Fact-checked by Grok 2 weeks ago

Intestinal epithelium

The intestinal epithelium is a single-layered columnar epithelium that lines the luminal surface of the small and large intestines, serving as a selective barrier that regulates the absorption of nutrients, water, and electrolytes while preventing the entry of pathogens, toxins, and harmful luminal contents. This dynamic tissue, organized into crypt-villus units in the small intestine and crypts in the colon, undergoes rapid renewal every 2–5 days in humans, driven by stem cells at the crypt base to maintain its integrity amid constant exposure to the gut environment. Composed of diverse cell types, including absorptive enterocytes, mucus-secreting goblet cells, antimicrobial Paneth cells, hormone-producing enteroendocrine cells, sensory tuft cells, and antigen-sampling M cells, the epithelium coordinates digestion, secretion, and immune surveillance. Structurally, the intestinal epithelium features tight junctions, adherens junctions, and desmosomes that form an apical junctional complex, with tight junctions—comprising proteins like claudins, , and tricellulin—acting as the primary regulators of paracellular permeability to maintain . These junctions create size- and charge-selective pathways: high-capacity "" pathways for small ions and solutes, and low-capacity "leak" pathways that restrict larger molecules and microbes, supported by a protective layer secreted by goblet cells that further shields the from direct bacterial contact. In the , villi and microvilli dramatically expand the surface area by up to 600-fold, enhancing absorption efficiency, while the colonic epithelium focuses more on and fermentation byproduct handling. Functionally, the not only absorbs essential nutrients via transporters but also secretes from Paneth cells and hormones from enteroendocrine cells to modulate gut , , and . It interacts bidirectionally with the , with tuft cells sensing microbial signals to initiate type 2 immune responses against parasites and goblet cells adjusting production in response to bacterial density, thereby preserving . Lgr5-positive crypt base columnar stem cells, sustained by niche signals like Wnt and pathways, ensure continuous regeneration, with remarkable allowing during injury to restore barrier integrity. Dysfunction in the intestinal epithelium, often involving increased permeability due to altered tight junction expression (e.g., elevated claudin-2 in inflammatory conditions), is implicated in diseases such as and celiac disease, highlighting its role as a therapeutic target for barrier-modulating agents.

Anatomy and Structure

Cell types

The intestinal epithelium comprises a diverse array of specialized types that arise from multipotent intestinal cells (ISCs), primarily Lgr5-positive cells located at the base of crypts in both the small and large intestines. These stem cells generate transit-amplifying progenitors that differentiate into the major epithelial lineages along the crypt-villus axis in the or crypt-surface axis in the colon, ensuring functional specialization for , , and barrier maintenance. Enterocytes, also known as absorptive columnar cells, form the predominant population and are characterized by their tall, columnar with an apical of densely packed microvilli that dramatically increase surface area for nutrient uptake. These cells are located primarily on the villi of the and the surface of the , originating from ISCs through high signaling that promotes an absorptive fate while repressing secretory differentiation. In contrast, goblet cells exhibit a distinctive goblet-shaped filled with secretory granules containing , enabling production; they are scattered throughout the in both crypts and villi, derived from Atoh1-expressing secretory progenitors regulated by low and signaling. Paneth cells, residing exclusively at the base of small intestinal crypts, possess a pyramidal shape with prominent secretory granules that store like ; they differentiate from ISCs via Wnt/β-catenin and signaling, providing niche support to neighboring stem cells. Enteroendocrine cells are rare, scattered hormone-secreting cells with a small, basally located and apical processes; they emerge from Neurog3-positive progenitors in the secretory lineage and are found along both crypts and villi in the small and large intestines. Tuft cells, named for their tuft-like bundles of apical microvilli, adopt a chemosensory morphology and derive from Atoh1- and Pou2f3-dependent secretory progenitors, appearing sporadically throughout the in both intestinal regions. M cells, specialized for sampling, display an irregular apical surface with microfolds rather than microvilli and are located within the follicle-associated overlying Peyer's patches, primarily in the ; their differentiation involves signaling, potentially from precursors or direct ISC lineage commitment. ISCs themselves are small, undifferentiated cells marked by expression, positioned at the base interspersed with Paneth cells in the , and they self-renew while producing all other epithelial lineages through asymmetric division and progenitor expansion. Proportions of these cell types vary regionally: in the , enterocytes constitute approximately 80-90% of the , goblet cells 5-10%, Paneth cells ~5%, and enteroendocrine and tuft cells each <1%, with M cells being very rare and confined to lymphoid areas. In the , enterocytes (or colonocytes) remain dominant but goblet cells increase to up to 50% to bolster the layer, while Paneth cells are absent and tuft cells persist at low levels.

Epithelial organization and renewal

The intestinal epithelium exhibits a highly organized along the crypt-villus axis in the , where and Paneth cells reside at the base of invaginated , transit-amplifying cells occupy the mid- region, and differentiated epithelial cells migrate upward to the villus tips before extrusion. This hierarchical arrangement ensures efficient renewal, with Paneth cells providing essential niche signals to support maintenance at the base. In the colon, are present but lack villi, resulting in a flat surface where cells migrate from bases to the luminal surface. Epithelial renewal is remarkably rapid, with the entire intestinal lining turning over every 3-5 days in humans, driven by continuous in the crypts and subsequent migration of daughter cells. Newly generated cells from the transit-amplifying compartment migrate upward at rates that sustain this turnover, reaching the villus tip or colonic surface for shedding, thereby maintaining epithelial integrity and function. This dynamic process replaces approximately 10^11 cells daily, adapting to physiological demands while minimizing disruption to barrier properties. At the core of this renewal is a hierarchy comprising active + crypt base columnar cells, which drive daily through rapid cycling and generation of progenitors, and quiescent +4 reserve cells positioned approximately four cells above the crypt base, which serve as a population activated during . + cells differentiate into transit-amplifying progenitors that amplify the output of differentiated lineages, while +4 cells, marked by genes like Bmi1, exhibit slower proliferation but contribute to long-term repopulation. Key signaling pathways orchestrate this hierarchy: Wnt signaling promotes proliferation and Paneth cell differentiation at the crypt base, while signaling regulates the balance between proliferation in transit-amplifying cells and differentiation into secretory or absorptive lineages as cells ascend. These pathways interact dynamically, with Wnt maintaining the niche and suppressing secretory fates to favor absorptive differentiation higher along the axis. Regional differences persist, as colonic crypts lack Paneth cells in the same density but retain Lgr5+ s and similar Wnt/-driven renewal, albeit with adjusted turnover kinetics suited to the absence of villi.

Cellular junctions

The intestinal epithelium relies on specialized cellular junctions to maintain structural , , and selective intercellular communication among its diverse types, such as enterocytes and goblet cells. These junctions form a belt-like network at the apical-lateral domains, ensuring mechanical cohesion and barrier formation while allowing coordinated signaling. In the dynamic of the gut, where epithelial cells undergo turnover, these structures are essential for preventing paracellular leakage and supporting . Tight junctions (TJs) represent the most apical junctional complex, forming a selective seal that regulates paracellular permeability. Composed primarily of transmembrane proteins including claudins and occludins, TJs create strand-like networks that intertwine between adjacent cells. Claudins, a family of over 24 members, form the backbone of these strands, with specific isoforms like claudin-1 and claudin-5 promoting barrier tightness, while claudin-2 facilitates cation-selective channels. Occludins, characterized by four transmembrane domains, associate with claudins to modulate strand stability and contribute to the seal against macromolecules. Cytoplasmic plaque proteins such as zonula occludens (ZO)-1, ZO-2, and ZO-3 anchor these transmembrane components to the actin cytoskeleton, enabling force transmission and junctional maturation. Adherens junctions (AJs) lie immediately basal to TJs and provide adhesive strength through calcium-dependent interactions. Central to AJs is the E-cadherin-catenin complex, where E-cadherin homodimers from neighboring cells engage extracellularly, while their cytoplasmic tails bind β-catenin and p120-catenin intracellularly. α-Catenin further links this complex to the actin cytoskeleton, facilitating actomyosin contractility and epithelial sheet integrity. This linkage is crucial for maintaining cell-cell adhesion in the mechanically stressed intestinal lining. Desmosomes, positioned more basally than , serve as robust anchors conferring mechanical resilience to the . These junctions feature desmosomal cadherins—desmogleins (e.g., desmoglein-2) and desmocollins (e.g., desmocollin-2)—which form heterophilic or homophilic bonds across the intercellular space. Intracellularly, these cadherins connect via proteins like plakoglobin and plakophilins to desmoplakin, which bundles filaments such as keratins 8 and 18. This architecture distributes tensile forces across the epithelial layer, essential for withstanding peristaltic shear in the intestine. Gap junctions enable direct cytoplasmic continuity for intercellular exchange, composed of connexin proteins that assemble into hexameric hemichannels (connexons). In the intestinal epithelium, predominant connexins include Cx32 and Cx43, which dock between cells to form aqueous pores permeable to ions, metabolites, and signaling molecules up to ~1 kDa, such as and ATP. These channels support synchronized cellular responses, including propagation of calcium waves among enterocytes. The assembly and regulation of these junctions involve coordinated molecular scaffolding and post-translational modifications. Zonula occludens proteins orchestrate TJ maturation by recruiting claudins and occludins to the apical membrane, often following AJ formation as a prerequisite. Phosphorylation by kinases such as (PKC) and tyrosine kinases modulates junctional dynamics; for instance, PKC enhances occludin localization to TJs, while promotes and barrier disruption. In and desmosomes, catenin and plakoglobin by Src kinases influences adhesion strength and cytoskeletal linkage. trafficking to junctions is similarly regulated by , with sites on Cx43 targeted by MAPK pathways to control gating and stability. These mechanisms ensure adaptive responses to environmental cues while preserving epithelial architecture.

Physiology

Absorption and secretion

The intestinal epithelium plays a central role in and homeostasis through coordinated and processes. primarily occurs via transcellular and paracellular pathways across enterocytes, the predominant in the villi, while is driven by specialized crypt cells, including for . These mechanisms ensure efficient uptake of dietary components and maintenance of luminal hydration, with energy provided by ion gradients. Transcellular absorption involves active transport across the apical and basolateral membranes of enterocytes. For carbohydrates, the sodium-glucose linked transporter 1 (SGLT1) facilitates and uptake in a sodium-dependent manner, with a stoichiometry of two sodium ions per , primarily in the . absorption relies on the proton-coupled transporter PEPT1, which handles di- and tripeptides generated from , contributing to nitrogen balance. absorption occurs through diffusion of free fatty acids and monoglycerides into enterocytes, followed by re-esterification into triglycerides and packaging into chylomicrons for secretion via into lacteals. Paracellular absorption supplements transcellular routes by allowing passive movement of ions, , and small solutes through tight junctions between epithelial cells. This pathway, often driven by solvent drag from osmotic flow generated by active solute , facilitates the uptake of ions like calcium in the and , modulated by junctional proteins such as claudins. in the intestinal epithelium maintains luminal conditions and supports . In crypt cells, the (CFTR) channel mediates into the , creating an osmotic that drives for hydration. Goblet cells, interspersed among enterocytes, secrete mucins to form a protective layer that aids in and nutrient passage. Paneth cells in the crypt base release , such as α-defensins, into the to regulate the local environment. Regional specialization enhances efficiency along the small intestine. The duodenum primarily absorbs iron via divalent metal transporter 1 (DMT1) and recycles bile salts through apical sodium-dependent bile acid transporter (ASBT). The ileum specializes in vitamin B12 uptake via the cubam receptor complex. pH gradients, lower in the duodenum and increasing distally, influence solubility and transport of ions like calcium. These processes are energetically dependent on the basolateral Na+/K+ ATPase pump, which hydrolyzes ATP to extrude three sodium ions and import two potassium ions per cycle, establishing the electrochemical gradient essential for secondary active transport via SGLT1 and other carriers.

Barrier function and permeability

The intestinal epithelium functions as a selective physical and biochemical barrier, allowing the controlled passage of essential molecules such as nutrients, ions, and water while restricting the entry of pathogens, toxins, and antigens from the luminal environment. This barrier selectivity is mediated by two main routes: transcellular transport across the epithelial cells and paracellular transport between adjacent cells. Disruption of this balance can lead to increased permeability, or "leakiness," potentially compromising host defense without affecting absorptive functions. Transcellular permeability occurs through the epithelial cell interior via vesicular transport mechanisms, including fluid-phase , , and , which enable the uptake and potential transfer of macromolecules across the cell. These processes are dynamically regulated by and cycles, where internalized vesicles are often routed to lysosomes for degradation to prevent translocation, ensuring the pathway remains selective and non-permissive to harmful agents. For example, in the neonatal intestine, facilitates transport but is downregulated in adults to enhance barrier tightness. Paracellular permeability is primarily controlled by tight junctions, which form pore pathways for small ions and uncharged solutes (up to ~4 ) and selective pathways for larger or charged molecules based on size and charge discrimination. Tight junction proteins such as claudins regulate these pathways, with the overall barrier integrity quantified by transepithelial electrical resistance (TER), which typically ranges from 100 to 200 Ω·cm² in the intestinal epithelium, reflecting primarily paracellular resistance. The tight junction structures enabling this permeability are discussed in the section on cellular junctions. Maintenance of barrier integrity relies on signaling molecules like interleukin-22 (), which sustains by inducing (AMPs) such as Reg3γ and β-defensins in epithelial cells, thereby limiting microbial invasion. also modulates in infected cells, promoting survival and regeneration of healthy epithelium via activation while facilitating the targeted elimination of compromised cells to avert dissemination. Secretory contributions to the barrier, such as production, aid in physical separation of luminal contents. Permeability is influenced by external factors, including diet and ; for instance, high-fructose diets can reduce proteins like and claudin-1, increasing leakiness, particularly under conditions that trigger release. , a precursor to haptoglobin-2, reversibly disassembles s in response to psychological or physiological via signaling, thereby elevating paracellular flux and potentially allowing translocation. Measurement of barrier permeability commonly employs Ussing chambers, an system where intestinal tissue is mounted between reservoirs to assess flux of inert probes (e.g., FITC-dextran for paracellular or lucifer yellow for transcellular routes) across the under physiological conditions, simultaneously monitoring TER to evaluate integrity. This technique provides quantitative insights into both pathway-specific permeability and overall in health and experimental perturbations.

Interactions with immune system and microbiota

The intestinal epithelium serves as a critical interface for immune surveillance, employing pattern recognition receptors (PRRs) to detect microbial and danger signals from the lumen. Toll-like receptors (TLRs), such as TLR4 and TLR5, are expressed on the apical surface of epithelial cells and recognize pathogen-associated molecular patterns (PAMPs) like lipopolysaccharide (LPS) from Gram-negative bacteria and flagellin from motile microbes, respectively, initiating downstream signaling via NF-κB to promote antimicrobial responses. In the cytoplasm, NOD-like receptors (NLRs), including NOD1 and NOD2, detect peptidoglycan fragments from bacterial cell walls, activating pathways that enhance autophagy and inflammasome assembly to maintain barrier integrity. Epithelial cells modulate immune responses by secreting cytokines that orchestrate leukocyte recruitment and production. For instance, upon PAMP stimulation, intestinal epithelial cells (IECs) produce interleukin-8 (IL-8), a potent that recruits neutrophils to the , facilitating rapid clearance. Additionally, IECs interact with underlying plasma cells to support secretory IgA (sIgA) production; through the release of factors like and BAFF, IECs promote B-cell differentiation and IgA class switching, enabling non-inflammatory neutralization of luminal antigens. The epithelium senses microbiota through specialized cells that bridge luminal contents and immune effectors. Tuft cells, a rare IEC subset, detect succinate from protozoan parasites via the succinate receptor SUCNR1 (GPR91), triggering IL-25 secretion that activates group 2 (ILC2s) to produce IL-13, amplifying mucus production and type 2 immunity. Microfold (M) cells in Peyer's patches sample particulate antigens and microbes from the lumen, transcytosing them to underlying dendritic cells for and T-cell priming, thus initiating adaptive responses without breaching the barrier. Feedback mechanisms reinforce epithelial-immune-microbiota harmony, with microbiota-derived (SCFAs) like butyrate binding to G-protein-coupled receptor 43 (GPR43) on IECs to upregulate proteins and , thereby strengthening barrier function. Recent 2025 studies highlight IEC involvement in microbiota via autoinducer-2 (AI-2), a signaling that modulates bacterial colonization and IEC responses; for example, AI-2 from commensals promotes epithelial repair and reduces inflammation by influencing microbial community dynamics and host signaling.

Functions in Homeostasis

Nutrient processing and transport

The intestinal epithelium plays a central role in integrating luminal with , primarily through the action of brush-border enzymes on the apical surface of enterocytes. These enzymes, such as lactase-phlorizin hydrolase and sucrase-isomaltase, catalyze the of disaccharides like and into absorbable monosaccharides, including glucose and , facilitating the final stages of breakdown that complement pancreatic secretions. Similarly, in processing, salts emulsify dietary fats in the proximal , enabling formation for efficient uptake, while the ileal actively reabsorbs over 95% of these salts via apical sodium-dependent transporters, supporting their recirculation to the liver through the and maintaining the enterohepatic cycle essential for ongoing fat . Once internalized, nutrients are processed within enterocytes and transported systemically to support metabolic . Glucose and other monosaccharides exit the basolateral primarily via facilitative transporters like GLUT2, which equilibrates intracellular sugars with the bloodstream, allowing rapid delivery to peripheral tissues. Lipids, re-esterified into triglycerides within the , are packaged into chylomicrons and secreted apically into lacteals, entering the before joining the venous circulation via the , thereby bypassing initial hepatic processing. Homeostatic regulation of nutrient processing involves enteroendocrine cells within the epithelium that sense luminal contents and release hormones to coordinate digestive responses. For instance, cholecystokinin (CCK) secreted by I-cells in response to fats and proteins stimulates gallbladder contraction and pancreatic enzyme release, enhancing luminal digestion while feedback mechanisms prevent overload. The epithelium adapts dynamically to nutritional states; postprandial hyperemia increases splanchnic blood flow by 30-50% to support heightened absorption rates, whereas fasting reduces epithelial cell turnover by up to 50%, conserving energy and minimizing unnecessary renewal. Quantitatively, the human intestinal epithelium absorbs approximately 9 liters of water and 100 grams of protein daily, underscoring its efficiency in maintaining fluid and nitrogen balance for systemic needs.

Defense and immune modulation

The intestinal epithelium serves as a frontline barrier against pathogens through the secretion of () primarily from Paneth cells, which reside at the base of small intestinal crypts. These cells produce α-defensins such as human defensin 5 (HD5) and HD6, which disrupt bacterial membranes and limit microbial proliferation in the crypt lumen. Paneth cells also secrete , an enzyme that hydrolyzes in bacterial cell walls, further contributing to innate antimicrobial defense. Complementing these mechanisms, goblet cells in the epithelium produce a layer rich in mucins, which physically traps pathogens and prevents their adhesion to epithelial surfaces, thereby facilitating clearance by and immune cells. Epithelial cells actively prime adaptive immune responses by secreting cytokines that modulate (DC) function and T cell differentiation. (TSLP) and interleukin-33 (IL-33), released from intestinal epithelial cells (IECs) in response to stress or microbial signals, condition DCs to promote type 2 immune responses and enhance to T cells. Additionally, IECs produce IL-23, which drives the differentiation and maintenance of Th17 cells, thereby supporting IL-17-mediated defense against extracellular bacteria and fungi at mucosal sites. Upon injury, the intestinal epithelium undergoes rapid through collective , where neighboring epithelial cells coordinate movement to reseal breaches without in the initial phase. This restitution process is accelerated by (EGF) signaling, which activates receptor tyrosine kinases on IECs to promote lamellipodia formation and directed migration. In response to viral and bacterial threats, IECs mount targeted defenses via cytokine production and inflammasome pathways. Type III interferons (IFN-λ), secreted by IECs upon viral recognition, induce an antiviral state in neighboring epithelial cells by upregulating interferon-stimulated genes, effectively restricting viral spread while minimizing inflammation compared to type I interferons. For bacterial pathogens, activation of inflammasomes such as NAIP/NLRC4 in IECs leads to caspase-1-mediated processing of IL-1β and IL-18, promoting and expulsion of infected cells to limit bacterial invasion. Aging impairs these defensive functions, with reduced production of like HD5 in the elderly intestinal epithelium, leading to diminished activity and heightened susceptibility to infections. Recent studies indicate that this decline correlates with altered composition and increased , exacerbating vulnerability in older individuals.

Regulation of microbial symbiosis

The intestinal epithelium plays a pivotal role in regulating microbial by creating specialized niches that support beneficial while maintaining host-microbial balance. One key mechanism involves the provision of -derived glycans, which serve as a primary nutrient source for such as . This mucin-degrading bacterium colonizes the layer, utilizing O-linked glycans through enzymes like sialidases and fucosidases to access carbon and nitrogen, thereby reinforcing the epithelial barrier and preventing excessive degradation that could lead to . Additionally, radial oxygen gradients established by the epithelium favor the growth of symbionts; oxygen diffuses from host tissues into the but is rapidly consumed by facultative anaerobes near the epithelium, creating a low-oxygen environment deeper in the that sustains obligate anaerobes essential for mutualistic interactions. Bidirectional signaling between the and further stabilizes , with microbial metabolites like butyrate—produced by fermentative —enhancing epithelial integrity. Butyrate acts as a (HDAC) inhibitor, promoting histone acetylation that upregulates proteins such as synaptopodin and claudins, thereby strengthening the barrier and reducing permeability to prevent microbial overgrowth. This metabolite also modulates epithelial to foster a symbiotic , illustrating how microbiota-derived signals reciprocally influence epithelial function. To prevent dysbiosis, the employs regulatory circuits involving specialized cells. Tuft cells, sensing microbial or parasitic cues via metabolites like succinate, activate an ILC2-dependent type 2 that limits helminth overgrowth and maintains microbial by promoting epithelial remodeling without excessive . Complementing this, secretory IgA (sIgA) produced by the coats , promoting their retention in the gut and metabolic fitness while inhibiting pathogenic invasion, thus ensuring a stable microbial community. Developmentally, the shapes epithelial maturation post-weaning, when microbial accelerates the transition from immature to adult-like . Neonatal primes differentiation and production, enhancing barrier maturation and adapting the to solid food , with disruptions leading to delayed . Recent advances as of 2025 highlight the role of extracellular vesicles (EVs) in IEC-microbiota communication, where bacterial EVs carry DNA for to other microbes and host cells, facilitating genetic material exchange that modulates symbiotic adaptation and epithelial responses.

Clinical Significance

Role in intestinal diseases

The intestinal epithelium plays a central role in the of (IBD), a group of chronic disorders including (CD) and (UC), where epithelial barrier dysfunction allows luminal antigens to penetrate the mucosa, perpetuating inflammation. In CD, inflammation is transmural, affecting all layers of the intestinal wall and often leading to fistulas and strictures, whereas UC is confined to the mucosal layer, primarily involving the colon with continuous inflammation. This barrier impairment precedes clinical symptoms and is driven by the IL-23/Th17 axis, where IL-23 promotes Th17 cell differentiation and production of pro-inflammatory cytokines like IL-17 and , which disrupt tight junctions via upregulation of claudin-2 and , increasing paracellular permeability. Epidemiological data indicate a rising of IBD in Western populations, with rates of approximately 0.7–0.8% in and higher in parts of (e.g., 0.83% in and 1.19% for Denmark as of 2025), reflecting ongoing increases despite stabilizing incidence in adults. In celiac disease, an autoimmune enteropathy triggered by gluten ingestion in genetically susceptible individuals, the intestinal epithelium exhibits heightened permeability due to gliadin-induced release of zonulin, a tight junction modulator that disassembles occludin and ZO-1 proteins, facilitating antigen translocation to the lamina propria and immune activation. This zonulin release is mediated by gliadin binding to CXCR3 receptors on enterocytes, activating a MyD88-dependent pathway that elevates intestinal permeability within hours of exposure. Concurrently, gliadin promotes intraepithelial lymphocytosis, characterized by an influx of CD8+ T cells and γδ T lymphocytes into the epithelium, contributing to villous atrophy and crypt hyperplasia. Infectious agents further highlight the epithelium's vulnerability, as seen in Clostridioides difficile infections where toxins A and B inactivate Rho (Rho, Rac, Cdc42), leading to F-actin and redistribution of proteins like and ZO-1 from membrane microdomains, thereby increasing paracellular permeability and enabling bacterial persistence. Similarly, , a leading cause of , replicates efficiently in human intestinal enteroids derived from cells, targeting differentiated enterocytes while relying on progenitor-derived monolayers for propagation, which disrupts epithelial integrity and impairs during acute . Beyond inflammatory and infectious conditions, epithelial alterations underpin other disorders, such as , where APC gene mutations in intestinal stem cells initiate tumorigenesis by destabilizing the β-catenin destruction complex, promoting Wnt signaling hyperactivation and clonal expansion of mutant crypts into adenomas. In following extensive resection, the remnant epithelium undergoes adaptive with increased villus height, crypt depth, and , enhancing absorptive surface area through upregulated transporters like SGLT1, though this compensation often remains insufficient for full nutritional autonomy.

Advances in research and therapy

Recent advances in intestinal epithelium research have leveraged three-dimensional (3D) models derived from intestinal stem cells to recapitulate the crypt-villus and enable high-fidelity simulations of epithelial . These patient-derived s maintain genomic and phenotypic to the donor tissue, facilitating personalized drug screening for (IBD) therapies by assessing epithelial responses to candidate compounds in a controlled . For instance, s have been used to evaluate the efficacy of novel agents in restoring barrier integrity during IBD flares, accelerating the identification of therapeutics that target epithelial dysfunction. Single-cell RNA sequencing (scRNA-seq) has unveiled significant heterogeneity in intestinal states, revealing dynamic transcriptional programs that govern epithelial renewal and . Studies have identified distinct subpopulations with varying proliferative capacities, influenced by niche signals, which contribute to tissue homeostasis under . In 2025, transcriptomic analyses demonstrated aging-related shifts in intestinal epithelial cells, including upregulated senescence-associated pathways and altered barrier gene expression in the colon, linking age-dependent decline to reduced regenerative potential. Therapeutic strategies targeting epithelial regeneration have progressed, with mesenchymal stem cell (MSC) transplants showing promise in preclinical and early clinical settings for repairing radiation- or chemotherapy-induced epithelial damage in IBD models. These cells promote epithelial proliferation and junctional integrity by secreting anti-inflammatory factors, with phase I/II trials reporting improved mucosal healing rates. Anti-TNF biologics, such as and , restore proteins like claudins and occludins in the intestinal epithelium, enhancing barrier function and reducing permeability in active patients. Fecal microbiota transplantation (FMT) modulates epithelial- symbiosis by enriching beneficial taxa that upregulate production and , thereby supporting epithelial and reducing inflammation in . Emerging technologies include CRISPR-Cas9 editing of intestinal epithelial cells (IECs) within organoids, enabling precise modeling of genetic variants associated with epithelial disorders and high-throughput screens for regulators of fate. This approach has identified key genes influencing epithelial maturation, with editing efficiencies exceeding 80% in human organoids. Additionally, AI-driven models predict by integrating molecular descriptors and epithelial transcriptomic data, aiding in the design of drugs that preserve barrier function without empirical testing. Despite these innovations, challenges persist in translating organoid-derived insights to applications, including discrepancies in vascularization, immune interactions, and long-term stability that limit direct comparability to native . Ethical concerns with human-derived organoids encompass for tissue donation, potential for unintended in complex models, and equitable access to personalized therapies derived from patient-specific lines.

References

  1. [1]
    The Intestinal Epithelium – Fluid Fate and Rigid Structure From ...
    The single-layered, simple epithelium of the gastro-intestinal tract controls nutrient uptake, coordinates our metabolism and shields us from pathogens.
  2. [2]
    The intestinal epithelial barrier: A therapeutic target? - PMC
    Jan 1, 2018 · A fundamental function of the intestinal epithelium is to act as a barrier that regulates interactions between the luminal contents.
  3. [3]
    Intestinal development and differentiation - PMC - PubMed Central
    In this review, we present an overview of intestinal development and cellular differentiation of the intestinal epithelium.
  4. [4]
    Identification of feature genes in intestinal epithelial cell types - NIH
    Nov 15, 2024 · Here, we unveiled the feature genes in different cell types of the human and mouse gut through single-cell RNA sequencing analysis.
  5. [5]
    Transcriptional programmes underlying cellular identity and ...
    Although the intestinal epithelium consists of a continuous single-cell layer, its diverse functions require the establishment of appropriate cell types, tissue ...Intestinal Cell Identities · Epigenetics In Identity And... · Histone Modifications
  6. [6]
    Intestinal Stem Cells - PMC - NIH
    Self-renewal in the intestinal epithelia is fueled by a population of undifferentiated intestinal stem cells (ISCs) that give rise to daughter or progenitor ...
  7. [7]
    The Intestinal Stem Cell Niche: Homeostasis and Adaptations - PMC
    The homeostatic constant regeneration of the intestinal epithelium is driven by active Lgr5+ stem cells (active intestinal stem cells [ISCs]) at the crypt bases ...Canonical Niche Pathways · Wnt Ligands · Emerging Niche Pathways
  8. [8]
    Intestinal stem cells and epithelial-mesenchymal interactions in the ...
    Stem cells, located in the crypt base, give rise to transit amplifying cells that differentiate into the four major epithelial cell types: enterocytes, goblet ...Figure 1 · Stem Cell Niche: The Tissue... · Stem Cell Therapy For...
  9. [9]
    Control of Intestinal Epithelial Proliferation and Differentiation - NIH
    ... rate in the adult human body, with complete turnover normally occurring every 3–5 days. The proliferating compartment is normally restricted to the crypt ...
  10. [10]
    Protein Turnover in Epithelial Cells and Mucus along the ... - NIH
    Jan 28, 2020 · The enterocytes, enteroendocrine, and goblet cells differentiate and migrate over 3–5 days from the crypt to the villus tip, where they ...
  11. [11]
    Intestinal epithelial regeneration: active versus reserve stem cells ...
    The intestinal epithelium has one of the highest turnover rates, with 10 11 epithelial cells (~200 g) being lost every day in humans.
  12. [12]
    The intestinal stem cell markers Bmi1 and Lgr5 identify two ... - NIH
    Bmi1 marks quiescent, injury-inducible reserve ISCs that exhibit striking functional distinctions from Lgr5 + ISCs.
  13. [13]
    Notch signaling modulates proliferation and differentiation of ...
    Notch signaling is known to regulate the proliferation and differentiation of intestinal stem and progenitor cells; however, direct cellular targets and ...
  14. [14]
    Opposing activities of Notch and Wnt signaling regulate intestinal ...
    Apr 7, 2015 · Notch blockade perturbs intestinal stem cell function by causing a derepression of the Wnt signaling pathway, leading to misexpression of prosecretory genes.
  15. [15]
    Cell-cell junctions: structure and regulation in physiology and ...
    The current review article presents our current state of knowledge on various cell-cell junctions, their molecular composition, and mechanisms regulating their ...2. Epithelial And... · 3. Tight Junctions (tjs) And... · 4. Cldn Family Of Tj...
  16. [16]
    The Organization of Tight Junctions in Epithelia - PubMed Central
    In epithelial tissue, cell–cell interactions are mediated by junctional complexes that consist of tight junctions (TJs), adherens junctions (AJs), desmosomes, ...Integral Membrane Proteins · Claudins · Cytoplasmic ProteinsMissing: paper | Show results with:paper
  17. [17]
    Full article: Epithelial integrity, junctional complexes, and biomarkers ...
    This review aims to describe the molecular structure of intestinal epithelial tight junction proteins and to characterize their organization and interaction.Occludin (ocln) · Claudins (cldns) · Zonula Occludens (zos)
  18. [18]
    Adherens junction proteins on the move—From the membrane to the ...
    In this review we discuss cell-cell adhesion complexes in the intestinal epithelial layer with a focus on the nuclear functions of adherens junction ...
  19. [19]
    Targeting desmosomal adhesion and signalling for intestinal barrier ...
    May 18, 2020 · The aim of this review is to summarize recent findings on the role of desmosomes in intestinal epithelial barrier regulation, inflammation and ...OVERVIEW OF THE... · THE INTEGRITY OF... · MECHANISMS UNDERLYING...
  20. [20]
    Desmosomes: Essential contributors to an integrated intercellular ...
    Dec 30, 2019 · Four types of intercellular junction are present in vertebrates: desmosomes, adherens junctions, tight junctions, and gap junctions.
  21. [21]
    Connexin and pannexin signaling in gastrointestinal and liver disease
    Gap junctions, which mediate intercellular communication, are key players in digestive homeostasis. They are also frequently involved in gastrointestinal ...
  22. [22]
    The Role of Connexins in Gastrointestinal Diseases - ScienceDirect
    Gap junctions connect cells and allow passage of molecules, ion, and electrical impulses between them.
  23. [23]
    Interplay between tight junctions & adherens junctions - ScienceDirect
    Sep 1, 2017 · Ultrastructural studies reveal that cells are connected by multiple junctional complexes, including the tight junctions, adherens junctions, gap ...Adherens Junctions · Tight Junctions · References (65)Missing: paper | Show results with:paper
  24. [24]
    The tight junction: a multifunctional complex
    The aim of this review is to present an updated overview of recent developments in the area of tight junction biology. In a relatively short time, our knowledge ...
  25. [25]
    Physiology of Intestinal Absorption and Secretion - PMC - NIH
    Virtually all nutrients from the diet are absorbed into blood across the highly polarized epithelial cell layer forming the small and large intestinal mucosa.
  26. [26]
    Sodium-glucose cotransport - PMC - PubMed Central - NIH
    Purpose of review​​ SGLT1 mediates almost all sodium-dependent glucose uptake in the small intestine, while in the kidney SGLT2, and to a lesser extent SGLT1, ...
  27. [27]
    Regulation profile of the intestinal peptide transporter 1 (PepT1) - PMC
    Dec 8, 2017 · The intestinal peptide transporter 1 (PepT1) was first identified in 1994. It plays a crucial role in the absorption of small peptides.
  28. [28]
    Intestinal Lipid Absorption and Lipoprotein Formation - PMC - NIH
    The data reviewed above suggest that chylomicron is the only pathways for triglyceride absorption. However, there are two pathways for the absorption of ...
  29. [29]
    Contribution of solvent drag through intercellular junctions ... - PubMed
    The results indicate that solvent drag through paracellular channels is the principal route for intestinal transport of glucose or amino acids.
  30. [30]
    Histology, Goblet Cells - StatPearls - NCBI Bookshelf
    Mar 15, 2023 · The primary function of goblet cells is to secrete mucin and create a protective mucus layer. Goblet cells are also thought to be involved with ...Introduction · Structure · Function · Tissue Preparation
  31. [31]
    IV. Paneth cell antimicrobial peptides and the biology of the mucosal ...
    In the mammalian small intestine, Paneth cells secrete granules that are rich in α-defensins and additional antimicrobial peptides into the lumen of the crypt.<|separator|>
  32. [32]
    Physiology, Sodium Potassium Pump - StatPearls - NCBI Bookshelf
    Mar 13, 2023 · The Na+K+ pump is an electrogenic ATPase that pumps 3 Na+ out and 2 K+ in, maintaining osmotic equilibrium and membrane potential.
  33. [33]
    Intestinal Barrier Function: Molecular Regulation and Disease ...
    The intestinal epithelium mediates selective permeability via two major routes: transepithelial/transcellular and paracellular pathways (Figure 1).
  34. [34]
    Probing paracellular -versus transcellular tissue barrier permeability ...
    Apr 18, 2019 · As an alternative mechanism to membrane perturbation, transcellular permeation may be achieved by transcytosis from the gut lumen to the ...
  35. [35]
    The Intestinal Barrier and Current Techniques for the Assessment of ...
    A crucial function of the intestinal epithelium is the maintenance of a proper barrier function, allowing the permeability of nutrients, water and ions, but ...
  36. [36]
    The role of IL-22 in intestinal health and disease - PMC - NIH
    Feb 13, 2020 · IL-22 regulates epithelial homeostasis, cell growth, mucus, and antimicrobial proteins. It can promote barrier repair, but also inflammation ...
  37. [37]
    Role of Stress on Driving the Intestinal Paracellular Permeability
    Nov 18, 2023 · The hallmark function of the gut barrier entails the selective transport of luminal substances toward blood circulation, and it is determined by ...<|control11|><|separator|>
  38. [38]
    The Ussing chamber system for measuring intestinal permeability in ...
    Jun 20, 2019 · An Ussing chamber was constructed and adapted to support both mouse and human tissue to measure intestinal permeability, using paracellular flux and electrical ...
  39. [39]
    NOD-like receptors mediate homeostatic intestinal epithelial barrier ...
    Sep 9, 2023 · It is believed that NOD-like receptors (NLRs) are the main mediators in maintaining intestinal mucosal barrier homeostasis, including regulation ...Nlrs Family And Signaling... · Nod-Like Receptors Regulate... · Role Of Nlr In Regulating...
  40. [40]
    Review Neutrophil–Epithelial Crosstalk During Intestinal Inflammation
    It is widely accepted that IL-8, a powerful chemoattractant released by IECs, effectively attracts neutrophils to the basolateral surface of the epithelium.
  41. [41]
    The intestine: A highly dynamic microenvironment for IgA plasma cells
    To increase the surface for food absorption, the small intestine is characterized by the presence of villi, whereas villi cannot be found in the caecum and the ...
  42. [42]
    Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 ...
    Our results identify intestinal tuft cells as critical sentinels in the gut epithelium that promote type 2 immunity in response to intestinal parasites.
  43. [43]
    M Cells: Intelligent Engineering of Mucosal Immune Surveillance
    M cells are specialized intestinal epithelial cells that provide the main machinery for sampling luminal microbes for mucosal immune surveillance.
  44. [44]
    GPR43 mediates microbiota metabolite SCFA regulation of ...
    Feb 7, 2018 · Our studies thus demonstrated that microbiota metabolites SCFA promoted IEC RegIIIγ and β-defensins in a GPR43-dependent manner.
  45. [45]
    Mechanisms and implications of the gut microbial modulation of ...
    Jun 10, 2025 · Furthermore, quorum-sensing molecules, including autoinducer-2 (AI-2), enable interspecies communication, orchestrating bacterial ...<|control11|><|separator|>
  46. [46]
    Intestinal Absorption of Bile Acids in Health and Disease - PMC
    The decrease in intestinal absorption reduces the circulating pool of bile acids, disrupting bile acid homeostasis and altering bile acid signaling processes.
  47. [47]
    Mechanisms of chylomicron uptake into lacteals - PMC
    Nearly all dietary lipid is transported in chylomicrons from the gut to the blood through the lymphatic system by entering specialized lymphatic vessels, ...
  48. [48]
    Regulation of Whole-Organ Pancreatic Secretion - NCBI - NIH
    CCK is secreted from the basolateral surface of a specific endocrine cell in the mucosa of the upper small intestine called the I cell [136].
  49. [49]
    Postprandial Hyperemia - The Gastrointestinal Circulation - NCBI - NIH
    It is well established that gastrointestinal blood flow increases after meals, a phenomenon referred to as postprandial or functional hyperemia.
  50. [50]
    Pre-therapy fasting slows epithelial turnover and modulates the ...
    Aug 26, 2020 · Acute fasting slows epithelial turnover and induces significant changes in body composition. Rats were given ad libitum access to food and water ...
  51. [51]
    Paneth cell α-defensins and enteric microbiota in health and disease
    Paneth cells also secrete other antimicrobial peptides, such as Reg3γ, and antimicrobial proteins including lysozyme and secretory phospholipase A2 [40, 41].Paneth Cells, A Lineage Of... · Paneth Cell Disruption... · Fig. 3
  52. [52]
    Reduced Paneth cell α-defensins in ileal Crohn's disease - PNAS
    Paneth cells (PC) are the major source of antimicrobial peptides in the small intestine, including human α-defensins HD5 and HD6. We tested the hypothesis ...
  53. [53]
    Paneth cells secrete lysozyme via secretory autophagy ... - Science
    Jul 27, 2017 · Paneth cells are specialized intestinal epithelial cells that secrete abundant antimicrobial proteins, including lysozyme; thus, disrupting ...
  54. [54]
    Role of mucus layers in gut infection and inflammation - PMC
    Pathogens infecting the small intestine have to avoid being trapped by the mucus. It is important for these pathogens to move efficiently towards the epithelium ...
  55. [55]
    Intestinal mucus components and secretion mechanisms: what we ...
    Apr 3, 2023 · The impairment or loss of the protective mucus layer may cause direct exposure of the intestinal epithelium to microorganisms or pathogens, ...
  56. [56]
    Role of thymic stromal lymphopoietin in allergy and beyond - Nature
    Jun 1, 2022 · Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat. Immunol. 3, 673–680 (2002). This study ...
  57. [57]
    Epithelial cell–derived cytokines: more than just signaling the alarm
    Indeed, TSLP can condition dendritic cells to initiate type 2 responses, and IL-33 may influence susceptibility to asthma through its role in establishing ...
  58. [58]
    Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial ...
    Intestinal Th17 cells are induced and accumulate in response to colonization with a subgroup of intestinal microbes such as segmented filamentous bacteria ...
  59. [59]
    IL-23 and Th17 cytokines in intestinal homeostasis - Nature
    Jul 2, 2008 · We will review the evidence linking the IL-23/Th17 axis to chronic intestinal inflammation and also will discuss its beneficial roles in intestinal protection ...
  60. [60]
    Mechanisms and modeling of wound repair in the intestinal epithelium
    In the small intestine, following barrier injury, there are three phases of wound healing: villus contraction, cell migration (restitution), and paracellular ...
  61. [61]
    Epithelial JAM-A is fundamental for intestinal wound repair in vivo
    Aug 9, 2022 · Such wound-associated epithelial cells undergo collective cell migration to initiate repair within a few hours of intestinal mucosal damage (1).Missing: post- | Show results with:post-
  62. [62]
    Wound healing of intestinal epithelial cells - PMC - PubMed Central
    It has been shown that the activation of specific signaling pathways is involved in intestinal epithelial wound repair. El-Assal et al[11] have demonstrated ...
  63. [63]
    Role of type I and type III interferons in gastrointestinal homeostasis ...
    Type III IFNs are prominent in antiviral protection of the intestinal epithelium, while type I IFNs play a larger role in non-epithelial cells. Type I IFNs can ...
  64. [64]
    Importance of Type I and III Interferons at Respiratory and Intestinal ...
    In the intestinal tract, type III IFNs have been shown to play a key role in helping to maintain this balance and protecting the intestinal epithelial cells ...
  65. [65]
    Epithelial Cell Inflammasomes in Intestinal Immunity and Inflammation
    Inflammasome formation and caspase-1 activation lead to cleavage and secretion of the active forms of IL-1 family member cytokines, such as IL-1β and IL-18.
  66. [66]
    Salmonella enterica Serovar Typhimurium Induces NAIP/NLRC4
    Jun 9, 2022 · NLRP3 and ASC are dispensable for inflammasome activation in response to Salmonella in human intestinal epithelial cells. Since the NAIP/NLRC4 ...
  67. [67]
    Lower human defensin 5 in elderly people compared to middle ...
    Jun 8, 2021 · This study aims to clarify relationships between the amount of human defensin 5 (HD5), a Paneth cell α-defensin, which is known to regulate the intestinal ...<|separator|>
  68. [68]
    Evidence that the loss of colonic anti-microbial peptides may ...
    This study supports the proposed model that aging-related loss of colonic crypt epithelial cell AMP gene expression promotes increased relative abundances of ...
  69. [69]
    Aging amplifies a gut microbiota immunogenic signature linked to ...
    Mar 27, 2024 · Aging is associated with low-grade inflammation that increases the risk of infection and disease, yet the underlying mechanisms remain unclear.
  70. [70]
    Sialidases and fucosidases of Akkermansia muciniphila are crucial ...
    Apr 1, 2023 · This study brings unprecedented mechanistic insight into the initiation of mucin O-glycan degradation by A. muciniphila and nutrient sharing between mucus- ...
  71. [71]
    Microbes vs. chemistry in the origin of the anaerobic gut lumen - PNAS
    Apr 2, 2018 · The gut microbes consume oxygen, which diffuses into the lumen from the intestinal tissue, maintaining the lumen in a deeply anaerobic state.
  72. [72]
    Microbiota-derived butyrate dynamically regulates intestinal ...
    May 12, 2020 · We showed the selective induction of SYNPO as an intestinal tight junction protein with a central role in epithelial barrier regulation.
  73. [73]
    Succinate-producing microbiota drives tuft cell hyperplasia to protect ...
    Nov 26, 2024 · The microbial metabolite succinate promotes tuft cell hyperplasia, which enhances the gut's ability to protect against intestinal infections ...
  74. [74]
    IgA regulates the composition and metabolic function of gut ...
    Jul 24, 2018 · IgA regulates the composition and metabolic function of gut microbiota by promoting symbiosis between bacteria.
  75. [75]
    Neonatal microbiota colonization primes maturation of goblet cell ...
    May 5, 2025 · Microbiota colonization drives postnatal GC maturation. After investigating microbiota influences on IML maturation, we examined how age and ...
  76. [76]
    Microbiota-derived extracellular vesicles: current knowledge, gaps ...
    Numerous studies have formed that BEVs contain DNA in the lumen and on the surface that they transport to other bacteria by horizontal transfer and that ...Missing: exchange | Show results with:exchange
  77. [77]
    Intestinal Barrier Dysfunction in Inflammatory Bowel Disease
    Sep 29, 2023 · This review aims to examine the role of intestinal barrier dysfunction in the development of inflammatory bowel disease
  78. [78]
    The Role of IL-23 in the Pathogenesis and Therapy of Inflammatory ...
    Jun 15, 2023 · IL-23 interacts with both the innate and adaptive immune systems, and IL-23/Th17 appears to be involved in the development of chronic intestinal inflammation.4.1. The Il-23-Th17 Axis · 4.3. Regulatory T Cells · 9. Treatment
  79. [79]
    Global evolution of inflammatory bowel disease across ... - Nature
    Apr 30, 2025 · The changing epidemiology of IBD is characterized in terms of incidence (new diagnoses reported per 100,000 person-years) and prevalence (total ...
  80. [80]
    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 ...α-Gliadin Affinity Column... · Cxcr3 Is Expressed Both In... · Figure 3. Cxcr3 Is Expressed...Missing: lymphocytosis | Show results with:lymphocytosis
  81. [81]
    Celiac Disease - StatPearls - NCBI Bookshelf - NIH
    Feb 4, 2025 · Gliadin also increases intestinal permeability, allowing more peptides to pass through the intestinal barrier and interact with the immune ...
  82. [82]
    Clostridium difficile Toxins Disrupt Epithelial Barrier Function by ...
    difficile toxins enhance paracellular permeability of T84 epithelial monolayers. T84 monolayers were incubated with C. difficile toxin A-10463 (TcdA, 240 ng/ml) ...
  83. [83]
    Replication of Human Noroviruses in Stem Cell-Derived ... - NIH
    We report successful cultivation of multiple HuNoV strains in enterocytes in stem cell-derived, nontransformed human intestinal enteroid monolayer cultures.<|separator|>
  84. [84]
    Polyclonality overcomes fitness barriers in Apc-driven tumorigenesis
    Oct 30, 2024 · APC-mutant intestinal stem cells ... The intestinal stem cell signature identifies colorectal cancer stem cells and predicts disease relapse.
  85. [85]
    Intestinal adaptation after massive intestinal resection - PMC - NIH
    Patients with short bowel syndrome require long term parenteral nutrition support. However, after massive intestinal resection the intestine undergoes ...
  86. [86]
    Intestinal organoids in inflammatory bowel disease - Frontiers
    Intestinal organoids are not only valuable for understanding disease mechanisms but are also revolutionizing drug discovery and personalized therapy for IBD.
  87. [87]
    Current applications of intestinal organoids: a review
    May 31, 2024 · We have summarized the current applications of intestinal organoid technology in disease modeling, drug screening, and regenerative medicine.
  88. [88]
    Stem cell-derived intestinal organoids: a novel modality for IBD
    Jul 21, 2023 · Intestinal organoids can be used for intestinal development and IBD disease modeling, drug/toxicity testing, and host-pathogen interaction ...
  89. [89]
    Intestinal cellular heterogeneity and disease development revealed ...
    Sep 1, 2022 · In this review, we summarize the single-cell RNA sequencing applications to understand intestinal cell characteristics, spatiotemporal evolution, and ...
  90. [90]
    Tissue and cellular spatiotemporal dynamics in colon aging - Nature
    Oct 22, 2025 · The cellular branch of the atlas encompassed a total of ~400,000 snRNA-seq profiles (n = 21), integrating newly generated and publicly available ...
  91. [91]
    The therapeutic potential of mesenchymal stem cells in intestinal ...
    Jul 21, 2025 · MSCs alleviate chemotherapy-induced intestinal damage by promoting the regeneration and repair of intestinal epithelial cells, thereby improving ...
  92. [92]
    The therapeutic potential of mesenchymal stem cells in intestinal ...
    Jul 21, 2025 · Following injury, the intestinal epithelium demonstrates significant repair and regenerative capacity. After radiation-induced stem cell ...
  93. [93]
    Anti-TNF-α antibodies improve intestinal barrier function in Crohn's ...
    The present study demonstrates that anti-TNF-α antibodies are able to restore the impaired intestinal permeability of patients with active Crohn's disease.Missing: biologics | Show results with:biologics
  94. [94]
    Therapeutic faecal microbiota transplantation controls intestinal ...
    Dec 5, 2018 · We show that therapeutic FMT reduces colonic inflammation and initiates the restoration of intestinal homeostasis through the simultaneous activation of ...
  95. [95]
    Fecal microbiota transplantation and next-generation therapies
    May 31, 2024 · Fecal microbiota transplantation has received considerable attention as a promising therapy to modulate the gut microbiome and restore microbial ...
  96. [96]
    An organoid-based CRISPR-Cas9 screen for regulators of intestinal ...
    Jul 12, 2023 · Our approach demonstrates the utility of organoid models in the identification of factors regulating cell fate and state transitions during tissue maturation.
  97. [97]
    Efficient genetic editing of human intestinal organoids using ...
    Oct 5, 2023 · Here, we describe an efficient method for intestinal organoid editing using a ribonucleoprotein-based CRISPR approach. Editing efficiencies of ...
  98. [98]
    Artificial Intelligence-Based Quantitative Structure–Property ...
    Apr 18, 2023 · Sun et al. focused on permeability prediction through the intestinal wall expressed as Peff (human effective intestinal membrane permeability).
  99. [99]
    Technological advances and challenges in constructing complex gut ...
    Aug 14, 2024 · Traditional organoid models, though providing valuable insights into intestinal biology, are limited by their lack of vascular structures, ...
  100. [100]
    Intestinal organoids: roadmap to the clinic
    Jun 25, 2021 · The purpose of this article is to outline the critical areas and challenges that must be addressed when transitioning laboratory-based discovery.<|separator|>
  101. [101]
    Organoids: a systematic review of ethical issues
    Jul 23, 2022 · Organoids might be better models than animals, for instance, for the purposes of drug screening, because they are grown from human, even patient ...
  102. [102]
    Ethical issues in human organoid and gastruloid research
    Mar 15, 2017 · Research involving human organoids and gastruloids involves ethical issues associated with their derivation as well as their current and future uses.