Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Pflugers Arch ; 2024 Jun 03.
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.

2.
Respir Res ; 24(1): 83, 2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36927357

ABSTRACT

BACKGROUND: The respiratory tract is protected from inhaled particles and microbes by mucociliary clearance, mediated by the mucus and the cilia creating a flow to move the mucus cephalad. Submucosal glands secrete linear MUC5B mucin polymers and because they pass through the gland duct before reaching the airway surface, bundled strands of 1000-5000 parallel molecules exit the glands. In contrast, the surface goblet cells secrete both MUC5AC and MUC5B. METHODS: We used mass-spectrometry based proteomic analysis of unstimulated and carbachol stimulated newborn wild-type (WT) and cystic fibrosis transmembrane conductance regulator (CFTR) null (CF) piglet airways to study proteins in the airway surface liquid and mucus, to investigate if levels of MUC5AC and MUC5B were affected by carbachol stimulation and whether the proteins clustered according to function. RESULTS: Proteins in the first four extracted fractions clustered together and the fifth fraction contained the mucus cluster, mucins and other proteins known to associate with mucins, whereas the traditional airway surface liquid proteins clustered to fraction 1-4 and were absent from the mucus fraction. Carbachol stimulation resulted in increased MUC5AC and MUC5B. CONCLUSIONS: These results indicate a distinct separation between proteins in the washable surface liquid and the mucus fraction. In fractions 1-4 from newborn CF piglets an additional cluster containing acute phase proteins was observed, suggesting an early inflammatory response in CF piglets. Alternatively, increased levels of these proteins could indicate altered lung development in the CF piglets. This observation suggests that CF airway disease is present at birth and thus, treatment should commence directly after diagnosis.


Subject(s)
Cystic Fibrosis , Animals , Swine , Cystic Fibrosis/metabolism , Proteome/metabolism , Carbachol , Proteomics , Mucus/metabolism , Mucins/metabolism , Goblet Cells/metabolism
3.
Am J Respir Crit Care Med ; 206(9): 1081-1095, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35776514

ABSTRACT

Rationale: MUC5AC (mucin 5AC, oligomeric gel-forming) and MUC5B (mucin 5B, oligomeric gel-forming) are the predominant secreted polymeric mucins in mammalian airways. They contribute differently to the pathogenesis of various muco-obstructive and interstitial lung diseases, and their genes are separately regulated, but whether they are packaged together or in separate secretory granules is not known. Objectives: To determine the packaging of MUC5AC and MUC5B within individual secretory granules in mouse and human airways under varying conditions of inflammation and along the proximal-distal axis. Methods: Lung tissue was obtained from mice stimulated to upregulate mucin production by the cytokines IL-1ß and IL-13 or by porcine pancreatic elastase. Human lung tissue was obtained from donated normal lungs, biopsy samples of transplanted lungs, and explanted lungs from subjects with chronic obstructive pulmonary disease. MUC5AC and MUC5B were labeled with antibodies from different animal species or, in mice only, by transgenic chimeric mucin-fluorescent proteins and imaged using widefield deconvolution or Airyscan fluorescence microscopy. Measurements and Main Results: In both mouse and human airways, most secretory granules contained both mucins interdigitating within the granules. Smaller numbers of granules contained MUC5B alone, and even fewer contained MUC5AC alone. Conclusions: MUC5AC and MUC5B are variably stored both in the same and in separate secretory granules of both mice and humans. The high fraction of granules containing both mucins under a variety of conditions makes it unlikely that their secretion can be differentially controlled as a therapeutic strategy. This work also advances knowledge of the packaging of mucins within secretory granules to understand mechanisms of epithelial stress in the pathogenesis of chronic lung diseases.


Subject(s)
Mucin-5B , Pulmonary Disease, Chronic Obstructive , Humans , Mice , Animals , Swine , Mucin 5AC , Lung/metabolism , Secretory Vesicles/metabolism , Mammals/metabolism
4.
Gut ; 70(6): 1117-1129, 2021 06.
Article in English | MEDLINE | ID: mdl-33177165

ABSTRACT

OBJECTIVE: The incidence of IBS increases following enteric infections, suggesting a causative role for microbial imbalance. However, analyses of faecal microbiota have not demonstrated consistent alterations. Here, we used metaproteomics to investigate potential associations between mucus-resident microbiota and IBS symptoms. DESIGN: Mucus samples were prospectively collected from sigmoid colon biopsies from patients with IBS and healthy volunteers, and their microbial protein composition analysed by mass spectrometry. Observations were verified by immunofluorescence, electron microscopy and real-time PCR, further confirmed in a second cohort, and correlated with comprehensive profiling of clinical characteristics and mucosal immune responses. RESULTS: Metaproteomic analysis of colon mucus samples identified peptides from potentially pathogenic Brachyspira species in a subset of patients with IBS. Using multiple diagnostic methods, mucosal Brachyspira colonisation was detected in a total of 19/62 (31%) patients with IBS from two prospective cohorts, versus 0/31 healthy volunteers (p<0.001). The prevalence of Brachyspira colonisation in IBS with diarrhoea (IBS-D) was 40% in both cohorts (p=0.02 and p=0.006 vs controls). Brachyspira attachment to the colonocyte apical membrane was observed in 20% of patients with IBS and associated with accelerated oro-anal transit, mild mucosal inflammation, mast cell activation and alterations of molecular pathways linked to bacterial uptake and ion-fluid homeostasis. Metronidazole treatment paradoxically promoted Brachyspira relocation into goblet cell secretory granules-possibly representing a novel bacterial strategy to evade antibiotics. CONCLUSION: Mucosal Brachyspira colonisation was significantly more common in IBS and associated with distinctive clinical, histological and molecular characteristics. Our observations suggest a role for Brachyspira in the pathogenesis of IBS, particularly IBS-D.


Subject(s)
Bacterial Proteins/analysis , Brachyspira/metabolism , Gram-Negative Bacterial Infections/epidemiology , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/pathology , Mucus/microbiology , Adult , Anti-Bacterial Agents/pharmacology , Biopsy , Brachyspira/drug effects , Brachyspira/isolation & purification , Case-Control Studies , Colon, Sigmoid/pathology , Diarrhea/etiology , Feces/microbiology , Female , Gastrointestinal Transit , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/pathology , Gram-Negative Bacterial Infections/physiopathology , Humans , Immunity, Mucosal , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Irritable Bowel Syndrome/complications , Irritable Bowel Syndrome/physiopathology , Male , Mast Cells , Metronidazole/pharmacology , Middle Aged , Mucus/chemistry , Prevalence , Prospective Studies , Proteomics , Severity of Illness Index , Young Adult
5.
Respir Res ; 22(1): 303, 2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34823518

ABSTRACT

BACKGROUND: The mucociliary clearance system driven by beating cilia protects the airways from inhaled microbes and particles. Large particles are cleared by mucus bundles made in submucosal glands by parallel linear polymers of the MUC5B mucins. However, the structural organization and function of the mucus generated in surface goblet cells are poorly understood. METHODS: The origin and characteristics of different mucus structures were studied on live tissue explants from newborn wild-type (WT), cystic fibrosis transmembrane conductance regulator (CFTR) deficient (CF) piglets and weaned pig airways using video microscopy, Airyscan imaging and electron microscopy. Bronchoscopy was performed in juvenile pigs in vivo. RESULTS: We have identified a distinct mucus formation secreted from the surface goblet cells with a diameter less than two micrometer. This type of mucus was named mucus threads. With time mucus threads gathered into larger mucus assemblies, efficiently collecting particles. The previously observed Alcian blue stained mucus bundles were around 10 times thicker than the threads. Together the mucus bundles, mucus assemblies and mucus threads cleared the pig trachea from particles. CONCLUSIONS: These results demonstrate that normal airway mucus is more complex and has a more variable structural organization and function than was previously understood. These observations emphasize the importance of studying young objects to understand the function of a non-compromised lung.


Subject(s)
Goblet Cells/physiology , Mucociliary Clearance/physiology , Mucus/cytology , Trachea/physiology , Animals , Bronchoscopy , Goblet Cells/cytology , Microscopy, Video , Models, Animal , Swine
6.
Am J Physiol Lung Cell Mol Physiol ; 318(6): L1270-L1279, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32348677

ABSTRACT

The organization of the normal airway mucus system differs in small experimental animals from that in humans and large mammals. To address normal murine airway mucociliary clearance, Alcian blue-stained mucus transport was measured ex vivo on tracheal tissues of naïve C57BL/6, Muc5b-/-, Muc5ac-/-, and EGFP-tagged Muc5b reporter mice. Close to the larynx with a few submucosal glands, the mucus appeared as thick bundles. More distally in the trachea and in large bronchi, Alcian blue-stained mucus was organized in cloud-like formations based on the Muc5b mucin. On tilted tissue, the mucus clouds moved upward toward the larynx with an average velocity of 12 µm/s compared with 20 µm/s for beads not associated with clouds. In Muc5ac-/- mice, Muc5b formed mucus strands attached to the tissue surface, while in Muc5b-/- mice, Muc5ac had a more variable appearance. The normal mouse lung mucus thus appears as discontinuous clouds, clearly different from the stagnant mucus layer in diseased lungs.


Subject(s)
Mucin-5B/metabolism , Mucus/metabolism , Respiratory System/metabolism , Animals , Biological Transport , Fluorescence , Goblet Cells/metabolism , Mice, Inbred C57BL , Mucin 5AC/metabolism , Mucous Membrane/metabolism , Trachea/metabolism
7.
J Biol Chem ; 293(15): 5746-5754, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29440393

ABSTRACT

Most MUC5B mucin polymers in the upper airways of humans and pigs are produced by submucosal glands. MUC5B forms N-terminal covalent dimers that are further packed into larger assemblies because of low pH and high Ca2+ in the secretory granule of the mucin-producing cell. We purified the recombinant MUC5B N-terminal covalent dimer and used single-particle electron microscopy to study its structure under intracellular conditions. We found that, at intragranular pH, the dimeric MUC5B organized into head-to-head noncovalent tetramers where the von Willebrand D1-D2 domains hooked into each other. These N-terminal tetramers further formed long linear complexes from which, we suggest, the mucin domains and their C termini project radially outwards. Using conventional and video microscopy, we observed that, upon secretion into the submucosal gland ducts, a flow of bicarbonate-rich fluid passes the mucin-secreting cells. We suggest that this unfolds and pulls out the MUC5B assemblies into long linear threads. These further assemble into thicker mucin bundles in the glandular ducts before emerging at the gland duct opening. We conclude that the combination of intracellular packing of the MUC5B mucin and the submucosal gland morphology creates an efficient machine for producing linear mucin bundles.


Subject(s)
Calcium/chemistry , Mucin-5B/chemistry , Protein Multimerization , Animals , Calcium/metabolism , Humans , Hydrogen-Ion Concentration , Mucin-5B/genetics , Mucin-5B/metabolism , Protein Domains , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Swine
8.
Biochem J ; 475(3): 691-704, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29335300

ABSTRACT

In the present study, we have applied ratiometric measurements of intracellular Ca2+ concentrations ([Ca2+]i) to show that extracellularly applied ATP (adenosine triphosphate) (100 µM) stimulates store-operated Ca2+ entry (SOCE) in 3T3-L1 adipocytes. ATP produced a rapid increase in [Ca2+]i consisting of an initial transient elevation followed by a sustained elevated phase that could be observed only in the presence of extracellular Ca2+ Gene expression data and [Ca2+]i recordings with uridine-5'-triphosphate or with the phospholipase C (PLC) inhibitor U73122 demonstrated the involvement of purinergic P2Y2 receptors and the PLC/inositol trisphosphate pathway. The [Ca2+]i elevation produced by reintroduction of a Ca2+-containing intracellular solution to adipocytes exposed to ATP in the absence of Ca2+ was diminished by known SOCE antagonists. The chief molecular components of SOCE, the stromal interaction molecule 1 (STIM1) and the calcium release-activated calcium channel protein 1 (ORAI1), were detected at the mRNA and protein level. Moreover, SOCE was largely diminished in cells where STIM1 and/or ORAI1 had been silenced by small interfering (si)RNA. We conclude that extracellular ATP activates SOCE in white adipocytes, an effect predominantly mediated by STIM1 and ORAI1.


Subject(s)
Adenosine Triphosphate/metabolism , Adipocytes, White/metabolism , ORAI1 Protein/genetics , Stromal Interaction Molecule 1/genetics , 3T3-L1 Cells , Animals , Calcium/metabolism , Calcium Signaling/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Multiprotein Complexes/genetics , ORAI1 Protein/metabolism , RNA, Small Interfering/genetics , Stromal Interaction Molecule 1/metabolism , TRPC Cation Channels/genetics
9.
Proc Natl Acad Sci U S A ; 113(48): 13833-13838, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27849619

ABSTRACT

The distal colon functions as a bioreactor and harbors an enormous amount of bacteria in a mutualistic relationship with the host. The microbiota have to be kept at a safe distance to prevent inflammation, something that is achieved by a dense inner mucus layer that lines the epithelial cells. The large polymeric nets made up by the heavily O-glycosylated MUC2 mucin forms this physical barrier. Proteomic analyses of mucus have identified the lectin-like protein ZG16 (zymogen granulae protein 16) as an abundant mucus component. To elucidate the function of ZG16, we generated recombinant ZG16 and studied Zg16-/- mice. ZG16 bound to and aggregated Gram-positive bacteria via binding to the bacterial cell wall peptidoglycan. Zg16-/- mice have a distal colon mucus layer with normal thickness, but with bacteria closer to the epithelium. Using distal colon explants mounted in a horizontal perfusion chamber we demonstrated that treatment of bacteria with recombinant ZG16 hindered bacterial penetration into the mucus. The inner colon mucus of Zg16-/- animals had a higher load of Gram-positive bacteria and showed bacteria with higher motility in the mucus close to the host epithelium compared with cohoused littermate Zg16+/+ The more penetrable Zg16-/- mucus allowed Gram-positive bacteria to translocate to systemic tissues. Viable bacteria were found in spleen and were associated with increased abdominal fat pad mass in Zg16-/- animals. The function of ZG16 reveals a mechanism for keeping bacteria further away from the host colon epithelium.


Subject(s)
Gram-Positive Bacteria/genetics , Lectins/genetics , Membrane Proteins/genetics , Proteomics , Animals , Colon/metabolism , Colon/microbiology , Digestive System/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Glycosylation , Gram-Positive Bacteria/metabolism , Host-Pathogen Interactions/genetics , Lectins/metabolism , Mice , Mice, Knockout , Mucus/metabolism , Mucus/microbiology , Symbiosis/genetics
10.
Eur Respir J ; 52(2)2018 08.
Article in English | MEDLINE | ID: mdl-29853489

ABSTRACT

The beneficial effect of anticholinergic therapy for chronic lung diseases such as chronic obstructive pulmonary disease (COPD) is well documented, although cholinergic stimulation paradoxically inhibits liquid absorption, increases ciliary beat frequency and increases airway surface liquid transport.Using pig tracheobronchial explants, we quantified basal mucus transport before as well as after incubation with the clinically used antimuscarinic compound ipratropium bromide (Atrovent) and stimulation with acetylcholine.As expected, surface liquid transport was increased by acetylcholine and carbachol. In contrast, the mucus bundles secreted from the submucosal glands normally transported on the cilia were stopped from moving by acetylcholine, an effect inhibited by ipratropium bromide. Interestingly, in pigs lacking a functional cystic fibrosis (CF) transmembrane conductance regulator (CFTR) channel, the mucus bundles were almost immobile. As in wild-type pigs, CF surface liquid transport increased after carbachol stimulation. The stagnant CF mucus bundles were trapped on the tracheal surface attached to the surface goblet cells. Pseudomonas aeruginosa bacteria were moved by the mucus bundles in wild-type but not CF pigs.Acetylcholine thus uncouples airway surface liquid transport from transport of the surface mucus bundles as the bundles are dynamically inhibited by acetylcholine and the CFTR channel, explaining initiation of CF and COPD, and opening novel therapeutic windows.


Subject(s)
Cholinergic Agents/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/drug therapy , Mucociliary Clearance , Mucus/metabolism , Animals , Cystic Fibrosis/metabolism , Disease Models, Animal , Epithelial Cells/drug effects , Humans , Pseudomonas aeruginosa/isolation & purification , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Swine
11.
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
12.
Biochem Biophys Res Commun ; 492(3): 331-337, 2017 10 21.
Article in English | MEDLINE | ID: mdl-28859985

ABSTRACT

To understand the mucociliary clearance system, mucins were visualized by light, confocal and electron microscopy, and mucus was stained by Alcian blue and tracked by video microscopy on tracheal explants of newborn piglets. We observed long linear mucus bundles that appeared at the submucosal gland openings and were transported cephalically. The mucus bundles were shown by mass spectrometry and immunostaining to have a core made of MUC5B mucin and were coated with MUC5AC mucin produced by surface goblet cells. The transport speed of the bundles was slower than the airway surface liquid flow. We suggest that the goblet cell MUC5AC mucin anchors the mucus bundles and thus controls their transport. Normal clearance of the respiratory tree of pigs and humans, both rich in submucosal glands, is performed by thick and long mucus bundles.


Subject(s)
Exocrine Glands/metabolism , Mucin 5AC/metabolism , Mucin-5B/metabolism , Mucociliary Clearance , Respiratory Mucosa/metabolism , Trachea/metabolism , Animals , Swine
13.
EMBO Rep ; 16(2): 164-77, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25525071

ABSTRACT

Two C57BL/6 mice colonies maintained in two rooms of the same specific pathogen-free (SPF) facility were found to have different gut microbiota and a mucus phenotype that was specific for each colony. The thickness and growth of the colon mucus were similar in the two colonies. However, one colony had mucus that was impenetrable to bacteria or beads the size of bacteria-which is comparable to what we observed in free-living wild mice-whereas the other colony had an inner mucus layer penetrable to bacteria and beads. The different properties of the mucus depended on the microbiota, as they were transmissible by transfer of caecal microbiota to germ-free mice. Mice with an impenetrable mucus layer had increased amounts of Erysipelotrichi, whereas mice with a penetrable mucus layer had higher levels of Proteobacteria and TM7 bacteria in the distal colon mucus. Thus, our study shows that bacteria and their community structure affect mucus barrier properties in ways that can have implications for health and disease. It also highlights that genetically identical animals housed in the same facility can have rather distinct microbiotas and barrier structures.


Subject(s)
Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Animals , Female , Mice , Mice, Inbred C57BL , Microbiota/physiology , Mucus/cytology , Mucus/microbiology , RNA, Ribosomal, 16S/genetics , Specific Pathogen-Free Organisms
14.
Clin Exp Pharmacol Physiol ; 44(6): 639-647, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28261854

ABSTRACT

The goal of this study was to determine whether the guluronate (G) rich alginate OligoG CF-5/20 (OligoG) could detach cystic fibrosis (CF) mucus by calcium chelation, which is also required for normal mucin unfolding. Since bicarbonate secretion is impaired in CF, leading to insufficient mucin unfolding and thereby attached mucus, and since bicarbonate has the ability to bind calcium, we hypothesized that the calcium chelating property of OligoG would lead to detachment of CF mucus. Indeed, OligoG could compete with the N-terminus of the MUC2 mucin for calcium binding as shown by microscale thermophoresis. Further, effects on mucus thickness and attachment induced by OligoG and other alginate fractions of different length and composition were evaluated in explants of CF mouse ileum mounted in horizontal Ussing-type chambers. OligoG at 1.5% caused effective detachment of CF mucus and the most potent alginate fraction tested, the poly-G fraction of about 12 residues, had similar potency compared to OligoG whereas mannuronate-rich (M) polymers had minimal effect. In conclusion, OligoG binds calcium with appropriate affinity without any overt harmful effect on the tissue and can be exploited for treating mucus stagnation.


Subject(s)
Alginates/chemistry , Alginates/pharmacology , Calcium/metabolism , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Mucus/drug effects , Mucus/metabolism , Alginates/metabolism , Alginates/therapeutic use , Animals , Chelating Agents/chemistry , Chelating Agents/metabolism , Chelating Agents/pharmacology , Chelating Agents/therapeutic use , Glucuronic Acid/chemistry , Glucuronic Acid/metabolism , Glucuronic Acid/pharmacology , Glucuronic Acid/therapeutic use , Hexuronic Acids/chemistry , Hexuronic Acids/metabolism , Hexuronic Acids/pharmacology , Hexuronic Acids/therapeutic use , Ileum/drug effects , Ileum/metabolism , Mice , Polymerization
15.
Proc Natl Acad Sci U S A ; 111(34): 12396-401, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25114233

ABSTRACT

The mucus that covers and protects the epithelium of the intestine is built around its major structural component, the gel-forming MUC2 mucin. The gel-forming mucins have traditionally been assumed to be secreted as nonattached. The colon has a two-layered mucus system where the inner mucus is attached to the epithelium, whereas the small intestine normally has a nonattached mucus. However, the mucus of the small intestine of meprin ß-deficient mice was now found to be attached. Meprin ß is an endogenous zinc-dependent metalloprotease now shown to cleave the N-terminal region of the MUC2 mucin at two specific sites. When recombinant meprin ß was added to the attached mucus of meprin ß-deficient mice, the mucus was detached from the epithelium. Similar to meprin ß-deficient mice, germ-free mice have attached mucus as they did not shed the membrane-anchored meprin ß into the luminal mucus. The ileal mucus of cystic fibrosis (CF) mice with a nonfunctional cystic fibrosis transmembrane conductance regulator (CFTR) channel was recently shown to be attached to the epithelium. Addition of recombinant meprin ß to CF mucus did not release the mucus, but further addition of bicarbonate rendered the CF mucus normal, suggesting that MUC2 unfolding exposed the meprin ß cleavage sites. Mucus is thus secreted attached to the goblet cells and requires an enzyme, meprin ß in the small intestine, to be detached and released into the intestinal lumen. This process regulates mucus properties, can be triggered by bacterial contact, and is nonfunctional in CF due to poor mucin unfolding.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Intestine, Small/metabolism , Metalloendopeptidases/metabolism , Mucin-2/metabolism , Mucus/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Germ-Free Life/physiology , Intestine, Small/microbiology , Metalloendopeptidases/deficiency , Metalloendopeptidases/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CFTR , Mice, Knockout , Molecular Sequence Data , Mucin-2/chemistry , Mucin-2/genetics , Protein Folding , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
16.
Clin Exp Pharmacol Physiol ; 42(1): 69-75, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25311799

ABSTRACT

Hypertonic saline inhalation has become a cornerstone in the treatment of cystic fibrosis (CF), but its effect on CF mucus is still not understood. In CF, mucus stagnates in the airways, causing mucus plugging, and forming a substrate for bacterial invasion. Using horizontal Ussing-type chambers to allow easy access to the tissue, we have recently shown that the small intestinal mucus of CF mice is attached to the epithelium and not freely movable as opposed to normal mucus, thus pointing to a similarity between the CF mucus in the ileum and airways. In the same type of system, we investigated how hypertonic saline affects mucus thickness, attachment and penetrability to fluorescent beads the size of bacteria in ileal explants from the cystic fibrosis transmembrane conductance regulator mutant (ΔF508) mouse, in order to characterize how this common therapy affects mucus properties. Hypertonic saline (1.75-5%) detached the mucus from the epithelium, but the mucus remained impenetrable to beads the size of bacteria. This approach might be used to test other mucolytic interventions in CF.


Subject(s)
Cystic Fibrosis/drug therapy , Intestine, Small/drug effects , Mucus/drug effects , Saline Solution, Hypertonic/therapeutic use , Animals , Cystic Fibrosis/pathology , Female , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucus/metabolism , Organ Culture Techniques , Saline Solution, Hypertonic/pharmacology
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL