Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 723: 150176, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-38820627

ABSTRACT

Epithelial homeostasis is fundamental for the physiological functions of colon tissue. Dysregulation of colon epithelial structure leads to abnormal immune responses and diseases such as inflammatory bowel disease. In this work we found long non-coding RNA DANCR was a novel regulator to colon epithelial homeostasis. Silencing DANCR resulted in decreased expression of epithelial barrier proteins and enhanced susceptibility to TNFα stimulation, which was accompanied by hyperactivation of the NF-κB pathway. Mechanistical studies revealed DANCR modulated the expression of a protein methyltransferase SET7 to suppress responses to TNFα, as well as the activity of NF-κB pathway. In summary, DANCR regulated colon epithelial homeostasis through modulating the TNFα/NF-κB axis. These findings cast light on the discovery of novel regulators to colon epithelial homeostasis and added new evidence to the physiological functions of DANCR.


Subject(s)
Colon , Homeostasis , NF-kappa B , RNA, Long Noncoding , Signal Transduction , Tumor Necrosis Factor-alpha , NF-kappa B/metabolism , Colon/metabolism , Humans , Tumor Necrosis Factor-alpha/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Intestinal Mucosa/metabolism , Animals , Epithelial Cells/metabolism
2.
Free Radic Biol Med ; 205: 224-233, 2023 08 20.
Article in English | MEDLINE | ID: mdl-37315703

ABSTRACT

Mucosal healing has emerged as a therapeutic goal to achieve lasting clinical remission in ulcerative colitis. Intestinal repair in response to inflammation presumably requires higher energy supplies for the restoration of intestinal barrier and physiological functions. However, epithelial energy metabolism during intestinal mucosal healing has been little studied, whereas inflammation-induced alterations have been reported in the main energy production site, the mitochondria. The aim of the present work was to assess the involvement of mitochondrial activity and the events influencing their function during spontaneous epithelial repair after colitis induction in mouse colonic crypts. The results obtained show adaptations of colonocyte metabolism during colitis to ensure maximal ATP production for supporting energetic demand by both oxidative phosphorylation and glycolysis in a context of decreased mitochondrial biogenesis and through mitochondrial function restoration during colon epithelial repair. In parallel, colitis-induced mitochondrial ROS production in colonic epithelial cells was rapidly associated with transient expression of GSH-related enzymes. Mitochondrial respiration in colonic crypts was markedly increased during both inflammatory and recovery phases despite decreased expression of several mitochondrial respiratory chain complex subunits after colitis induction. Rapid induction of mitochondrial fusion was associated with mitochondrial function restoration. Finally, in contrast with the kinetics expression of genes involved in mitochondrial oxidative metabolism and in glycolysis, the expression of glutaminase was markedly reduced in the colonic crypts both during colitis and repair phases. Overall, our data suggest that the epithelial repair after colitis induction is characterized by a rapid and transient increased capacity for mitochondrial ATP production in a context of apparent restoration of mitochondrial biogenesis and metabolic reorientation of energy production. The potential implication of energy production adaptations within colonic crypts to sustain mucosal healing in a context of altered fuel supply is discussed.


Subject(s)
Colitis , Animals , Mice , Colitis/chemically induced , Colitis/genetics , Colon/metabolism , Inflammation/metabolism , Mitochondria/metabolism , Intestinal Mucosa/metabolism , Energy Metabolism , Adenosine Triphosphate/metabolism , Dextran Sulfate , Mice, Inbred C57BL , Disease Models, Animal
3.
Food Chem Toxicol ; 173: 113611, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36657700

ABSTRACT

Microcystin-leucine arginine (MCLR) is a phycotoxin produced by cyanobacteria. As a hepatotoxin, increasing evidence suggests that it has some negative effects on the mammal gastrointestinal tract, but further studies are warranted. In this study, we investigated the effects of MCLR on the intestinal epithelial microenvironment by oral administration of MCLR. As expected, MCLR at doses of 200 and 400 µg kg-1 bw showed hepatorenal toxicity in rats but without significant gastrointestinal symptoms. MCLR exposure decreased the thickness of the colonic epithelial mucus layer, and down-regulated the expression of main mucin protein (MUC2), cytoskeletal assembly-related genes (Arpc1a, Enah) and cytoskeletal stability-related genes (Ptk2, Prkca, Actn1, Pxn, Tln1, Cttn, Vcl) in colonic tissue to varying degrees, but did not affect the expression of cell connection-related genes including Zo1, Ocln, Cldn2 and Cdh1. In addition, MCLR exposure had a limited effect on gut bacterial diversity but clearly enriched specific bacteria. Prevotella, which plays a crucial role in balancing health and disease, was inhibited, whereas Muribaculaceae concerning the epithelial barrier, was promoted. Together, our findings demonstrate that MCLR exposure can weaken the colonic epithelial barrier by interfering with the stability of the cytoskeleton, which in turn exacerbates the homeostasis maintenance in the intestinal microenvironment.


