Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Pflugers Arch ; 476(8): 1209-1219, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38829391

ABSTRACT

The intestinal epithelium is covered by mucus that protects the tissue from the luminal content. Studies have shown that anion secretion via the cystic fibrosis conductance regulator (Cftr) regulates mucus formation in the small intestine. However, mechanisms regulating mucus formation in the colon are less understood. The aim of this study was to explore the role of anion transport in the regulation of mucus formation during steady state and in response to carbamylcholine (CCh) and prostaglandin E2 (PGE2). The broad-spectrum anion transport inhibitor 4,4'-diisothiocyanatostilbene-2,2'-disulfonate (DIDS), CftrdF508 (CF) mice, and the slc26a3 inhibitor SLC26A3-IN-2 were used to inhibit anion transport. In the distal colon, steady-state mucus expansion was reduced by SLC26A3-IN-2 and normal in CF mice. PGE2 stimulated mucus expansion without de novo mucus release in wild type (WT) and CF colon via slc26a3 sensitive mechanisms, while CCh induced de novo mucus secretion in WT but not in CF colon. However, when added simultaneously, CCh and PGE2 stimulated de novo mucus secretion in the CF colon via DIDS-sensitive pathways. A similar response was observed in CF ileum that responded to CCh and PGE2 with DIDS-sensitive de novo mucus secretion. In conclusion, this study suggests that slc26a3 regulates colonic mucus expansion, while Cftr regulates CCh-induced de novo mucus secretion from ileal and distal colon crypts. Furthermore, these findings demonstrate that in the absence of a functional Cftr channel, parallel stimulation with CCh and PGE2 activates additional anion transport processes that help release mucus from intestinal goblet cells.


Subject(s)
Carbachol , Colon , Cystic Fibrosis Transmembrane Conductance Regulator , Dinoprostone , Mucus , Sulfate Transporters , Animals , Dinoprostone/metabolism , Dinoprostone/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Sulfate Transporters/metabolism , Sulfate Transporters/genetics , Mice , Colon/metabolism , Colon/drug effects , Mucus/metabolism , Mucus/drug effects , Carbachol/pharmacology , Mice, Inbred C57BL , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Antiporters/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Male
2.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33431687

ABSTRACT

Goblet cells (GCs) are specialized cells of the intestinal epithelium contributing critically to mucosal homeostasis. One of the functions of GCs is to produce and secrete MUC2, the mucin that forms the scaffold of the intestinal mucus layer coating the epithelium and separates the luminal pathogens and commensal microbiota from the host tissues. Although a variety of ion channels and transporters are thought to impact on MUC2 secretion, the specific cellular mechanisms that regulate GC function remain incompletely understood. Previously, we demonstrated that leucine-rich repeat-containing protein 26 (LRRC26), a known regulatory subunit of the Ca2+-and voltage-activated K+ channel (BK channel), localizes specifically to secretory cells within the intestinal tract. Here, utilizing a mouse model in which MUC2 is fluorescently tagged, thereby allowing visualization of single GCs in intact colonic crypts, we show that murine colonic GCs have functional LRRC26-associated BK channels. In the absence of LRRC26, BK channels are present in GCs, but are not activated at physiological conditions. In contrast, all tested MUC2- cells completely lacked BK channels. Moreover, LRRC26-associated BK channels underlie the BK channel contribution to the resting transepithelial current across mouse distal colonic mucosa. Genetic ablation of either LRRC26 or BK pore-forming α-subunit in mice results in a dramatically enhanced susceptibility to colitis induced by dextran sodium sulfate. These results demonstrate that normal potassium flux through LRRC26-associated BK channels in GCs has protective effects against colitis in mice.


Subject(s)
Colitis/genetics , Large-Conductance Calcium-Activated Potassium Channels/genetics , Mucin-2/genetics , Animals , Colitis/pathology , Colitis/prevention & control , Colitis/therapy , Colon/metabolism , Colon/pathology , Disease Models, Animal , Goblet Cells/metabolism , Goblet Cells/pathology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Membrane Potentials/genetics , Mice , Patch-Clamp Techniques
3.
J Allergy Clin Immunol ; 144(4): 1058-1073.e3, 2019 10.
Article in English | MEDLINE | ID: mdl-31175877

ABSTRACT

BACKGROUND: Food-induced anaphylaxis (FIA) is an IgE-dependent immune response that can affect multiple organs and lead to life-threatening complications. The processes by which food allergens cross the mucosal surface and are delivered to the subepithelial immune compartment to promote the clinical manifestations associated with food-triggered anaphylaxis are largely unexplored. OBJECTIVE: We sought to define the processes involved in the translocation of food allergens across the mucosal epithelial surface to the subepithelial immune compartment in FIA. METHODS: Two-photon confocal and immunofluorescence microscopy was used to visualize and trace food allergen passage in a murine model of FIA. A human colon cancer cell line, RNA silencing, and pharmacologic approaches were used to identify the molecular regulation of intestinal epithelial allergen uptake and translocation. Human intestinal organoid transplants were used to demonstrate the conservation of these molecular processes in human tissues. RESULTS: Food allergens are sampled by using small intestine (SI) epithelial secretory cells (termed secretory antigen passages [SAPs]) that are localized to the SI villous and crypt region. SAPs channel food allergens to lamina propria mucosal mast cells through an IL-13-CD38-cyclic adenosine diphosphate ribose (cADPR)-dependent process. Blockade of IL-13-induced CD38/cADPR-dependent SAP antigen passaging in mice inhibited induction of clinical manifestations of FIA. IL-13-CD38-cADPR-dependent SAP sampling of food allergens was conserved in human intestinal organoids. CONCLUSION: We identify that SAPs are a mechanism by which food allergens are channeled across the SI epithelium mediated by the IL-13/CD38/cADPR pathway, regulate the onset of FIA reactions, and are conserved in human intestine.


Subject(s)
Allergens/immunology , Anaphylaxis/immunology , Food Hypersensitivity/immunology , Interleukin-13/immunology , Intestinal Mucosa/immunology , Allergens/metabolism , Anaphylaxis/metabolism , Animals , Food Hypersensitivity/metabolism , Humans , Immunoglobulin E/immunology , Interleukin-13/metabolism , Intestinal Mucosa/metabolism , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID
4.
Immunol Rev ; 260(1): 8-20, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24942678

ABSTRACT

The gastrointestinal tract is covered by mucus that has different properties in the stomach, small intestine, and colon. The large highly glycosylated gel-forming mucins MUC2 and MUC5AC are the major components of the mucus in the intestine and stomach, respectively. In the small intestine, mucus limits the number of bacteria that can reach the epithelium and the Peyer's patches. In the large intestine, the inner mucus layer separates the commensal bacteria from the host epithelium. The outer colonic mucus layer is the natural habitat for the commensal bacteria. The intestinal goblet cells secrete not only the MUC2 mucin but also a number of typical mucus components: CLCA1, FCGBP, AGR2, ZG16, and TFF3. The goblet cells have recently been shown to have a novel gate-keeping role for the presentation of oral antigens to the immune system. Goblet cells deliver small intestinal luminal material to the lamina propria dendritic cells of the tolerogenic CD103(+) type. In addition to the gel-forming mucins, the transmembrane mucins MUC3, MUC12, and MUC17 form the enterocyte glycocalyx that can reach about a micrometer out from the brush border. The MUC17 mucin can shuttle from a surface to an intracellular vesicle localization, suggesting that enterocytes might control and report epithelial microbial challenge. There is communication not only from the epithelial cells to the immune system but also in the opposite direction. One example of this is IL10 that can affect and improve the properties of the inner colonic mucus layer. The mucus and epithelial cells of the gastrointestinal tract are the primary gate keepers and controllers of bacterial interactions with the host immune system, but our understanding of this relationship is still in its infancy.


Subject(s)
Enterocytes/physiology , Gastrointestinal Tract/immunology , Goblet Cells/physiology , Mucins/physiology , Mucous Membrane/immunology , Mucus/physiology , Animals , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Humans , Immune System , Mucous Membrane/metabolism , Mucous Membrane/microbiology , Mucus/chemistry , Mucus/microbiology , Peyer's Patches/immunology
5.
Immunology ; 152(4): 613-627, 2017 12.
Article in English | MEDLINE | ID: mdl-28746740

ABSTRACT

The intestinal lamina propria (LP) contains antigen-presenting cells with features of dendritic cells and macrophages, collectively referred to as mononuclear phagocytes (MNPs). Association of MNPs with the epithelium is thought to play an important role in multiple facets of intestinal immunity including imprinting MNPs with the ability to induce IgA production, inducing the expression of gut homing molecules on T cells, facilitating the capture of luminal antigens and microbes, and subsequent immune responses in the mesenteric lymph node (MLN). However, the factors promoting this process in the steady state are largely unknown, and in vivo models to test and confirm the importance of LP-MNP association with the epithelium for these outcomes are unexplored. Evaluation of epithelial expression of chemoattractants in mice where MNP-epithelial associations were impaired suggested CCL20 as a candidate promoting epithelial association. Expression of CCR6, the only known receptor for CCL20, was required for MNPs to associate with the epithelium. LP-MNPs from CCR6-/- mice did not display defects in acquiring antigen and stimulating T-cell responses in ex vivo assays or in responses to antigen administered systemically. However, LP-MNPs from CCR6-deficient mice were impaired at acquiring luminal and epithelial antigens, inducing IgA production in B cells, inducing immune responses in the MLN, and capturing and trafficking luminal commensal bacteria to the MLN. These findings identify a crucial role for CCR6 in promoting LP-MNPs to associate with the intestinal epithelium in the steady state to perform multiple functions promoting gut immune homeostasis.


Subject(s)
Dendritic Cells/immunology , Genomic Imprinting/immunology , Immunologic Surveillance , Intestinal Mucosa/immunology , Macrophages/immunology , Receptors, CCR6/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Dendritic Cells/cytology , Humans , Macrophages/cytology , Mice , Mice, Knockout , Receptors, CCR6/genetics , T-Lymphocytes/cytology , T-Lymphocytes/immunology
6.
Pflugers Arch ; 467(7): 1403-1415, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25139191

ABSTRACT

The colonic mucosa protects itself from the luminal content by secreting mucus that keeps the bacteria at a distance from the epithelium. For this barrier to be effective, the mucus has to be constantly replenished which involves exocytosis and expansion of the secreted mucins. Mechanisms involved in regulation of mucus exocytosis and expansion are poorly understood, and the aim of this study was to investigate whether epithelial anion secretion regulates mucus formation in the colon. The muscarinic agonist carbachol was used to induce parallel secretion of anions and mucus, and by using established inhibitors of ion transport, we studied how inhibition of epithelial transport affected mucus formation in mouse colon. Anion secretion and mucin exocytosis were measured by changes in membrane current and epithelial capacitance, respectively. Mucus thickness measurements were used to determine the carbachol effect on mucus growth. The results showed that the carbachol-induced increase in membrane current was dependent on NKCC1 co-transport, basolateral K(+) channels and Cftr activity. In contrast, the carbachol-induced increase in capacitance was partially dependent on NKCC1 and K(+) channel activity, but did not require Cftr activity. Carbachol also induced an increase in mucus thickness that was inhibited by the NKCC1 blocker bumetanide. However, mice that lacked a functional Cftr channel did not respond to carbachol with an increase in mucus thickness, suggesting that carbachol-induced mucin expansion requires Cftr channel activity. In conclusion, these findings suggest that colonic epithelial transport regulates mucus formation by affecting both exocytosis and expansion of the mucin molecules.


Subject(s)
Colon/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Intestinal Mucosa/metabolism , Potassium Channels/metabolism , Solute Carrier Family 12, Member 2/metabolism , Animals , Carbachol/pharmacology , Chlorides/metabolism , Colon/cytology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Exocytosis , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Ion Transport , Mice , Mice, Inbred C57BL , Mucins/metabolism , Muscarinic Agonists/pharmacology , Potassium/metabolism , Potassium Channels/genetics , Solute Carrier Family 12, Member 2/genetics
7.
Proc Natl Acad Sci U S A ; 109(15): 5645-50, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-22451922

ABSTRACT

MUC2, the major colonic mucin, forms large polymers by N-terminal trimerization and C-terminal dimerization. Although the assembly process for MUC2 is established, it is not known how MUC2 is packed in the regulated secretory granulae of the goblet cell. When the N-terminal VWD1-D2-D'D3 domains (MUC2-N) were expressed in a goblet-like cell line, the protein was stored together with full-length MUC2. By mimicking the pH and calcium conditions of the secretory pathway we analyzed purified MUC2-N by gel filtration, density gradient centrifugation, and transmission electron microscopy. At pH 7.4 the MUC2-N trimer eluted as a single peak by gel filtration. At pH 6.2 with Ca(2+) it formed large aggregates that did not enter the gel filtration column but were made visible after density gradient centrifugation. Electron microscopy studies revealed that the aggregates were composed of rings also observed in secretory granulae of colon tissue sections. The MUC2-N aggregates were dissolved by removing Ca(2+) and raising pH. After release from goblet cells, the unfolded full-length MUC2 formed stratified layers. These findings suggest a model for mucin packing in the granulae and the mechanism for mucin release, unfolding, and expansion.


Subject(s)
Calcium/metabolism , Gels/metabolism , Mucin-2/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Goblet Cells/metabolism , Goblet Cells/ultrastructure , Hydrogen-Ion Concentration , Mice , Mice, Inbred C57BL , Models, Molecular , Mucin-2/chemistry , Mucin-2/ultrastructure , Protein Structure, Tertiary
8.
Gut ; 63(2): 281-91, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23426893

ABSTRACT

OBJECTIVE: The inner mucus layer in mouse colon normally separates bacteria from the epithelium. Do humans have a similar inner mucus layer and are defects in this mucus layer a common denominator for spontaneous colitis in mice models and ulcerative colitis (UC)? METHODS AND RESULTS: The colon mucus layer from mice deficient in Muc2 mucin, Core 1 O-glycans, Tlr5, interleukin 10 (IL-10) and Slc9a3 (Nhe3) together with that from dextran sodium sulfate-treated mice was immunostained for Muc2, and bacterial localisation in the mucus was analysed. All murine colitis models revealed bacteria in contact with the epithelium. Additional analysis of the less inflamed IL-10(-/-) mice revealed a thicker mucus layer than wild-type, but the properties were different, as the inner mucus layer could be penetrated both by bacteria in vivo and by fluorescent beads the size of bacteria ex vivo. Clear separation between bacteria or fluorescent beads and the epithelium mediated by the inner mucus layer was also evident in normal human sigmoid colon biopsy samples. In contrast, mucus on colon biopsy specimens from patients with UC with acute inflammation was highly penetrable. Most patients with UC in remission had an impenetrable mucus layer similar to that of controls. CONCLUSIONS: Normal human sigmoid colon has an inner mucus layer that is impenetrable to bacteria. The colon mucus in animal models that spontaneously develop colitis and in patients with active UC allows bacteria to penetrate and reach the epithelium. Thus colon mucus properties can be modulated, and this suggests a novel model of UC pathophysiology.


Subject(s)
Colitis, Ulcerative/microbiology , Colitis/microbiology , Colon/microbiology , Intestinal Mucosa/microbiology , Mucin-2/metabolism , Mucus/microbiology , Adolescent , Adult , Aged , Animals , Colitis/metabolism , Colitis/pathology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colon/metabolism , Colon/pathology , Female , Humans , In Situ Hybridization, Fluorescence , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Middle Aged , Young Adult
9.
Am J Physiol Cell Physiol ; 305(4): C457-67, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23784542

ABSTRACT

We have reported that transmembrane mucin MUC17 binds PDZ protein PDZK1, which retains MUC17 apically in enterocytes. MUC17 and transmembrane mucins MUC3 and MUC12 are suggested to build the enterocyte apical glycocalyx. Carbachol (CCh) stimulation of the small intestine results in gel-forming mucin secretion from goblet cells, something that requires adjacent enterocytes to secrete chloride and bicarbonate for proper mucin formation. Surface labeling and confocal imaging demonstrated that apically expressed MUC17 in Caco-2 cells and Muc3(17) in murine enterocytes were endocytosed upon stimulation with CCh. Relocation of MUC17 in response to CCh was specific as MUC3 and MUC12 did not relocate following CCh stimulation. MUC17 colocalized with PDZK1 under basal conditions, while MUC17 relocated to the terminal web and into early endosomes after CCh stimulation. CCh stimulation concomitantly internalized the Na(+/)H(+) exchanger 3 (NHE3) and recruited cystic fibrosis transmembrane conductance regulator (CFTR) to the apical membranes, a process that was important for CFTR-mediated bicarbonate secretion necessary for proper gel-forming mucin unfolding. The reason for the specific internalization of MUC17 is not understood, but it could limit the diffusion barrier for ion secretion caused by the apical enterocyte glycocalyx or alternatively act to sample luminal bacteria. Our results reveal well-orchestrated mucus secretion and trafficking of ion channels and the MUC17 mucin.


Subject(s)
Carbachol/pharmacology , Cell Membrane/drug effects , Cholinergic Agonists/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Endocytosis/drug effects , Enterocytes/drug effects , Mucins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Biotinylation , Caco-2 Cells , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Endosomes/drug effects , Endosomes/metabolism , Enterocytes/metabolism , Humans , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Protein Transport , Sodium-Hydrogen Exchanger 3 , Time Factors
10.
Am J Physiol Gastrointest Liver Physiol ; 305(5): G341-7, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23832518

ABSTRACT

Colon has been shown to have a two-layered mucus system where the inner layer is devoid of bacteria. However, a complete overview of the mouse gastrointestinal mucus system is lacking. We now characterize mucus release, thickness, growth over time, adhesive properties, and penetrability to fluorescent beads from stomach to distal colon. Colon displayed spontaneous mucus release and all regions released mucus in response to carbachol and PGE2, except the distal colon and domes of Peyer's patches. Stomach and colon had an inner mucus layer that was adherent to the epithelium. In contrast, the small intestine and Peyer's patches had a single mucus layer that was easily aspirated. The inner mucus layer of the distal colon was not penetrable to beads the size of bacteria and the inner layer of the proximal colon was only partly penetrable. In contrast, the inner mucus layer of stomach was fully penetrable, as was the small intestinal mucus. This suggests a functional organization of the intestinal mucus system, where the small intestine has loose and penetrable mucus that may allow easy penetration of nutrients, in contrast to the stomach, where the mucus provides physical protection, and the colon, where the mucus separates bacteria from the epithelium. This knowledge of the mucus system and its organization improves our understanding of the gastrointestinal tract physiology.


Subject(s)
Colon/metabolism , Gastric Mucosa/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Mucus/metabolism , Peyer's Patches/metabolism , Adhesiveness , Animals , Carbachol/pharmacology , Colon/drug effects , Colon/microbiology , Dinoprostone/pharmacology , Female , Fluorescent Dyes/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/microbiology , Intestinal Absorption , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Intestine, Small/drug effects , Intestine, Small/microbiology , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Video , Mucus/microbiology , Permeability , Peyer's Patches/drug effects , Time Factors
11.
Am J Physiol Gastrointest Liver Physiol ; 305(5): G348-56, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23832517

ABSTRACT

The mucus that protects the surface of the gastrointestinal tract is rich in specialized O-glycoproteins called mucins, but little is known about other mucus proteins or their variability along the gastrointestinal tract. To ensure that only mucus was analyzed, we combined collection from explant tissues mounted in perfusion chambers, liquid sample preparation, single-shot mass spectrometry, and specific bioinformatics tools, to characterize the proteome of the murine mucus from stomach to distal colon. With our approach, we identified ∼1,300 proteins in the mucus. We found no differences in the protein composition or abundance between sexes, but there were clear differences in mucus along the tract. Noticeably, mucus from duodenum showed similarities to the stomach, probably reflecting the normal distal transport. Qualitatively, there were, however, fewer differences than might had been anticipated, suggesting a relatively stable core proteome (∼80% of the total proteins identified). Quantitatively, we found significant differences (∼40% of the proteins) that could reflect mucus specialization throughout the gastrointestinal tract. Hierarchical clustering pinpointed a number of such proteins that correlated with Muc2 (e.g., Clca1, Zg16, Klk1). This study provides a deeper knowledge of the gastrointestinal mucus proteome that will be important in further understanding this poorly studied mucosal protection system.


Subject(s)
Colon/metabolism , Gastric Mucosa/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Mucin 5AC/metabolism , Mucin-2/metabolism , Proteomics , Animals , Biotinylation , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Cluster Analysis , Female , Male , Mice , Mice, Inbred C57BL , Mucus/metabolism , Proteomics/methods , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
12.
Am J Physiol Gastrointest Liver Physiol ; 302(4): G430-8, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22159279

ABSTRACT

The colon mucus layers minimize the contact between the luminal flora and the epithelial cells, and defects in this barrier may lead to colonic inflammation. We now describe an ex vivo method for analysis of mucus properties in human colon and mouse small and large intestine. Intestinal explants were mounted in horizontal perfusion chambers. The mucus surface was visualized by adding charcoal particles on the apical side, and mucus thickness was measured using a micropipette. Mucus thickness, adhesion, and growth rate were recorded for 1 h. In mouse and human colon, the ability of the mucus to act as a barrier to beads the size of bacteria was also evaluated. Tissue viability was monitored by transepithelial potential difference. In mouse ileum, the mucus could be removed by gentle aspiration, whereas in colon ∼40 µm of the mucus remained attached to the epithelial surface. Both mouse and human colon had an inner mucus layer that was not penetrated by the fluorescent beads. Spontaneous mucus growth was observed in human (240 µm/h) and mouse (100 µm/h) colon but not in mouse ileum. In contrast, stimulation with carbachol induced a higher mucus secretion in ileum than colon (mouse ileum: Δ200 µm, mouse colon: Δ130 µm, human colon: Δ140 µm). In conclusion, while retaining key properties from the mucus system in vivo, this setup also allows for studies of the highly dynamic mucus system under well-controlled conditions.


Subject(s)
Colon/physiology , Ileum/physiology , Intestinal Mucosa/physiology , Mucus/physiology , Animals , Colon/anatomy & histology , Colon/pathology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Humans , Ileum/anatomy & histology , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/pathology , Mice , Mucus/cytology , Mucus/microbiology
13.
Cell Mol Life Sci ; 68(22): 3635-41, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21947475

ABSTRACT

In discussions on intestinal protection, the protective capacity of mucus has not been very much considered. The progress in the last years in understanding the molecular nature of mucins, the main building blocks of mucus, has, however, changed this. The intestinal enterocytes have their apical surfaces covered by transmembrane mucins and the whole intestinal surface is further covered by mucus, built around the gel-forming mucin MUC2. The mucus of the small intestine has only one layer, whereas the large intestine has a two-layered mucus where the inner, attached layer has a protective function for the intestine, as it is impermeable to the luminal bacteria.


Subject(s)
Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/chemistry , Intestinal Mucosa/metabolism , Animals , Enterocytes/chemistry , Enterocytes/cytology , Enterocytes/metabolism , Humans , Immunity, Mucosal/immunology , Intestinal Mucosa/microbiology , Intestines/anatomy & histology , Intestines/microbiology , Intestines/physiology , Models, Molecular , Mucins/chemistry , Mucins/metabolism
14.
Nat Rev Gastroenterol Hepatol ; 19(12): 785-803, 2022 12.
Article in English | MEDLINE | ID: mdl-36097076

ABSTRACT

The intestinal tract faces numerous challenges that require several layers of defence. The tight epithelium forms a physical barrier that is further protected by a mucus layer, which provides various site-specific protective functions. Mucus is produced by goblet cells, and as a result of single-cell RNA sequencing identifying novel goblet cell subpopulations, our understanding of their various contributions to intestinal homeostasis has improved. Goblet cells not only produce mucus but also are intimately linked to the immune system. Mucus and goblet cell development is tightly regulated during early life and synchronized with microbial colonization. Dysregulation of the developing mucus systems and goblet cells has been associated with infectious and inflammatory conditions and predisposition to chronic disease later in life. Dysfunctional mucus and altered goblet cell profiles are associated with inflammatory conditions in which some mucus system impairments precede inflammation, indicating a role in pathogenesis. In this Review, we present an overview of the current understanding of the role of goblet cells and the mucus layer in maintaining intestinal health during steady-state and how alterations to these systems contribute to inflammatory and infectious disease.


Subject(s)
Goblet Cells , Mucins , Humans , Goblet Cells/pathology , Goblet Cells/physiology , Mucins/genetics , Mucus , Intestines , Homeostasis , Intestinal Mucosa/pathology
15.
Front Immunol ; 13: 953196, 2022.
Article in English | MEDLINE | ID: mdl-36177011

ABSTRACT

The intestinal tract is an ecosystem in which the resident microbiota lives in symbiosis with its host. This symbiotic relationship is key to maintaining overall health, with dietary habits of the host representing one of the main external factors shaping the microbiome-host relationship. Diets high in fiber and low in fat and sugars, as opposed to Western and high-fat diets, have been shown to have a beneficial effect on intestinal health by promoting the growth of beneficial bacteria, improve mucus barrier function and immune tolerance, while inhibiting pro-inflammatory responses and their downstream effects. On the contrary, diets low in fiber and high in fat and sugars have been associated with alterations in microbiota composition/functionality and the subsequent development of chronic diseases such as food allergies, inflammatory bowel disease, and metabolic disease. In this review, we provided an updated overview of the current understanding of the connection between diet, microbiota, and health, with a special focus on the role of Western and high-fat diets in shaping intestinal homeostasis by modulating the gut microbiota.


Subject(s)
Ecosystem , Gastrointestinal Microbiome , Diet, High-Fat , Dietary Fiber , Mucus , Sugars
16.
Am J Physiol Gastrointest Liver Physiol ; 300(6): G1080-5, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21454447

ABSTRACT

Irritable bowel syndrome (IBS) is often seen in women, and symptom severity is known to vary over the menstrual cycle. In addition, activation of the hypothalamic-pituitary-adrenal (HPA) axis enhances symptomology and patients with IBS have increased activation of the amygdala, a brain region known to facilitate HPA output. However, little is known about the effects of amygdala activation during different stages of the menstrual cycle. We therefore investigated the effects of amygdala activation on somatic and visceral pain perception over the rat estrous cycle. Female Wistar rats were implanted with either corticosterone (Cort) or cholesterol as a control onto the dorsal margin of the central amygdala. Visceral sensitivity was quantified by recording the visceromotor response (VMR) to colorectal distension (CRD) and somatic sensitivity was assessed via the Von Frey test. In cholesterol controls, both visceral and somatic sensitivity varied over the estrous cycle. Rats in proestrus/estrus responded to CRD with an increased VMR compared with rats in metestrus/diestrus. Somatic sensitivity followed a similar pattern with enhanced sensitivity during proestrus/estrus compared with metestrus/diestrus. Elevated amygdala Cort induced visceral hypersensitivity during metestrus/diestrus but had no effect during proestrus/estrus. In contrast, elevated amygdala Cort increased somatic sensitivity during both metestrus/diestrus and proestrus/estrous. These results suggests that amygdala activation by Cort eliminates spontaneously occurring differences in visceral and somatic pain perception, which could explain the lowered pain thresholds and higher incidence of somatic pain observed in women with IBS.


Subject(s)
Abdominal Pain/physiopathology , Amygdala/drug effects , Colon/innervation , Corticosterone/administration & dosage , Estrous Cycle , Irritable Bowel Syndrome/physiopathology , Amygdala/physiopathology , Analysis of Variance , Animals , Behavior, Animal , Drug Implants , Female , Ovariectomy , Pain Measurement , Pain Perception , Pain Threshold , Pressure , Rats , Rats, Wistar
17.
Elife ; 102021 10 22.
Article in English | MEDLINE | ID: mdl-34677124

ABSTRACT

Intestinal goblet cells maintain the protective epithelial barrier through mucus secretion and yet sample lumenal substances for immune processing through formation of goblet cell associated antigen passages (GAPs). The cellular biology of GAPs and how these divergent processes are balanced and regulated by goblet cells remains unknown. Using high-resolution light and electron microscopy, we found that in mice, GAPs were formed by an acetylcholine (ACh)-dependent endocytic event remarkable for delivery of fluid-phase cargo retrograde into the trans-golgi network and across the cell by transcytosis - in addition to the expected transport of fluid-phase cargo by endosomes to multi-vesicular bodies and lysosomes. While ACh also induced goblet cells to secrete mucins, ACh-induced GAP formation and mucin secretion were functionally independent and mediated by different receptors and signaling pathways, enabling goblet cells to differentially regulate these processes to accommodate the dynamically changing demands of the mucosal environment for barrier maintenance and sampling of lumenal substances.


Cells in the gut need to be protected against the many harmful microbes which inhabit this environment. Yet the immune system also needs to 'keep an eye' on intestinal contents to maintain tolerance to innocuous substances, such as those from the diet. The 'goblet cells' that are part of the gut lining do both: they create a mucus barrier that stops germs from invading the body, but they also can pass on molecules from the intestine to immune cells deep in the tissue to promote tolerance. This is achieved through a 'GAP' mechanism. A chemical messenger called acetylcholine can trigger both mucus release and the GAP process in goblet cells. Gustafsson et al. investigated how the cells could take on these two seemingly opposing roles in response to the same signal. A fluorescent molecule was introduced into the intestines of mice, and monitored as it pass through the goblet cells. This revealed how the GAP process took place: the cells were able to capture molecules from the intestines, wrap them in internal sack-like vesicles and then transport them across the entire cell. To explore the role of acetylcholine, Gustafsson et al. blocked the receptors that detect the messenger at the surface of goblet cells. Different receptors and therefore different cascades of molecular events were found to control mucus secretion and GAP formation; this explains how the two processes can be performed in parallel and independently from each other. Understanding how cells relay molecules to the immune system is relevant to other tissues in contact with the environment, such as the eyes, the airways, or the inside of the genital and urinary tracts. Understanding, and then ultimately harnessing this mechanism could help design of new ways to deliver drugs to the immune system and alter immune outcomes.


Subject(s)
Antigens/metabolism , Goblet Cells/metabolism , Transcytosis , Transport Vesicles/physiology , Animals , Mice
18.
Lab Anim (NY) ; 49(3): 79-88, 2020 03.
Article in English | MEDLINE | ID: mdl-32042160

ABSTRACT

The intestinal immune system samples luminal contents to induce adaptive immune responses that include tolerance in the steady state and protective immunity during infection. How luminal substances are delivered to the immune system has not been fully investigated. Goblet cells have an important role in this process by delivering luminal substances to the immune system through the formation of goblet cell-associated antigen passages (GAPs). Soluble antigens in the intestinal lumen are transported across the epithelium transcellularly through GAPs and delivered to dendritic cells for presentation to T cells and induction of immune responses. GAPs can be identified and quantified by using the ability of GAP-forming goblet cells to take up fluorescently labeled dextran. Here, we describe a method to visualize GAPs and other cells that have the capacity to take up luminal substances by intraluminal injection of fluorescent dextran in mice under anesthesia, tissue sectioning for slide preparation and imaging with fluorescence microscopy. In contrast to in vivo two-photon imaging previously used to identify GAPs, this technique is not limited by anatomical constraints and can be used to visualize GAP formation throughout the length of the intestine. In addition, this method can be combined with common immunohistochemistry protocols to visualize other cell types. This approach can be used to compare GAP formation following different treatments or changes to the luminal environment and to uncover how sampling of luminal substances is altered in pathophysiological conditions. This protocol requires 8 working hours over 2-3 d to be completed.


Subject(s)
Antigens/metabolism , Colon/immunology , Dendritic Cells/immunology , Goblet Cells/immunology , Immunologic Surveillance , Intestine, Small/immunology , Animals , Antigen Presentation/drug effects , Antigens/immunology , Cells, Cultured , Dendritic Cells/drug effects , Dextrans/administration & dosage , Fluorescent Dyes/administration & dosage , Goblet Cells/drug effects , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Microbiota/immunology , Ovalbumin/administration & dosage , Research Design
19.
Mucosal Immunol ; 13(2): 271-282, 2020 03.
Article in English | MEDLINE | ID: mdl-31819172

ABSTRACT

Tolerance to innocuous antigens from the diet and the commensal microbiota is a fundamental process essential to health. Why tolerance is efficiently induced to substances arising from the hostile environment of the gut lumen is incompletely understood but may be related to how these antigens are encountered by the immune system. We observed that goblet cell associated antigen passages (GAPs), but not other pathways of luminal antigen capture, correlated with the acquisition of luminal substances by lamina propria (LP) antigen presenting cells (APCs) and with the sites of tolerance induction to luminal antigens. Strikingly this role extended beyond antigen delivery. The GAP function of goblet cells facilitated maintenance of pre-existing LP T regulatory cells (Tregs), imprinting LP-dendritic cells with tolerogenic properties, and facilitating LP macrophages to produce the immunomodulatory cytokine IL-10. Moreover, tolerance to dietary antigen was impaired in the absence of GAPs. Thus, by delivering luminal antigens, maintaining pre-existing LP Tregs, and imprinting tolerogenic properties on LP-APCs GAPs support tolerance to substances encountered in the hostile environment of the gut lumen.


Subject(s)
Antigen-Presenting Cells/immunology , Dendritic Cells/immunology , Goblet Cells/immunology , Macrophages/immunology , Mucous Membrane/immunology , T-Lymphocytes, Regulatory/immunology , Administration, Oral , Animals , Antigen Presentation , Antigens/immunology , Cells, Cultured , GTPase-Activating Proteins/metabolism , Immune Tolerance , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
20.
JCI Insight ; 5(15)2020 08 06.
Article in English | MEDLINE | ID: mdl-32759496

ABSTRACT

Allergic disorders, characterized by Th2 immune responses to environmental substances, are increasingly common in children in Western societies. Multiple studies indicate that breastfeeding, early complementary introduction of food allergens, and antibiotic avoidance in the first year of life reduces allergic outcomes in at-risk children. Why the benefit of these practices is restricted to early life is largely unknown. We identified a preweaning interval during which dietary antigens are assimilated by the colonic immune system. This interval is under maternal control via temporal changes in breast milk, coincides with an influx of naive T cells into the colon, and is followed by the development of a long-lived population of colonic peripherally derived Tregs (pTregs) that can be specific for dietary antigens encountered during this interval. Desynchronization of mothers and offspring produced durable deficits in these pTregs, impaired tolerance to dietary antigens introduced during and after this preweaning interval, and resulted in spontaneous Th2 responses. These effects could be rescued by pTregs from the periweaning colon or by Tregs generated in vitro using periweaning colonic antigen-presenting cells. These findings demonstrate that mothers and their offspring are synchronized for the development of a balanced immune system.


Subject(s)
Allergens/immunology , Colon/immunology , Food Hypersensitivity/prevention & control , Immune Tolerance/immunology , Milk/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Animals , Animals, Newborn , Antigen-Presenting Cells/immunology , Female , Food Hypersensitivity/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Mothers , Ovalbumin/immunology , Weaning
SELECTION OF CITATIONS
SEARCH DETAIL