Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34548396

ABSTRACT

Elevated levels of MUC5AC, one of the major gel-forming mucins in the lungs, are closely associated with chronic obstructive lung diseases such as chronic bronchitis and asthma. It is not known, however, how the structure and/or gel-making properties of MUC5AC contribute to innate lung defense in health and drive the formation of stagnant mucus in disease. To understand this, here we studied the biophysical properties and macromolecular assembly of MUC5AC compared to MUC5B. To study each native mucin, we used Calu3 monomucin cultures that produced MUC5AC or MUC5B. To understand the macromolecular assembly of MUC5AC through N-terminal oligomerization, we expressed a recombinant whole N-terminal domain (5ACNT). Scanning electron microscopy and atomic force microscopy imaging indicated that the two mucins formed distinct networks on epithelial and experimental surfaces; MUC5B formed linear, infrequently branched multimers, whereas MUC5AC formed tightly organized networks with a high degree of branching. Quartz crystal microbalance-dissipation monitoring experiments indicated that MUC5AC bound significantly more to hydrophobic surfaces and was stiffer and more viscoelastic as compared to MUC5B. Light scattering analysis determined that 5ACNT primarily forms disulfide-linked covalent dimers and higher-order oligomers (i.e., trimers and tetramers). Selective proteolytic digestion of the central glycosylated region of the full-length molecule confirmed that MUC5AC forms dimers and higher-order oligomers through its N terminus. Collectively, the distinct N-terminal organization of MUC5AC may explain the more adhesive and unique viscoelastic properties of branched, highly networked MUC5AC gels. These properties may generate insight into why/how MUC5AC forms a static, "tethered" mucus layer in chronic muco-obstructive lung diseases.


Subject(s)
Epithelial Cells/metabolism , Mucin 5AC/chemistry , Mucin 5AC/metabolism , Mucin-5B/chemistry , Mucin-5B/metabolism , Respiratory Mucosa/metabolism , Cells, Cultured , Epithelial Cells/cytology , Humans , Respiratory Mucosa/cytology
2.
Biochem Biophys Res Commun ; 529(4): 984-990, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32819609

ABSTRACT

OBJECTIVE: To investigate the effects of macromolecular crowding on the folding and aggregation of MUC5AC with different levels of glycosylation during refolding. METHODS: Part 1:An in vitro catalytic reaction comprising the ppGalNAc T2 enzyme, uridine-5'-diphospho-N-galactosamine (UDP-GalNAc) and an 11-amino acid peptide substrate, was used to assess the enzyme activity of the ppGalNAc T2 enzyme in macromolecular crowding environment respectively with bovine serum albumin (BSA), polyethylene glycol (PEG2000), Dextran70 and Ficoll70 at different concentration and temperature. Part 2: The recombinant MUC5AC was expressed in HEK293 cells and purified by nickel column chromatography. The purified protein was treated with PNGase F, and the degree of glycosylation was analyzed by SDS-PAGE. Macromolecular crowding was simulated using PEG2000 at the concentrations of 50, 100, and 200 g/L. Deglycosylated-MUC5AC (d-MUC5AC) and glycosylated MUC5AC (g-MUC5AC) were denatured by GdnHCl and renatured by dilution in a refolding buffer. Protein aggregation was monitored continuously by absorbance reading at 488 nm using a UV spectrophotometer at 25 °C. The refolded proteins were centrifuged, the protein concentration of the supernatant was measured, and refolding yield in different refolding buffers was determined. RESULTS: Enzyme activityof ppGalNAc T2 was observed to increase with increasing crowding agent concentration, with highest enzyme activity at 200 g/L. Compared with the group in the absence of crowding reagent, the refolding yield of g-MUC5AC and d-MUC5AC were reduced significantly in the presence of different concentrations of PEG2000 (200, 100, and 50 g/L). Compared with the dilute solution, aggregation increased significantly in the presence of PEG2000, especially at 200 g/L. Moreover, in the crowded reagent with the same concentration, the refolding yield of d-MUC5AC was higher than that of g-MUC5AC, whereas the degree of aggregation of d-MUC5AC was lower than that of g-MUC5AC. CONCLUSION: The crowded intracellular environment reduces the refolding rate of MUC5AC and strongly induces the misfolding and aggregation of glycosylated MUC5AC.