Subject(s)
Cyanobacteria , Microcystins , Rats , Animals , Microcystins/toxicity , Marine Toxins/metabolism , Liver , Cytoskeleton/metabolism , Cyanobacteria/metabolism , Mammals , Cortactin/metabolism , Cortactin/pharmacology
4.
FEBS J ; 289(23): 7466-7485, 2022 12.
Article in English | MEDLINE | ID: mdl-35730982

ABSTRACT

The basement membrane (BM) underlying epithelial tissue is a thin layer of extracellular matrix that governs tissue integrity and function. Epithelial BMs are generally assembled using BM components secreted from two origins: epithelium and stroma. Although de novo BM formation involves self-assembly processes of large proteins, it remains unclear how stroma-derived macromolecules are transported and assembled, specifically in the BM region. In this study, we established an in vitro co-culture model of BM formation in which DLD-1 human colon epithelial cells were cultured on top of collagen I gel containing human embryonic OUMS-36T-2 fibroblasts as stromal cells. A distinct feature of our system is represented by OUMS-36T-2 cells which are almost exclusively responsible for synthesis of collagen IV, a major BM component. Exploiting this advantage, we found that collagen IV incorporation was significantly impaired in culture conditions where OUMS-36T-2 cells were not allowed to directly contact DLD-1 cells. Soluble collagen IV, once diluted in the culture medium, did not accumulate in the BM region efficiently. Live imaging of fluorescently tagged collagen IV revealed that OUMS-36T-2 cells deposited collagen IV aggregates directly onto the basal surface of DLD-1 cells. Collectively, these results indicate a novel mode of collagen IV deposition in which fibroblasts proximal to epithelial cells exclusively contribute to collagen IV assembly during BM formation.


Subject(s)
Colon , Epithelial Cells , Humans , Basement Membrane , Collagen
5.
Adv Biol (Weinh) ; 6(11): e2200129, 2022 11.
Article in English | MEDLINE | ID: mdl-35773243

ABSTRACT

The human colon plays a critical role in fluid and salt absorption and harbors the largest immune compartment. There is a widespread need for in vitro models of human colon physiology with its innate immune system. A method is described to produce a cassette with a network of struts supporting a suspended, non-chemically cross-linked collagen hydrogel scaffold compatible with the co-culture of primary gastrointestinal epithelium and migratory inflammatory cells. The epithelial monolayer cultured on the suspended collagen possesses a population of polarized and differentiated cells similar to that present in vivo. This epithelial layer displays proper barrier function with a transepithelial electrical resistance (TEER) ≥ 1,500 Ω cm2 and an apparent permeability ≤10-5 cm2 s-1 . Immune cells plated on the basal face of the scaffold transmigrated over a period of 24 h to the epithelial layer in response to epithelial production of IL-8 induced by luminal stimulation of Clostridium difficile Toxin A. These studies demonstrate that this in vitro platform possesses a functional primary colonic epithelial layer with an immune cell compartment capable of recruitment in response to pro-inflammatory cues coming from the epithelium.


Subject(s)
Colon , Hydrogels , Humans , Hydrogels/pharmacology , Cells, Cultured , Collagen , Cell Communication
6.
Am J Physiol Cell Physiol ; 322(5): C960-C966, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35385327

ABSTRACT

The syndecans are a family of transmembrane proteoglycans that are widespread in mammalian tissues. Located at the cell surface membrane, they contribute to modulating the composition of the extracellular matrix via glycosaminoglycan chains (GAGs) attached to their extracellular domains. Syndecans can interact with a variety of extracellular ligands through their core proteins and GAGs, and may also transmit signals through their transmembrane domain to regulate intracellular functions. These properties enable syndecan to modulate glycocalyx formation, epithelial cell-to-cell connections for cell barrier formation, and epithelial cell-lamina propria interactions in the colon epithelium, all of which are crucial for the homeostasis of this tissue. Inflammation induces structural alterations of the colon epithelium, and accumulating evidence suggests that syndecan expression might play important regulatory functions during inflammation. This review summarizes the possible roles of syndecans in maintaining tissue homeostasis in the colon epithelium, especially under inflammation.


Subject(s)
Colon , Inflammation , Animals , Colon/metabolism , Epithelium/metabolism , Homeostasis , Mammals/metabolism , Syndecans/metabolism
7.
Cell Rep ; 37(11): 110068, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34910915

ABSTRACT

Microenvironmental factors modulating age-related DNA damage are unclear. Non-pituitary growth hormone (npGH) is induced in human colon, non-transformed human colon cells, and fibroblasts, and in 3-dimensional intestinal organoids with age-associated DNA damage. Autocrine/paracrine npGH suppresses p53 and attenuates DNA damage response (DDR) by inducing TRIM29 and reducing ATM phosphorylation, leading to reduced DNA repair and DNA damage accumulation. Organoids cultured up to 4 months exhibit aging markers, p16, and SA-ß-galactosidase and decreased telomere length, as well as DNA damage accumulation, with increased npGH, suppressed p53, and attenuated DDR. Suppressing GH in aged organoids increases p53 and decreases DNA damage. WT mice exhibit age-dependent colon DNA damage accumulation, while in aged mice devoid of colon GH signaling, DNA damage remains low, with elevated p53. As age-associated npGH induction enables a pro-proliferative microenvironment, abrogating npGH signaling could be targeted as anti-aging therapy by impeding DNA damage and age-related pathologies.


Subject(s)
Aging , Carrier Proteins/physiology , Colon/pathology , DNA Damage , Fibroblasts/pathology , Human Growth Hormone/metabolism , Intestinal Mucosa/pathology , Animals , Colon/metabolism , DNA Repair , Fibroblasts/metabolism , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
8.
Nutrients ; 13(2)2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33498991

ABSTRACT

The large intestinal epithelium is confronted with the necessity to adapt quickly to varying levels of oxygenation. In contrast to other tissues, it meets this requirement successfully and remains unharmed during (limited) hypoxic periods. The large intestine is also the site of bacterial fermentation producing short-chain fatty acids (SCFA). Amongst these SCFA, butyrate has been reported to ameliorate many pathological conditions. Thus, we hypothesized that butyrate protects the colonocytes from hypoxic damage. We used isolated porcine colon epithelium mounted in Ussing chambers, incubated it with or without butyrate and simulated hypoxia by changing the gassing regime to test this hypothesis. We found an increase in transepithelial conductance and a decrease in short-circuit current across the epithelia when simulating hypoxia for more than 30 min. Incubation with 50 mM butyrate significantly ameliorated these changes to the epithelial integrity. In order to characterize the protective mechanism, we compared the effects of butyrate to those of iso-butyrate and propionate. These two SCFAs exerted similar effects to butyrate. Therefore, we propose that the protective effect of butyrate on colon epithelium under hypoxia is not (only) based on its nutritive function, but rather on the intracellular signaling effects of SCFA.


Subject(s)
Butyrates/pharmacology , Colon/drug effects , Epithelium/drug effects , Fatty Acids, Volatile/metabolism , Gene Expression Regulation/drug effects , Oxygen/pharmacology , Animals , Biological Transport , Colon/metabolism , Electrophysiology , Epithelium/metabolism , Fatty Acids, Volatile/analysis , Fermentation/physiology , Hypoxia/physiopathology , Male , Oxygen/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Swine/physiology , Tissue Culture Techniques
9.
Trends Neurosci ; 43(3): 170-181, 2020 03.
Article in English | MEDLINE | ID: mdl-31983457

ABSTRACT

Visceral hypersensitivity and pain result, at least in part, from increased excitability of primary afferents that innervate the colon. In addition to intrinsic changes in these neurons, emerging evidence indicates that changes in lining epithelial cells may also contribute to increased excitability. Here we review recent studies on how colon epithelial cells communicate directly with colon afferents. Specifically, anatomical studies revealed specialized synaptic connections between epithelial cells and nerve fibers and studies using optogenetic activation of the epithelium showed initiation of pain-like responses. We review the possible mechanisms of epithelial-neuronal communication and provide an overview of the possible neurotransmitters and receptors involved. Understanding the biology of this interface and how it changes in pathological conditions may provide new treatments for visceral pain conditions.


Subject(s)
Visceral Pain , Colon , Communication , Humans , Neurons , Optogenetics
10.
Toxicology ; 412: 1-11, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30439556

ABSTRACT

Butyrate helps to maintain colon homeostasis and exhibits chemopreventive effects in colon epithelium. We examined the interactive effects of butyrate and benzo[a]pyrene (BaP), dietary carcinogen, in regulation of expression of a panel of phase I and II xenobiotic metabolizing enzymes (XMEs) in human colon cells. In human colon carcinoma HCT-116 and HT-29 cell lines, butyrate alone increased mRNA levels of some enzymes, such as N-acetyltransferases (in particular NAT2). In combination with BaP, butyrate potentiated induction of cytochrome P450 family 1 enzymes (CYP1A1), aldo-keto reductases (AKR1C1) or UDP-glucuronosyltransferases (UGT1A1). There were some notable differences between cell lines, as butyrate potentiated induction of NAD(P)H:quinone oxidoreductase 1 (NQO1) and UGT1A4 only in HCT-116 cells, and it even repressed AKR1C3 induction in HT-29 cells. Butyrate also promoted induction of CYP1, NQO1, NAT2, UGT1A1 or UGT1A4 in human colon Caco-2 cells, in a differentiation-dependent manner. Differentiated Caco-2 cells exhibited a higher inducibility of selected XME genes than undifferentiated cells. Butyrate increased induction of enzymatic activities of NATs, NQO1 and UGTs by BaP in HCT-116 and HT29 cells, whereas in differentiated Caco-2 cells it helped to increase only enzymatic activity of NQO1 and UGTs. Together, the present data suggest that butyrate may modulate expression/activities of several enzymes involved in metabolism of carcinogens in colon. In some cases (NAT2, UGT1 A1), this was linked to inhibition of histone deacetylases (HDAC), as confirmed by using HDAC inhibitor trichostatin A. These results may have implications for our understanding of the role of butyrate in regulation of XMEs and carcinogen metabolism in colon.


Subject(s)
Benzo(a)pyrene/toxicity , Butyrates/pharmacology , Carcinogens/toxicity , Oxidoreductases/metabolism , Transferases/metabolism , Cell Line , Colon/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Oxidoreductases/genetics , Transferases/genetics , Xenobiotics/metabolism
11.
J Nutr Sci Vitaminol (Tokyo) ; 64(4): 287-291, 2018.
Article in English | MEDLINE | ID: mdl-30175793

ABSTRACT

[6]-Gingerol possesses various beneficial pharmacological and therapeutic properties, including anti-carcinogenic and anti-inflammatory properties and the ability to regulate intestinal contraction. Recently, our group observed that the serosal administration of [6]-gingerol stimulated electrogenic sodium absorption in the rat colon via the capsaicin receptor, TRPV1. TRPV1 is known to be expressed in both the mucosal epithelium and the muscle layers in the colon. In the present study, we assessed whether [6]-gingerol stimulated sodium absorption via TRPV1 in the colonic mucosal epithelium. We compared the effect of [6]-gingerol on TRPV1-dependent colonic sodium absorption in the colon preparation with or without muscle layer. All experiments were performed by measuring the transmural potential difference (ΔPD) in an Ussing chamber system. [6]-Gingerol induced positive ΔPD when administered to the serosal side of the colon, and this effect was significantly larger in the colon preparation without muscle layer than in that with the muscle layer. In the colon preparation without muscle layer, the [6]-gingerol-dependent induction of ΔPD was markedly suppressed by mucosal addition of amiloride, a selective inhibitor of epithelial sodium channel. ΔPD induction by [6]-gingerol was considerably diminished by capsazepine, an inhibitor of the capsaicin receptor TRPV1, but not by AP-18, an inhibitor of TRPA1. These results suggest that [6]-gingerol induces amiloride-sensitive electrogenic sodium absorption in the rat colon via TRPV1 expressed in the colonic mucosal epithelium, and that this effect is independent of TRPV1 in the colonic muscle layer.


Subject(s)
Catechols/pharmacology , Colon/drug effects , Fatty Alcohols/pharmacology , Gastrointestinal Agents/pharmacology , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Sodium, Dietary/metabolism , TRPV Cation Channels/agonists , Amiloride/pharmacology , Animals , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Colon/physiology , Epithelial Sodium Channel Blockers/pharmacology , In Vitro Techniques , Intestinal Mucosa/physiology , Membrane Potentials/drug effects , Membrane Transport Modulators/pharmacology , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Oximes/pharmacology , Rats, Sprague-Dawley , TRPA1 Cation Channel/antagonists & inhibitors , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism
12.
Cells Tissues Organs ; 203(5): 267-286, 2017.
Article in English | MEDLINE | ID: mdl-28052271

ABSTRACT

Continuous cell lines have become indispensable tools that have enabled investigations into cellular mechanisms by increasing experimental reproducibility and sample availability, and decreasing the use of experimental animals. To facilitate studies of epithelial barrier function of the porcine colon, we aimed to establish an epithelial cell line with an extended replicative capacity. Cells were isolated from the proximal colon of a 3-week-old piglet and transduced using a recombinant retroviral vector construct containing the simian virus 40 large T antigen (SV40 TAg). We established a clonal epithelial cell line, referred to as PoCo83-3, that stably expressed the SV40 TAg, verified at mRNA and protein levels. PoCo83-3 showed epithelial cell-specific features, such as cobblestone-like morphology, dome structure formation, the presence of apical microvilli, and the expression of keratin 18, E-cadherin and the tight junction-associated proteins zonula occludens-1, occludin, and claudin-1. To validate PoCo83-3 as an in vitro model in epithelial barrier research, proinflammatory cytokine-inducible alterations in barrier integrity were demonstrated by incubating the cells with TNF-α and IFN-γ for 48 h. These cytokine treatments promoted a decreased transepithelial electrical resistance. In summary, PoCo83-3 exhibited an extended life span and a differentiated phenotype while maintaining epithelial characteristics. Based on these results, we present this cell line as a valuable in vitro model for investigations of epithelial barrier function in the porcine colon.


Subject(s)
Antigens, Viral, Tumor/genetics , Cell Culture Techniques/methods , Colon/cytology , Epithelial Cells/cytology , Transduction, Genetic , Animals , Cell Line , Cell Separation/methods , Cell Survival , Cells, Cultured , Colon/metabolism , Cryopreservation/methods , Epithelial Cells/metabolism , Genetic Vectors/genetics , Karyotype , Male , Swine
13.
Elife ; 52016 12 20.
Article in English | MEDLINE | ID: mdl-27996939

ABSTRACT

Previously we reported that Src-associated-substrate-during-mitosis-of-68kDa (Sam68/KHDRBS1) is pivotal for DNA damage-stimulated NF-κB transactivation of anti-apoptotic genes (Fu et al., 2016). Here we show that Sam68 is critical for genotoxic stress-induced NF-κB activation in the γ-irradiated colon and animal and that Sam68-dependent NF-κB activation provides radioprotection to colon epithelium in vivo. Sam68 deletion diminishes γ-irradiation-triggered PAR synthesis and NF-κB activation in colon epithelial cells (CECs), thus hampering the expression of anti-apoptotic molecules in situ and facilitating CECs to undergo apoptosis in mice post whole-body γ-irradiation (WBIR). Sam68 knockout mice suffer more severe damage in the colon and succumb more rapidly from acute radiotoxicity than the control mice following WBIR. Our results underscore the critical role of Sam68 in orchestrating genotoxic stress-initiated NF-κB activation signaling in the colon tissue and whole animal and reveal the pathophysiological relevance of Sam68-dependent NF-κB activation in colonic cell survival and recovery from extrinsic DNA damage.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colon/radiation effects , Gamma Rays , Intestinal Mucosa/radiation effects , NF-kappa B p50 Subunit/metabolism , RNA-Binding Proteins/metabolism , Animals , Mice, Knockout
14.
J Innate Immun ; 1(2): 145-52, 2009.
Article in English | MEDLINE | ID: mdl-20046221

ABSTRACT

One major route of intoxication by Bacillus anthracis (anthrax) spores is via their ingestion and subsequent uptake by the intestinal epithelium. Anthrax edema toxin (ETx) is an adenylate cyclase that causes persistent elevation of cAMP in intoxicated cells. NADPH oxidase enzymes (Nox1-Nox5, Duox1 and 2) generate reactive oxygen species (ROS) as components of the host innate immune response to bacteria, including Nox1 in gastrointestinal epithelial tissues. We show that ETx effectively inhibits ROS formation by Nox1 in HT-29 colon epithelial cells. This inhibition requires the PKA-mediated phosphorylation of the Nox1-regulatory component, NoxA1, and the subsequent binding of 14-3-3zeta. Inhibition of Nox1-mediated ROS formation in the gut epithelium may be a mechanism used by B. anthracis to circumvent the innate immune response.


Subject(s)
Anthrax/immunology , Antigens, Bacterial/metabolism , Bacillus anthracis/enzymology , Bacterial Toxins/metabolism , Colon/metabolism , Intestinal Mucosa/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Cell Line, Tumor , Colon/microbiology , Down-Regulation , Host-Pathogen Interactions , Humans , Immunity, Innate , Intestinal Mucosa/microbiology , NADPH Oxidase 1 , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL