translocation and improved barrier integrity.

Pharmacological and Microbial Therapies

Pharmacological therapies targeting intestinal permeability primarily focus on modulating proteins and reducing inflammation to restore barrier integrity. Larazotide acetate, a antagonist, inhibits the disassembly of s induced by in celiac disease, thereby decreasing paracellular permeability. In a phase II , the 0.5 mg dose of larazotide acetate led to a reduction of 50% or more in weekly average celiac disease abdominal domain scores from baseline for at least 6 weeks in treated patients compared to (P = 0.029). Although advanced to phase III trials for celiac disease, development was discontinued in 2022 due to insufficient efficacy endpoints, but earlier data support its role in symptom alleviation alongside a . Corticosteroids, such as , are employed for acute flares in (IBD) to suppress inflammation and enhance epithelial barrier function. , with its topical action in the gut, restores intestinal permeability as measured by the lactulose/ ratio in patients during active inflammation. 5-Aminosalicylic acid (5-ASA) derivatives, like mesalamine, stabilize s in by modulating junctional complexes, including upregulation of and claudin-1 expression, thereby reducing epithelial permeability and promoting barrier repair. Microbial therapies leverage the gut microbiome to reinforce intestinal barrier function through probiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT). , particularly strains like Lactobacillus rhamnosus GG (LGG) at doses of 10^9 CFU/day, improve epithelial integrity by enhancing protein expression and reducing permeability in inflammatory conditions. A 2025 systematic review and of 46 randomized controlled trials involving over 3,200 participants demonstrated that , including LGG, significantly lowered serum levels (standardized mean difference [SMD] = -0.49, 95% CI: -0.79 to -0.18) and (LPS) (SMD = -0.54, 95% CI: -1.01 to -0.07), indicating reduced intestinal permeability. Synbiotics, combining with prebiotics, further bolster barrier function in leaky gut models by promoting beneficial microbial growth and decreasing release, as evidenced in clinical and preclinical studies where they attenuated stressor-induced permeability increases. Postbiotics, such as culture supernatants from Lactobacillus reuteri ZJ617, prevent LPS-induced damage to the intestinal barrier by enhancing antioxidant activity and integrity, mitigating and permeability in mouse models of acute injury. Emerging microbial interventions include FMT, which transfers donor to reshape the recipient's gut ecosystem and repair barrier defects. In IBS with (IBS-D), clinical trials from 2023 to 2025 report FMT efficacy rates of 30-65%, defined by at least 30% improvement in IBS severity scoring system scores, with endoscopic or delivery showing superior outcomes over in reducing symptoms and permeability markers. Preclinical data also highlight arginase-2 inhibitors, such as nor-NOHA, which prevent aging-related intestinal leaks by preserving availability and stability in rodent models, offering potential for age-associated barrier dysfunction. These therapies collectively aim to normalize permeability, with efficacy often assessed via urinary sugar ratios or serum biomarkers, complementing broader modulatory strategies.

Controversies and Emerging Research

Leaky Gut Syndrome

, as described in , refers to a hypothetical condition in which damage to the intestinal lining allows undigested food particles, toxins, and to "leak" into the bloodstream, purportedly triggering a wide array of symptoms including chronic fatigue, food allergies, skin issues, and joint pain. This concept gained prominence in the through nutritionists and alternative health practitioners who promoted it as an underlying cause of diverse health problems, often without established diagnostic criteria or standardized testing protocols. Proponents typically attribute the syndrome to factors like poor diet, stress, or use, suggesting it leads to and immune dysregulation. As of 2025, the scientific consensus holds that is not recognized as a formal , distinguishing it from the well-documented phenomenon of increased intestinal permeability observed in specific gastrointestinal diseases such as celiac disease or . While alterations in gut barrier function are real and measurable in certain pathologies, the broader claims of as a standalone entity lack robust evidence, including the absence of validated biomarkers or specific treatments beyond general gut health support. For instance, proposed therapies akin to unproven interventions like have not demonstrated efficacy in controlled studies for this purported syndrome. Common misconceptions surrounding include unsubstantiated causal links to conditions, where proponents claim toxin leakage directly initiates , despite 2025 reviews emphasizing that such associations require evidence of causation and reliable biomarkers, which are currently absent. These overreaches often ignore that while intestinal hyperpermeability can contribute to immune responses in established diseases, the syndrome's systemic effects—such as widespread allergies or —are not supported by mechanistic studies without objective permeability assessments. In research contexts, the term "leaky gut" is employed descriptively to denote barrier defects in experimental models or clinical conditions, rather than endorsing the as a diagnostic category. The public perception of has fueled a substantial market for dietary supplements, including and glutamine-based products, estimated at over $2.3 billion globally in 2024, driven by marketing. However, randomized controlled trials evaluating these supplements for syndrome-related symptoms generally report benefits no greater than , with only limited evidence for permeability improvements in targeted subgroups, underscoring the need for evidence-based approaches over anecdotal remedies.

Recent Advances and Future Directions

Recent research has illuminated the intricate connections within the gut-organ axis, particularly the role of intestinal barrier dysfunction in cardiovascular diseases through lipopolysaccharide (LPS) translocation. A 2024 review highlights how impaired gut permeability facilitates LPS leakage, activating systemic inflammation via Toll-like receptor 4 (TLR4) pathways, thereby exacerbating heart failure and atherosclerosis. Similarly, studies in 2025 have demonstrated that gut microbiota dysbiosis amplifies cardiac remodeling by promoting LPS-induced myocardial inflammation. In aging populations, maturation influences rectal and intestinal permeability, with age-related shifts leading to barrier weakening. A 2025 study in aged mice revealed microbiota-dependent increases in intestinal permeability and reduced protein ZO-1 expression, underscoring the need for microbiome-targeted interventions to mitigate frailty. Between 2023 and 2025, key highlights include evidence that diet-induced weight loss reverses permeability in metabolic dysfunction-associated (MASH); a 2024 clinical study showed significant reductions in intestinal permeability markers alongside improvements in MASH following caloric restriction. Postbiotics have emerged as promising agents for barrier maintenance, with a 2025 trial demonstrating that postbiotic supplementation enhances integrity and reduces in healthy adults. Additionally, AI-driven multi-omics approaches are enabling personalized , integrating genomic, metagenomic, and metabolomic data to predict permeability-related susceptibility. Despite these advances, critical gaps persist, particularly in distinguishing causal from correlative roles of intestinal permeability in the gut-brain axis and . Limited longitudinal data hinder causal inference, as most evidence relies on cross-sectional associations between barrier dysfunction, alterations, and depressive symptoms. Beyond , standardized biomarkers remain underdeveloped; while fecal calprotectin and serum intestinal fatty acid-binding protein (I-FABP) show promise, their validation across diverse populations is inconsistent, complicating clinical translation. Looking ahead, targeted modulators of tight junctions, such as agents enhancing claudin expression, hold potential for restoring barrier function in inflammatory conditions. Microbiome engineering via CRISPR offers innovative avenues, enabling precise editing of gut bacteria to bolster barrier integrity and reduce pathogen translocation. Ongoing clinical trials are exploring preventive strategies in at-risk groups like the elderly, including prebiotic interventions to improve permeability and mitigate age-related inflammation. Challenges include ethical concerns in probe studies, where invasive permeability assessments raise issues of participant burden and safety in vulnerable cohorts. Furthermore, the underrepresentation of diverse ethnic and socioeconomic groups in limits generalizability, necessitating inclusive cohorts to address global variations in permeability-related diseases.

References

  1. [1]
    Intestinal permeability – a new target for disease prevention and ...
    Intestinal permeability, which is a feature of intestinal barrier function, is increasingly recognized as being of relevance for health and disease.
  2. [2]
    The Role of Intestinal Permeability in Gastrointestinal Disorders and ...
    Aug 26, 2021 · Abstract. An increased intestinal permeability has been described in various gastrointestinal and non-gastrointestinal disorders.
  3. [3]
    Intestinal permeability disturbances: causes, diseases and therapy
    Sep 28, 2024 · Nowadays, a pathological increase in the permeability of the intestinal barrier (the so-called leaky gut) is increasingly being diagnosed.
  4. [4]
    The Role of Intestinal Permeability in Gastrointestinal Disorders and ...
    An increased intestinal transcellular permeability found in different disease conditions has been postulated to be critical for the migration of bacteria and ...
  5. [5]
    The Intestinal Barrier and Current Techniques for the Assessment of ...
    Intestinal permeability is defined as the passage of a solute through a simple membrane and can be measured by recording the passage of permeability markers ...
  6. [6]
    Intestinal permeability – a new target for disease prevention and ...
    Nov 18, 2014 · Intestinal permeability, which is a feature of intestinal barrier function, is increasingly recognized as being of relevance for health and disease.
  7. [7]
    Gut microbiota, intestinal permeability, and systemic inflammation
    Jul 28, 2023 · The maintenance of a stable intestinal barrier is crucial to prevent luminal substances and pathogens from entering the internal environment.
  8. [8]
    The History of the Intestinal Microbiota and the Gut-Brain Axis - PMC
    Dec 15, 2022 · In the 19th century, physicians tended to blame the gastrointestinal system for its influence on mind [1,9]. In the 20th century, scientists ...
  9. [9]
    Tight junctions: from molecules to gastrointestinal diseases - NIH
    Increased intestinal permeability can be due to tight junction-dependent and tight junction-independent mechanisms. Two tight junction-dependent permeability ...
  10. [10]
    Mechanisms regulating intestinal barrier integrity and its ... - Nature
    Aug 16, 2018 · Tight junction complexes are the rate-limiting factor for paracellular permeability; they are programmed to rapidly open and seal the barrier in ...
  11. [11]
    Intestinal mucus components and secretion mechanisms: what we ...
    Apr 3, 2023 · The hierarchical structure of the mucus barrier is key in preventing bacterial penetration and blocking direct contact of bacteria and the ...
  12. [12]
    Layered defense: how mucus and tight junctions seal the intestinal ...
    Jul 13, 2017 · This review focuses on how intestinal mucus and intercellular tight junctions (TJs) act together to maintain the integrity of the colonic barrier.
  13. [13]
    Intestinal barrier permeability: the influence of gut microbiota ...
    Mucus and epithelium are the most important components of the intestinal barrier that limit the development of inflammation. The mucous layer consists of two ...
  14. [14]
    Imaging therapeutic peptide transport across intestinal barriers - PMC
    These pathways include (Fig. 1, right) active transcytosis mediated vesicular transport, passive transcellular transport through epithelial cells, and ...Missing: PEPT1 | Show results with:PEPT1
  15. [15]
    Bacterial colonization and intestinal mucosal barrier development
    This review summarizes recent advances in understanding the complex ecosystem of intestinal microbiota and its role in regulating intestinal barrier function.
  16. [16]
    Intestinal Barrier Function: Molecular Regulation and Disease ...
    Transcellular permeability is associated with solute or water movement through intestinal epithelial cells. Paracellular permeability is associated with ...
  17. [17]
    Paracellular permeability and tight junction regulation in gut health ...
    Apr 25, 2023 · Intestinal permeability can reflect contributions of three distinct pathways: the pore pathway, the leak pathway and the unrestricted pathway ( ...
  18. [18]
    Role of Stress on Driving the Intestinal Paracellular Permeability
    Nov 18, 2023 · The paracellular route allows the passage of ions and small molecules between the spaces of the lateral membrane of the epithelial cells (4).
  19. [19]
    Na(+)-D-glucose cotransporter SGLT1 is pivotal for intestinal ...
    Dec 12, 2011 · The primary transporters that mediate transcellular movements of d-glucose in small intestine have been identified and include the Na+-d-glucose ...
  20. [20]
    Breaking the Barrier: The Role of Gut Epithelial Permeability in ... - NIH
    1). The paracellular and the transcellular endocytosis pathways are the most relevant for intestinal permeability, with most research to date focused on the ...
  21. [21]
    Integrative physiology of transcellular and paracellular intestinal ...
    Jul 15, 2017 · Transcellular absorption: glucose from the lumen (top of the figure) can move across the epithelium by uptake via the Na+-coupled glucose ...
  22. [22]
    Structure-function analysis of tight junction-directed permeation ...
    The intestinal epithelium is a semi-permeable barrier that limits the non-selective absorption of hydrophilic molecules with a molecular weight of > ∼500 Da [1] ...
  23. [23]
    Determination and prediction of permeability across intestinal ...
    Jan 14, 2020 · Apparent permeability coefficients (Papp) across a human intestinal epithelial Caco-2 cell monolayer were measured for a range of industrial ...Missing: formula | Show results with:formula
  24. [24]
    Short-Chain Fatty Acids Manifest Stimulative and Protective Effects ...
    Aug 23, 2018 · Short-Chain Fatty Acids Manifest Stimulative and Protective Effects on Intestinal Barrier Function Through the Inhibition of NLRP3 Inflammasome ...Missing: enhance | Show results with:enhance
  25. [25]
    Biological Function of Short-Chain Fatty Acids and Its Regulation on ...
    Oct 18, 2021 · This review summarizes the biological functions of SCFAs and their role in gut health, as well as nutritional strategies to regulate SCFA ...
  26. [26]
    Glucagon-like peptide-2 enhances intestinal epithelial ... - PubMed
    GLP-2 enhances intestinal epithelial barrier function by affecting both paracellular and transcellular pathways and thus may be of therapeutic value.
  27. [27]
    Glucagon-Like Peptide-2 Reduces Intestinal Permeability But Does ...
    GLP-2 has also been shown to enhance transcellular and paracellular gut barrier function, by increasing transepithelial resistance in the normal gut and after ...
  28. [28]
    Glutamine supplementation decreases intestinal permeability and ...
    Conclusions: GLN had a positive impact on the intestinal barrier by reducing permeability and bacterial translocation to physiologic levels and preserving ...
  29. [29]
    Glutamine and whey protein improve intestinal permeability and ...
    Glutamine, the major fuel for the enterocytes, may improve IP. Aim: We evaluated the effect of oral glutamine on IP and intestinal morphology in patients with ...
  30. [30]
    Gliadin, zonulin and gut permeability: Effects on celiac ... - PubMed
    We concluded that gliadin activates zonulin signaling irrespective of the genetic expression of autoimmunity, leading to increased intestinal permeability to ...
  31. [31]
    Interferon-gamma and tumor necrosis factor-alpha disrupt epithelial ...
    We found that TNF-alpha and IFN-gamma synergistically affected the epithelial barrier and disrupted the structure of TJs.
  32. [32]
    Proinflammatory cytokines disrupt epithelial barrier function by ...
    Our findings for the first time clearly separate the proapoptotic effects of IFN-gamma and TNF-alpha from their abilities to disrupt barrier function.
  33. [33]
    Relevance of oxidative stress for small intestinal injuries induced by ...
    Dec 13, 2024 · Nonaspirin NSAIDs-induced intestinal mucosal injury is associated with antioxidant systems, resulting in increased oxidative stress.
  34. [34]
    Evidence that chronic alcohol exposure promotes intestinal ... - NIH
    Evidence that chronic alcohol exposure promotes intestinal oxidative stress, intestinal hyperpermeability and endotoxemia prior to development of alcoholic ...
  35. [35]
    Psychological stress and corticotropin-releasing hormone increase ...
    Acute psychological stress increases small intestinal permeability in humans. Peripheral CRH reproduces the effect of stress and DSCG blocks the effect of ...Missing: CRF | Show results with:CRF
  36. [36]
    Psychosocial stress-induced intestinal permeability in healthy humans
    Chronic peripheral administration of corticotropin-releasing factor causes colonic barrier dysfunction similar to psychological stress. Am. J. Physiol ...
  37. [37]
    Salmonella infection upregulates the leaky protein claudin-2 in ...
    We have shown that Salmonella targets the tight junction protein claudin-2 to facilitate bacterial invasion. We speculate that this disruption of barrier ...
  38. [38]
    Knockout animals and natural mutations as experimental and ...
    Many pathologies of knockout mice and mutations of the claudin and tricellulin genes in hereditary human diseases have been interpreted as defects in TJ ...2.4. Claudin-14 Gene... · 2.6. Claudin-16 Mutations... · 4. Tricellulin Mutations
  39. [39]
    Claudin-1 Regulates Intestinal Epithelial Homeostasis through the ...
    Jul 7, 2014 · Claudin-1 helps regulate intestinal epithelial homeostasis through the regulation of Notch-signaling. An upregulated claudin-1 expression induces MMP-9 and p- ...Figure 2. Claudin-1... · Figure 3. Claudin-1 Tg Mice... · Figure 7. Claudin-1...Missing: CLDN1 | Show results with:CLDN1
  40. [40]
    How to Perform and Interpret Intestinal Permeability - NIH
    Lactulose and mannitol ratio is the most commonly used test for assessment of small intestinal permeability and the most reliable method for measurement of ...Missing: review | Show results with:review<|control11|><|separator|>
  41. [41]
    Methods to determine intestinal permeability and bacterial ... - NIH
    Intestinal permeability can be assessed through enteral administration of non-digestible markers, which ideally should cross the mucosal barrier by non-mediated ...
  42. [42]
    Fecal zonulin is elevated in Crohn's disease and in cigarette smokers
    For serum zonulin, a median value of 34 ng/mL (± 14 ng/mL) was declared as normal for healthy persons. A reference value of 50 µg/g for normal fecal ...
  43. [43]
    Zonulin Upregulation Is Associated With Increased Gut Permeability ...
    May 1, 2006 · The increased zonulin levels correlated with increased intestinal permeability in vivo and changes in claudin-1, claudin-2, and myosin IXB genes ...
  44. [44]
    Local barrier dysfunction identified by confocal laser ... - Gut
    Assessment of barrier function and cell shedding by confocal endomicroscopy promises to be a simple, fast and practical method of predicting relapse of IBD.
  45. [45]
    Small Intestinal Permeability and Gut-Transit Time Determined with ...
    May 28, 2018 · 4-kDa FITC dextran measured in plasma 1 h after its oral application is a marker of small intestinal permeability.
  46. [46]
    The Ussing chamber system for measuring intestinal permeability in ...
    Jun 20, 2019 · The Ussing chamber system can be utilised as a valuable tool for measuring gut integrity. Here we describe step-by-step methodology required to ...
  47. [47]
    A guide to Ussing chamber studies of mouse intestine
    The Ussing chamber provides a physiological system to measure the transport of ions, nutrients, and drugs across various epithelial tissues.
  48. [48]
    Stool multi-omics for the study of host–microbe interactions in ...
    Dec 11, 2022 · Stool-based multi-omics is a promising approach for characterizing host-gut microbiome interactions using deep integration of technologies.
  49. [49]
    Elevated fecal calprotectin is associated with gut microbial dysbiosis ...
    Jun 12, 2024 · Fecal calprotectin is an established marker of gut inflammation in inflammatory bowel disease (IBD). Elevated levels of fecal calprotectin ...Missing: permeability | Show results with:permeability
  50. [50]
    Zonulin as a Biomarker for the Development of Celiac Disease
    Dec 8, 2023 · Intestinal permeability has been implicated in various gastrointestinal diseases, including celiac disease (CeD). CeD is a T cell-mediated ...
  51. [51]
    Gliadin Induces an Increase in Intestinal Permeability and Zonulin ...
    Gliadin induces an MyD88-dependent zonulin release that leads to increased intestinal permeability, a postulated early element in the pathogenesis of celiac ...
  52. [52]
    Intestinal Permeability in Inflammatory Bowel Disease
    A defective mucosal barrier may result in increased intestinal permeability which promotes the exposition to luminal content and triggers an immunological ...
  53. [53]
    TNF-α-induced increase in intestinal epithelial tight junction ...
    These results demonstrate for the first time that TNF-α-induced increase in Caco-2 TJ permeability was mediated by NF-κB activation.
  54. [54]
    Hyperglycemia drives intestinal barrier dysfunction and risk for ...
    Our results mechanistically link hyperglycemia and intestinal barrier function with systemic infectious and inflammatory consequences of obesity and diabetes.
  55. [55]
    The Role for Gut Permeability in the Pathogenesis of Type 1 Diabetes
    Often referred to as a “leaky gut”, its mechanistic impact on the pathogenesis of T1D remains unclear. Hypotheses that this defect influences immune responses ...Missing: HbA1c | Show results with:HbA1c
  56. [56]
    Gut permeability is related to body weight, fatty liver disease, and ...
    These findings suggest that the Lac:Man ratio could be a valuable marker of intestinal permeability in patients with obesity and metabolic disorders. In ...
  57. [57]
    Intestinal Barrier Permeability in Obese Individuals with or without ...
    Sep 3, 2022 · Altered intestinal barrier permeability has been associated with obesity and its metabolic and inflammatory complications in animal models.
  58. [58]
    Intestinal Permeability in Disorders of Gut–Brain Interaction
    Sep 3, 2024 · This review highlights the techniques for assessment of barrier function, appraises evidence for barrier dysfunction in DGBI including mechanistic studies in ...
  59. [59]
    Intestinal permeability assessed by serum zonulin in liver cirrhosis
    Apr 18, 2025 · It plays a crucial role in the rapid, reversible, and dynamic regulation of intestinal permeability, making it a key marker of this process. In ...
  60. [60]
    Serum zonulin levels in patients with liver cirrhosis - NIH
    Increased gut permeability and bacterial translocation play an important role in liver cirrhosis. Zonulin is a recently recognized protein involved in the ...
  61. [61]
    Celiac Disease and Non-celiac Wheat Sensitivity: State of Art of Non ...
    Sep 8, 2020 · Innate and adaptive immune response (IL15, IL8, IELs, Th1/Th17, antibodies production), increased intestinal permeability, Altered expression ...
  62. [62]
    Latest In vitro and in vivo models of celiac disease - PMC - NIH
    The increased intestinal permeability in celiac disease allows for paracellular transfer of gluten derived peptides to the lamina propria, resulting in the ...
  63. [63]
  64. [64]
    Role of Metabolic Endotoxemia in Systemic Inflammation ... - PubMed
    Jan 11, 2021 · LPS activates Toll-like receptor-4 (TLR4) leading to the production of numerous pro-inflammatory cytokines and, hence, low-grade systemic ...
  65. [65]
    A high-fat meal induces low-grade endotoxemia - ScienceDirect.com
    Background: Bacterial endotoxin is a potently inflammatory antigen that is abundant in the human gut. Endotoxin circulates at low concentrations in the ...
  66. [66]
    Gut dysbiosis-derived low-grade endotoxemia: A common basis for ...
    Gut dysbiosis is characterized by bacteria overgrowth that ultimately leads to increased intestinal barrier permeability and translocation of bacteria or ...<|separator|>
  67. [67]
    Intestinal permeability in human cardiovascular diseases - Frontiers
    Jul 16, 2024 · The results of the meta-analysis verified that the intestinal barrier was damaged and intestinal permeability was increased in patients with cardiovascular ...Abstract · Introduction · Methods · Discussion
  68. [68]
    Intestinal Permeability and Depression—A Narrative Review ... - MDPI
    There is ample evidence supporting associations between zonulin and mental illness, although findings remain partially inconclusive.Missing: Beck | Show results with:Beck
  69. [69]
    Microbiota–gut–brain axis and its therapeutic applications ... - Nature
    Feb 16, 2024 · There is a growing recognition of the role of gut microbiome in neurodegenerative diseases. Notably, early microbiome changes were detected in ...
  70. [70]
    Serum lipopolysaccharide binding protein (LBP) and metabolic ...
    Jul 16, 2025 · Our findings suggest a potential association between higher levels of LBP and MetS, with MetS cases exhibiting elevated LBP levels compared to non-MetS ...
  71. [71]
    A review of gut failure as a cause and consequence of critical illness
    Feb 26, 2025 · In critical illness, all elements of gut function are perturbed. Dysbiosis develops as the gut microbial community loses taxonomic diversity and new virulence ...
  72. [72]
    Wistar Scientists Discover p53 Can “Read” Cellular Signals to Direct ...
    Oct 2, 2025 · Wistar Scientists Discover p53 Can “Read” Cellular Signals to Direct Immune Response, Upending 30 Years of Scientific Consensus Press ...Missing: intestinal permeability metastasis 2024
  73. [73]
    Potential effects of gut microbiota on host cancers: focus on ...
    In this review, we discuss how the gut microbiota play a role in human cancer and its application in anticancer therapy.
  74. [74]
    Impact of Diet-Modulated Butyrate Production on Intestinal Barrier ...
    Oct 13, 2018 · A suppressed production of butyrate is crucial, as this short-chain fatty acid (SCFA) can play a key role not only in colonic health and ...
  75. [75]
    High fiber dietary and sodium butyrate attenuate experimental ...
    Mar 22, 2018 · Short chain fatty acids in feces were determined by HPLC. The ratio of CD4 + C25 + Foxp3+ Treg and CD4 + IL-17 + Th17 were evaluated by flow ...
  76. [76]
    Can Diet Alter the Intestinal Barrier Permeability in Healthy People ...
    Jun 14, 2024 · In respect to infants fed formula or breast milk, those breastfed presented a higher lactulose/mannitol ratio, indicating greater IBP.
  77. [77]
    Effect of the Mediterranean diet on the faecal long-chain fatty acid ...
    We recently showed that adherence to the Mediterranean diet increased the proportion of plasma n-3 PUFA, which was associated with an improved intestinal ...
  78. [78]
    Fermented Foods, Health and the Gut Microbiome - PMC
    Apr 6, 2022 · This review clearly shows that fermented foods can affect the gut microbiome in both the short and long term, and should be considered an important element of ...
  79. [79]
    Kefir and Intestinal Microbiota Modulation: Implications in Human ...
    Feb 21, 2021 · Fermented foods are able to modify the composition of the intestinal microbiota, improve the control of intestinal permeability, increase its ...
  80. [80]
    A systematic review and meta-analysis of clinical trials on the effects ...
    Oct 13, 2024 · The meta-analysis demonstrated a significant reduction in intestinal permeability with glutamine supplementation exceeding 30 mg/day for durations of less than ...Missing: 10g | Show results with:10g
  81. [81]
    A systematic review and meta-analysis of clinical trials on the effects ...
    Overall, glutamine supplementation did not significantly affect intestinal permeability, and the meta-analysis demonstrated a significant reduction in ...
  82. [82]
    Omega Fatty Acids and Inflammatory Bowel Diseases: An Overview
    Sep 30, 2019 · The results showed that ω3 fatty acids reduce intestinal inflammation, induce and maintain clinical remission in UC patients.
  83. [83]
    Interplay of n-3 Polyunsaturated Fatty Acids, Intestinal Inflammation ...
    In inflammatory bowel diseases (IBDs), such as ulcerative colitis and Crohn's disease, doses between 1000 and 3000 mg per day of EPA and DHA have been shown to ...3. From The Celiac Immune... · 6. Pufas And The Gut... · 7. N-3 Pufa: Effects On The...
  84. [84]
    Is There an Exercise-Intensity Threshold Capable of Avoiding ... - NIH
    There is some evidence that vigorous endurance training (≥60 min and ≥70% of maximum work capacity) may lead to injury and increased intestinal permeability.
  85. [85]
    Intensity-specific considerations for exercise for patients with ...
    Feb 20, 2023 · showed that IBD patients had an improvement in intestinal permeability after completing moderate-intensity aerobic exercise as measured by ...
  86. [86]
    The effect of a mindfulness-based therapy on different biomarkers ...
    Apr 8, 2020 · CRF also increases intestinal permeability by causing mast cells to release tumour necrosis factor alpha and proteases. Thus, targeting CRF ...Methods · Study Design · Discussion
  87. [87]
    Psychological stress in inflammatory bowel disease - Frontiers
    It has been reported that psychological stress is involved in the permeability, motility, sensitivity, and secretion of the intestine, composition of gut ...
  88. [88]
    Markers of Intestinal Permeability Are Rapidly Improved by Alcohol ...
    May 14, 2021 · Our data suggest that one-week of abstinence improves markers of intestinal barrier function and liver health in ALD patients.
  89. [89]
    Markers of Intestinal Permeability Are Rapidly Improved by Alcohol ...
    May 12, 2021 · Serum markers of intestinal permeability in controls and patients with ALD before and after alcohol withdrawal. (A) Zonulin and (B) I-FABP ...
  90. [90]
    Changes in intestinal permeability and gut microbiota following diet ...
    Sep 28, 2024 · Weight loss improves metabolic dysfunction-associated steatohepatitis (MASH). We investigated whether there were associated changes in ...
  91. [91]
    Changes in intestinal permeability and gut microbiota following diet ...
    Sep 28, 2024 · Preclinical studies support a causal role for the gut microbiota in MASH and suggest that large weight loss reduces markers of intestinal ...
  92. [92]
    [PDF] Larazotide Acetate for Persistent Symptoms of Celiac Disease ...
    CONCLUSIONS: Larazotide acetate 0.5 mg reduced signs and symptoms in CeD patients on a GFD better than a GFD alone. Although results were mixed, this study was ...
  93. [93]
    Phase 3 study of larazotide acetate to treat celiac disease ...
    Jun 21, 2022 · The Phase 3 clinical trial investigating larazotide acetate as a treatment for celiac disease was discontinued today by 9 Meters Biopharma, Inc.Missing: zonulin inhibitor
  94. [94]
    Corticosteroid enhances epithelial barrier function in intestinal ... - NIH
    In one previous study, CS was shown to restore an increased intestinal permeability as determined by the urinary lactulose/mannitol ratio in CD patients [14].
  95. [95]
    Mesalamine and azathioprine modulate junctional complexes and ...
    Feb 26, 2019 · This study provides mechanistic evidence that molecular actions of 5-ASA and AZTP on intestinal epithelia are fundamental in the resolution of barrier ...
  96. [96]
    Reinforcing gut integrity: A systematic review and meta-analysis of ...
    This systematic review and meta-analysis explores the effects of pro- and synbiotic, or prebiotic administration, on various intestinal permeability markers.
  97. [97]
    Human Intestinal Barrier: Effects of Stressors, Diet, Prebiotics, and ...
    In summary, probiotics and synbiotics enhance intestinal barrier function in response to stressor or disease states. Future studies should address the changes ...
  98. [98]
    Unlocking the power of probiotics, postbiotics: targeting apoptosis ...
    Furthermore, the culture supernatant of L. reuteri ZJ617 attenuates lipopolysaccharide (LPS)-induced acute liver injury by strengthening intestinal barrier ...<|control11|><|separator|>
  99. [99]
    Efficacy of faecal microbiota transplantation for patients with irritable ...
    This study demonstrates the effectiveness of FMT in the treatment of IBS. ... The response criteria for IBS-D are an improvement at least 30% in the ...
  100. [100]
    Clinical efficacy and safety of faecal microbiota transplantation in the ...
    Sep 18, 2024 · This study suggests that patients with IBS may benefit from FMT especially when it is administered via endoscopy, nasojejunal tube or rectal enema.
  101. [101]
    Impairments of intestinal arginine and NO metabolisms trigger aging ...
    Intestinal arginine and NO metabolisms could be a target in the prevention of aging-associated intestinal barrier dysfunction and subsequently decline and ' ...
  102. [102]
    Leaky Gut Syndrome: Symptoms, Diet, Tests & Treatment
    Leaky gut syndrome is a hypothetical condition in which your intestines become permeable to toxins that leak into your bloodstream and trigger systemic ...
  103. [103]
    Debunking the Myth of 'Leaky Gut Syndrome' - badgut.org
    Feb 20, 2024 · Some alternative medicine practitioners claim that leaky gut syndrome is a prevalent problem responsible for ill health in many people. However ...
  104. [104]
    Leaky gut: What is it, and what does it mean for you? - Harvard Health
    Sep 12, 2023 · An unhealthy gut lining may have large cracks or holes, allowing partially digested food, toxins, and bugs to penetrate the tissues beneath it.
  105. [105]
    Leaky Gut Syndrome: Myths and Management - PMC
    Leaky gut syndrome is a condition widely popularized in the lay literature, although it is not currently accepted as a formal medical diagnosis.
  106. [106]
    [PDF] Leaky Gut Syndrome: Myths and Management
    May 5, 2024 · Abstract: Leaky gut syndrome is a condition widely popularized in the lay literature, although it is not currently accepted as a formal ...<|separator|>
  107. [107]
    You Probably Don't Have a Leaky Gut - McGill University
    Feb 23, 2024 · Leaky gut syndrome is not a real diagnosis, but if you step through the looking-glass like Alice in Wonderland, it will become real.
  108. [108]
    The Myths and Scientific Realities About the Gut Microbiota - MDPI
    Leaky Gut Syndrome Is a Clearly Defined Clinical Diagnosis. The phrase leaky gut syndrome is often used in popular media and by alternative health ...
  109. [109]
    Leaky gut in systemic inflammation: exploring the link ... - PubMed
    Dec 6, 2024 · This review outlines current literature on the leaky gut in aging, how leaky gut might contribute to systemic inflammation, and the links ...
  110. [110]
    Leaky Gut Supplements Market Research Report 2033
    As per our latest market intelligence, the Global Leaky Gut Supplements market size was valued at $2.3 billion in 2024, and is forecasted to hit $5.1 billion by ...
  111. [111]
    Leaky Gut and the Ingredients That Help Treat It: A Review - MDPI
    A Randomized Clinical Trial (RCT) with a larger sample size is suggested to confirm the positive benefits of using probiotics and glutamine. Even though ...
  112. [112]
    New insights into the intestinal barrier through “gut-organ” axes and ...
    In recent years, the concept of the “gut-organ” axis has gained significant popularity, revealing a profound interconnection between the gut and other organs.
  113. [113]
    Gut microbiota dysbiosis exacerbates heart failure by the LPS-TLR4 ...
    Jul 10, 2025 · Gut microbiota dysbiosis exacerbates heart failure by activating myocardial inflammation through the LPS-TLR4/NF-κB signalling pathway.
  114. [114]
    Age-related alterations in gut homeostasis are microbiota dependent
    Mar 25, 2025 · In the present study, aged mice presented increased intestinal barrier permeability and decreased expression of the tight junction protein ZO-1.Missing: rectal | Show results with:rectal
  115. [115]
    Postbiotic supplementation promotes gut barrier integrity and ...
    Discussion: These findings support the use of postbiotics as a safe and effective nutritional approach to help maintain gastrointestinal and immune health in ...
  116. [116]
    Systems Biology of Human Microbiome for the Prediction of ...
    Sep 1, 2024 · This mini-review explores the relationship between multiomics, precision medicine, and machine learning to improve our understanding of the gut ...
  117. [117]
    Biomarkers of intestinal permeability are linked to incident ...
    Lipopolysaccharide causes an increase in intestinal tight junction permeability in vitro and in vivo by inducing enterocyte membrane expression and localization ...<|control11|><|separator|>
  118. [118]
    Present and Future Therapeutic Approaches to Barrier Dysfunction
    Similarly, in colonic cell lines, EcN EV promoted upregulation of ZO-1 and claudin-14, and induced IL-22 expression reinforcing the intestinal barrier (186).
  119. [119]
    CRISPR-Assisted Probiotic and In Situ Engineering of Gut Microbiota
    May 16, 2025 · The CRISPR/Cas system offers a promising approach to targeted gut microbiome manipulation. There are two major strategies to edit microbiota ...
  120. [120]
    Study Details | Pectin, Aging and Intestinal Barrier Function
    Jul 15, 2016 · As intestinal barrier function may be compromised in the elderly we will investigate whether the effects of pectin on selected parameters of gut ...
  121. [121]
    Assessment of the Effect of Intestinal Permeability Probes (Lactulose ...
    Aug 6, 2025 · Study protocol submitted to Massey University Human Ethics Committee. (PDF). S3 File. Information sheet given to participants.
  122. [122]
    Challenges in IBD Research 2024: Preclinical Human IBD ... - NIH
    We provide a comprehensive overview of current gaps in inflammatory bowel diseases research that relate to preclinical research and deliver actionable ...