Subject(s)
Dextrans/pharmacology , Ficoll/pharmacology , Mucin 5AC/metabolism , Polyethylene Glycols/pharmacology , Protein Processing, Post-Translational , Serum Albumin, Bovine/pharmacology , Amino Acid Sequence , Animals , Cattle , Cloning, Molecular , Dextrans/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Ficoll/chemistry , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glycosylation/drug effects , HEK293 Cells , Humans , Kinetics , Mucin 5AC/chemistry , Peptides/chemical synthesis , Peptides/metabolism , Polyethylene Glycols/chemistry , Protein Aggregates/drug effects , Protein Folding/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serum Albumin, Bovine/chemistry , Uridine Diphosphate N-Acetylgalactosamine/analogs & derivatives , Uridine Diphosphate N-Acetylgalactosamine/chemistry , Uridine Diphosphate N-Acetylgalactosamine/metabolism
3.
Biol Pharm Bull ; 43(2): 356-360, 2020.
Article in English | MEDLINE | ID: mdl-32009121

ABSTRACT

Galectins are a group of animal lectins characterized by their specificity for ß-galactosides. Of these, galectin-2 (Gal-2) is predominantly expressed in the gastrointestinal tract. In the current study, we used a mouse gastric mucous fraction to investigate whether Gal-2 is secreted from epithelial cells and identify its potential ligands in gastric mucus. Gal-2 was detected in the mouse gastric mucous fraction and could be eluted from it by the addition of lactose. Affinity chromatography using recombinant mouse galectin-2 (mGal-2)-immobilized adsorbent and subsequent LC-MS/MS identified MUC5AC, one of the major gastric mucin glycoproteins, as a potential ligand of mGal-2. Furthermore, MUC5AC was detected in the mouse gastric mucous fraction by Western blotting, and recombinant mGal-2 was adsorbed to this fraction in a carbohydrate-dependent manner. These results suggested that Gal-2 and MUC5AC in mouse gastric mucus interact in a ß-galactoside-dependent manner, resulting in a stronger barrier structure protecting the mucosal surface.


Subject(s)
Galectin 2/chemistry , Gastrointestinal Tract/chemistry , Mucin 5AC/chemistry , Animals , Humans , Lactose , Mice , Mucus , Stomach
4.
Biomacromolecules ; 20(12): 4332-4344, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31721560

ABSTRACT

Recent research indicates that the progression of Parkinson's disease can start from neurons of the enteric nervous system, which are in close contact with the gastrointestinal epithelium: α-synuclein molecules can be transferred from these epithelial cells in a prion-like fashion to enteric neurons. Thin mucus layers constitute a defense line against the exposure of noninfected cells to potentially harmful α-synuclein species. We show that-despite its mucoadhesive properties-α-synuclein can translocate across mucin hydrogels, and this process is accompanied by structural rearrangements of the mucin molecules within the gel. Penetration experiments with different α-synuclein variants and synthetic peptides suggest that two binding sites on α-synuclein are required to accomplish this rearrangement of the mucin matrix. Our results support the notion that the translocation of α-synuclein across mucus barriers observed here might be a critical step in the infection of the gastrointestinal epithelium and the development of Parkinson's disease.


Subject(s)
Hydrogels/chemistry , Mucin 5AC/chemistry , alpha-Synuclein/chemistry , Animals , Cattle , Gastric Mucosa/chemistry , Gastric Mucosa/metabolism , Humans , Intestinal Mucosa/chemistry , Intestinal Mucosa/metabolism , Mucin 5AC/metabolism , Parkinson Disease/metabolism , Swine , alpha-Synuclein/metabolism
5.
Mol Cell Proteomics ; 16(5): 743-758, 2017 05.
Article in English | MEDLINE | ID: mdl-28461410

ABSTRACT

The mucin O-glycosylation of 10 individuals with and without gastric disease was examined in depth in order to generate a structural map of human gastric glycosylation. In the stomach, these mucins and their O-glycosylation protect the epithelial surface from the acidic gastric juice and provide the first point of interaction for pathogens such as Helicobacter pylori, reported to cause gastritis, gastric and duodenal ulcers and gastric cancer. The rational of the present study was to map the O-glycosylation that the pathogen may come in contact with. An enormous diversity in glycosylation was found, which varied both between individuals and within mucins from a single individual: mucin glycan chain length ranged from 2-13 residues, each individual carried 34-103 O-glycan structures and in total over 258 structures were identified. The majority of gastric O-glycans were neutral and fucosylated. Blood group I antigens, as well as terminal α1,4-GlcNAc-like and GalNAcß1-4GlcNAc-like (LacdiNAc-like), were common modifications of human gastric O-glycans. Furthemore, each individual carried 1-14 glycan structures that were unique for that individual. The diversity and alterations in gastric O-glycosylation broaden our understanding of the human gastric O-glycome and its implications for gastric cancer research and emphasize that the high individual variation makes it difficult to identify gastric cancer specific structures. However, despite the low number of individuals, we could verify a higher level of sialylation and sulfation on gastric O-glycans from cancerous tissue than from healthy stomachs.


Subject(s)
Gastric Mucins/chemistry , Polysaccharides/chemistry , Blood Group Antigens/chemistry , Chromatography, Liquid , Epitopes/metabolism , Gastric Mucins/metabolism , Humans , Mucin 5AC/chemistry , Mucin 5AC/metabolism , Polysaccharides/metabolism , Tandem Mass Spectrometry
6.
Int J Mol Sci ; 20(23)2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31771101

ABSTRACT

Trefoil factor family 2 (TFF2) and the mucin MUC6 are co-secreted from human gastric and duodenal glands. TFF2 binds MUC6 as a lectin and is a constituent of the gastric mucus. Herein, we investigated human gastric extracts by FPLC and identified mainly high- but also low-molecular-mass forms of TFF2. From the high-molecular-mass forms, TFF2 can be completely released by boiling in SDS or by harsh denaturing extraction. The low-molecular-mass form representing monomeric TFF2 can be washed out in part from gastric mucosa specimens with buffer. Overlay assays with radioactively labeled TFF2 revealed binding to the mucin MUC6 and not MUC5AC. This binding is modulated by Ca2+ and can be blocked by the lectin GSA-II and the monoclonal antibody HIK1083. TFF2 binding was also inhibited by Me-ß-Gal, but not the α anomer. Thus, both the α1,4GlcNAc as well as the juxtaperipheral ß-galactoside residues of the characteristic GlcNAcα1→4Galß1→R moiety of human MUC6 are essential for TFF2 binding. Furthermore, there are major differences in the TFF2 binding characteristics when human is compared with the porcine system. Taken together, TFF2 appears to fulfill an important role in stabilizing the inner insoluble gastric mucus barrier layer, particularly by its binding to the mucin MUC6.


Subject(s)
Gastric Mucosa , Mucin-6 , Trefoil Factor-2 , Calcium/chemistry , Calcium/metabolism , Female , Gastric Mucosa/chemistry , Gastric Mucosa/metabolism , Humans , Male , Mucin 5AC/chemistry , Mucin 5AC/metabolism , Mucin-6/chemistry , Mucin-6/metabolism , Trefoil Factor-2/chemistry , Trefoil Factor-2/metabolism
7.
Chemistry ; 23(4): 946-952, 2017 Jan 18.
Article in English | MEDLINE | ID: mdl-27859731

ABSTRACT

The use of native chemical ligation at selenocysteine (Sec) residues with peptide thioesters and additive-free selenocystine ligation with peptides bearing phenyl selenoesters, in concert with one-pot oxidative deselenization chemistry, is described. These approaches provide a simple and rapid method for accessing native peptides with serine in place of Sec at the ligation junction. The efficiency of both variants of the one-pot ligation-oxidative deselenization chemistry is probed through the synthesis of a MUC5AC-derived glycopeptide.


Subject(s)
Cystine/analogs & derivatives , Organoselenium Compounds/chemistry , Selenocysteine/chemistry , Chromatography, High Pressure Liquid , Cystine/chemistry , Glycopeptides/chemical synthesis , Glycopeptides/chemistry , Humans , Mass Spectrometry , Mucin 5AC/chemistry , Oxidation-Reduction
8.
Biomacromolecules ; 18(8): 2454-2462, 2017 Aug 14.
Article in English | MEDLINE | ID: mdl-28635258

ABSTRACT

In the human body, high-molecular-weight glycoproteins called mucins play a key role in protecting epithelial surfaces against pathogenic attack, controlling the passage of molecules toward the tissue and enabling boundary lubrication with very low friction coefficients. However, neither the molecular mechanisms nor the chemical motifs of those biomacromolecules involved in these fundamental processes are fully understood. Thus, identifying the key features that render biomacromolecules such as mucins outstanding boundary lubricants could set the stage for creating versatile artificial superlubricants. We here demonstrate the importance of the hydrophobic terminal peptide domains of porcine gastric mucin (MUC5AC) and human salivary mucin (MUC5B) in the processes of adsorbing to and lubricating a hydrophobic PDMS surface. Tryptic digestion of those mucins results in removal of those terminal domains, which is accompanied by a loss of lubricity as well as surface adsorption. We show that this loss can in part be compensated by attaching hydrophobic phenyl groups to the glycosylated central part of the mucin macromolecule. Furthermore, we demonstrate that the simple biopolysaccharide dextran can be functionalized with hydrophobic groups which confers efficient surface adsorption and good lubricity on PDMS to the polysaccharide.


Subject(s)
Dimethylpolysiloxanes/chemistry , Lubricants/chemistry , Mucin 5AC/chemistry , Animals , Hydrophobic and Hydrophilic Interactions , Protein Domains , Swine
9.
Biomacromolecules ; 18(11): 3654-3664, 2017 Nov 13.
Article in English | MEDLINE | ID: mdl-28903557

ABSTRACT

The details of how a mucus hydrogel forms from its primary structural component, mucin polymers, remain incompletely resolved. To explore this, we use a combination of macrorheology and single-particle tracking to investigate the bulk and microscopic mechanical properties of reconstituted MUC5AC mucin gels. We find that analyses of thermal fluctuations on the length scale of the micrometer-sized particles are not predictive of the linear viscoelastic response of the mucin gels, and that taken together, the results from both techniques help to provide complementary insight into the structure of the network. In particular, we show that macroscopic stiffening of MUC5AC gels can be brought about in different ways by targeting specific associations within the network using environmental triggers such as modifications to the pH, surfactant, and salt concentration. Our work may be important for understanding how environmental factors, including pathogens and therapeutic agents, alter the mechanical properties of fully constituted mucus.


Subject(s)
Hydrogels/chemistry , Mucin 5AC/chemistry , Rheology , Animals , Hydrogen-Ion Concentration , Mucin 5AC/genetics , Particle Size , Polymers/chemistry , Salts/chemistry , Surface-Active Agents/chemistry , Swine , Viscosity
10.
Angew Chem Int Ed Engl ; 54(43): 12716-21, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26384718

ABSTRACT

Despite the unique chemical properties of selenocysteine (Sec), ligation at Sec is an under-utilized methodology for protein synthesis. We describe herein an unprecedented protocol for the conversion of Sec to serine (Ser) in a single, high-yielding step. When coupled with ligation at Sec, this transformation provides a new approach to programmed ligations at Ser residues. This new reaction is compatible with a wide range of functionality, including the presence of unprotected amino acid side chains and appended glycans. The utility of the methodology is demonstrated in the rapid synthesis of complex glycopeptide fragments of the epithelial glycoproteins MUC5AC and MUC4 and through the total synthesis of the structured, cysteine (Cys)-free protein eglin C.


Subject(s)
Cysteine/chemistry , Glycopeptides/chemical synthesis , Selenocysteine/chemistry , Amino Acid Sequence , Animals , Glycopeptides/chemistry , Hirudo medicinalis/chemistry , Humans , Molecular Sequence Data , Mucin 5AC/chemical synthesis , Mucin 5AC/chemistry , Mucin-4/chemical synthesis , Mucin-4/chemistry , Oxidation-Reduction , Proteins/chemical synthesis , Proteins/chemistry
11.
J Proteome Res ; 13(3): 1293-306, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24471499

ABSTRACT

Glycosylation plays an important role in epithelial cancers, including pancreatic ductal adenocarcinoma. However, little is known about the glycoproteome of the human pancreas or its alterations associated with pancreatic tumorigenesis. Using quantitative glycoproteomics approach, we investigated protein N-glycosylation in pancreatic tumor tissue in comparison with normal pancreas and chronic pancreatitis tissue. The study lead to the discovery of a roster of glycoproteins with aberrant N-glycosylation level associated with pancreatic cancer, including mucin-5AC (MUC5AC), carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), insulin-like growth factor binding protein (IGFBP3), and galectin-3-binding protein (LGALS3BP). Pathway analysis of cancer-associated aberrant glycoproteins revealed an emerging phenomenon that increased activity of N-glycosylation was implicated in several pancreatic cancer pathways, including TGF-ß, TNF, NF-kappa-B, and TFEB-related lysosomal changes. In addition, the study provided evidence that specific N-glycosylation sites within certain individual proteins can have significantly altered glycosylation occupancy in pancreatic cancer, reflecting the complexity of the molecular mechanisms underlying cancer-associated glycosylation events.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Gene Expression Regulation, Neoplastic , Glycoproteins/chemistry , Neoplasm Proteins/chemistry , Pancreatic Neoplasms/genetics , Pancreatitis/genetics , Amino Acid Sequence , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoembryonic Antigen/chemistry , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Case-Control Studies , Chronic Disease , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Humans , Insulin-Like Growth Factor Binding Protein 3/chemistry , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Molecular Sequence Data , Mucin 5AC/chemistry , Mucin 5AC/genetics , Mucin 5AC/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatitis/metabolism , Pancreatitis/pathology , Proteomics
12.
Biopolymers ; 101(12): 1154-64, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25041765

ABSTRACT

The structures of purified soluble porcine gastric (Muc5ac) and duodenal (Muc2) mucin solutions at neutral and acidic pH were examined using small-angle X-ray scattering and small-angle neutron scattering experiments. We provide evidence for the morphology of the network above the semidilute overlap concentration and above the entanglement concentration. Furthermore, we investigated the gelation of both types of mucin solutions in response to a reduction in pH, where we observed the formation of large-scale heterogeneities within the polymer solutions, typical of microphase-separated gels. The concentration dependence of the inhomogeneity length scale (Ξ) and the amplitude of the excess scattering intensity [I(ex) (0)] are consistent with previously studied gelled synthetic polymeric systems. The persistence lengths of the chains were found to be similar for both Muc5ac and Muc2 from Kratky plots of the neutron data (8 ± 2 nm).


Subject(s)
Gastrointestinal Tract/metabolism , Mucin 5AC/chemistry , Mucin-2/chemistry , Neutron Diffraction , Scattering, Small Angle , X-Ray Diffraction , Animals , Hydrogen-Ion Concentration , Solubility , Sus scrofa
13.
Int J Biol Macromol ; 267(Pt 2): 131434, 2024 May.
Article in English | MEDLINE | ID: mdl-38614182

ABSTRACT

The gastrointestinal (GI) tract's mucus layer serves as a critical barrier and a mediator in drug nanoparticle delivery. The mucus layer's diverse molecular structures and spatial complexity complicates the mechanistic study of the diffusion dynamics of particulate materials. In response, we developed a bi-component coarse-grained mucus model, specifically tailored for the colorectal cancer environment, that contained the two most abundant glycoproteins in GI mucus: Muc2 and Muc5AC. This model demonstrated the effects of molecular composition and concentration on mucus pore size, a key determinant in the permeability of nanoparticles. Using this computational model, we investigated the diffusion rate of polyethylene glycol (PEG) coated nanoparticles, a widely used muco-penetrating nanoparticle. We validated our model with experimentally characterized mucus pore sizes and the diffusional coefficients of PEG-coated nanoparticles in the mucus collected from cultured human colorectal goblet cells. Machine learning fingerprints were then employed to provide a mechanistic understanding of nanoparticle diffusional behavior. We found that larger nanoparticles tended to be trapped in mucus over longer durations but exhibited more ballistic diffusion over shorter time spans. Through these discoveries, our model provides a promising platform to study pharmacokinetics in the GI mucus layer.


Subject(s)
Mucus , Nanoparticles , Polyethylene Glycols , Humans , Nanoparticles/chemistry , Diffusion , Polyethylene Glycols/chemistry , Mucus/metabolism , Mucus/chemistry , Mucin-2/metabolism , Mucin-2/chemistry , Mucin 5AC/metabolism , Mucin 5AC/chemistry , Intestinal Mucosa/metabolism , Gastrointestinal Tract/metabolism , Goblet Cells/metabolism , Models, Biological
14.
Cancer Sci ; 104(10): 1278-84, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23809433

ABSTRACT

Early and specific diagnosis is critical for treatment of cholangiocarcinoma (CCA). In this study, a carbohydrate antigen-S27 (CA-S27) monoclonal antibody (mAb) was established using pooled CCA tissue-extract as immunogen. The epitope recognized by CA-S27-mAb was a new Lewis-a (Le(a)) associated modification of MUC5AC mucin. A Soybean agglutinin/CA-S27-mAb sandwich ELISA to determine CA-S27 in serum was successfully developed. High level of CA-S27 was detected in serum of CCA patients and could differentiate CCA patients from those of gastro-intestinal cancers, hepatomas, benign hepatobiliary diseases and healthy subjects with high sensitivity (87.5%) and high negative predictive value (90.4%). The level of serum CA-S27 was dramatically reduced after tumor removal, indicating tumor origin of CA-S27. Patients with high serum CA-S27 had significantly shorter survivals than those with low serum CA-S27 regardless of serum MUC5AC levels. Fucosyltransferase-III (FUT3) was shown to be a regulator of CA-S27 expression. Suppression of CA-S27 expression with siRNA-FUT3 or neutralization with CA-S27 mAb significantly reduced growth, adhesion, invasion and migration potentials of CCA cells in vitro. In summary, we demonstrate that serum CA-S27, a novel carbohydrate antigen, has potential as diagnostic and prognostic markers for CCA patients. CA-S27 involves in promoting cell growth, adhesion, migration and invasion of CCA cells.


Subject(s)
Antigens, Neoplasm/immunology , Bile Duct Neoplasms/diagnosis , Bile Ducts, Intrahepatic/immunology , Cholangiocarcinoma/diagnosis , Epitopes/immunology , Mucin 5AC/immunology , Oligosaccharides/immunology , Aged , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/blood , Bile Duct Neoplasms/chemistry , Bile Duct Neoplasms/immunology , Bile Ducts, Intrahepatic/chemistry , Cell Adhesion/physiology , Cholangiocarcinoma/chemistry , Cholangiocarcinoma/immunology , Diagnosis, Differential , Digestive System Neoplasms/diagnosis , Epithelial Cells/chemistry , Epithelial Cells/immunology , Epitopes/blood , Female , Fucosyltransferases/antagonists & inhibitors , Fucosyltransferases/physiology , Humans , Lewis Blood Group Antigens , Liver Diseases/diagnosis , Male , Middle Aged , Molecular Sequence Data , Mucin 5AC/blood , Mucin 5AC/chemistry , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Predictive Value of Tests , RNA Interference , Sensitivity and Specificity , Survival Analysis , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology
15.
Anal Biochem ; 439(2): 204-11, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23643621

ABSTRACT

We have determined the molar proportions of the MUC5AC and MUC6 mucus glycoproteins (mucins) in mucus from the normal and pathological human gastric antrum using a least-squares minimization analysis applied to amino acid compositions. We noted that the content of MUC5AC mucin in mucus from individuals without gastroduodenal disease was very high, suggesting that the integrity and barrier properties of the adherent gastric mucus layer are normally maintained by building-block structures formed from this mucin alone. We observed that the molar content of MUC6 mucin doubled (without significance) in mucus from patients with duodenal ulcer, and increased five times (with high significance) in mucus from patients with gastric ulcer, when compared with that in mucus from individuals without gastroduodenal disease.


Subject(s)
Duodenal Ulcer/metabolism , Mucin 5AC/metabolism , Mucin-6/metabolism , Mucus/chemistry , Stomach Ulcer/metabolism , Adult , Aged , Humans , Middle Aged , Mucin 5AC/chemistry , Mucin 5AC/genetics , Mucin-6/chemistry , Mucin-6/genetics
16.
J Proteome Res ; 11(5): 2904-11, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22439797

ABSTRACT

Significant efforts are underway to develop new biomarkers from pancreatic cyst fluid. Previous research has made use of cyst fluid collected from surgically removed cysts, but the clinical implementation of biomarkers would use cyst fluid collected by endoscopic ultrasound-guided, fine-needle aspiration (EUS-FNA). The purpose of this study was to investigate the clinical applicability of cyst fluid research obtained using surgical specimens. Matched pairs of operating-room collected (OR) and EUS-FNA samples from 12 patients were evaluated for the levels of three previously described biomarkers, CA 19-9, CEA, and glycan levels detected by wheat germ agglutinin on MUC5AC (MUC5AC-WGA). CA 19-9 and MUC5AC-WGA correlated well between the sample types, although CEA was more variable between the sample types for certain patients. The variability was not due to the time delay between EUS-FNA and OR collection or differences in total protein concentrations but may be caused by contamination of the cyst fluid with blood proteins. The classification of each patient based on thresholds for each marker was perfectly consistent between sample types for CA 19-9 and MUC5AC-WGA and mostly consistent for CEA. Therefore, results obtained using OR-collected pancreatic cyst fluid samples should reliably transfer to the clinical setting using EUS-FNA samples.


Subject(s)
Biopsy, Fine-Needle/methods , Cyst Fluid/chemistry , Endosonography/methods , Pancreatic Cyst/chemistry , Biomarkers/chemistry , Blood Proteins/metabolism , CA-19-9 Antigen/chemistry , Humans , Mucin 5AC/chemistry , Operating Rooms/methods , Pancreatic Cyst/pathology , Pancreatic Cyst/surgery , Reproducibility of Results , Specimen Handling
17.
Glycobiology ; 22(3): 429-38, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22042768

ABSTRACT

Mucin-type glycosylation [α-N-acetyl-D-galactosamine (α-GalNAc)-O-Ser/Thr] on proteins is initiated biosynthetically by 16 homologous isoforms of GalNAc-Ts (uridine diphosphate-GalNAc:polypeptide N-acetylgalactosaminyltransferases). All the GalNAc-Ts consist of a catalytic domain and a lectin domain. Previous reports of GalNAc-T assays toward peptides and α-GalNAc glycopeptides showed that the lectin domain recognized the sugar on the substrates and affected the reaction; however, the details are not clear. Here, we report a new strategy to give insight on the sugar recognition ability and the function of the GalNAc-T3 lectin domain using chemically synthesized natural-type (α-GalNAc-O-Thr) and unnatural-type [ß-GalNAc-O-Thr, α-Fuc-O-Thr and ß-GlcNAc-O-Thr] MUC5AC glycopeptides. GalNAc-T3 is one of isoforms expressed in various organs, its substrate specificity extensively characterized and its anomalous expression has been identified in several types of cancer (e.g. pancreas and stomach). The glycopeptides used in this study were designed based on a preliminary peptide assay with a sequence derived from the MUC5AC tandem repeat. Through GalNAc-T3 and lectin-inactivated GalNAc-T3, competition assays between the glycopeptide substrates and product analyses (MALDI-TOF MS, RP-HPLC and ETD-MS/MS), we show that the lectin domain strictly recognized GalNAc on the substrate and this specificity controlled the glycosylation pathway.


Subject(s)
Fucose/chemistry , Galactosamine/chemistry , Glycopeptides/chemistry , Peptides/chemistry , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Cell Line , Chromatography, High Pressure Liquid , Enzyme Assays , Glycopeptides/chemical synthesis , Glycopeptides/isolation & purification , Glycosylation , Humans , Molecular Sequence Data , Mucin 5AC/chemistry , N-Acetylgalactosaminyltransferases , Peptides/chemical synthesis , Peptides/isolation & purification , Protein Binding , Protein Structure, Tertiary , Substrate Specificity , Polypeptide N-acetylgalactosaminyltransferase
18.
Mediators Inflamm ; 2012: 265714, 2012.
Article in English | MEDLINE | ID: mdl-22577246

ABSTRACT

To examine the in vivo effects of the 15-member macrolide, azithromycin (AZM), on mucus hypersecretion, we induced hypertrophic and metaplastic changes of goblet cells in rat nasal epithelium by intranasal instillation of ovalbumin (OVA) in OVA-sensitized rats, or by intranasal lipopolysaccharides (LPS) instillation. Oral administration of AZM (5-10 mg/kg) or clarithromycin (CAM, 5-10 mg/kg) significantly inhibited OVA- and LPS-induced mucus production, whereas josamycin (JM) or ampicillin (ABPC) showed no effect. In vitro effects of AZM on airway epithelial cells were examined using NCI-H292 cells and human nasal epithelial cells cultured in air-liquid interface. Mucus secretion was evaluated by enzyme-linked immunosorbent assay using an anti-MUC5AC monoclonal antibody. AZM or CAM significantly inhibited tumor necrosis factor-α (TNF-α) (20 ng/mL)-induced MUC5AC secretion from NCI-H292 cells at 10⁻6-10⁻7 M, whereas JM or ABPC showed no effect. AZM significantly inhibited TNF-α (20 ng/mL)-induced MUC5AC secretion from human nasal epithelial cells at 10⁻4 M. MUC5AC mRNA expression was also significantly inhibited. These results indicate that the 15-member macrolide, AZM, exerts direct inhibitory effects on mucus secretion from airway epithelial cells and that it may be useful for the treatment of mucus hypersecretion caused by allergic inflammation and LPS stimulation.


Subject(s)
Azithromycin/pharmacology , Bronchi/metabolism , Gene Expression Regulation , Mucus/drug effects , Trachea/metabolism , Ampicillin/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/chemistry , Bronchi/drug effects , Cell Line, Tumor , Clarithromycin/pharmacology , Epithelial Cells/cytology , Goblet Cells/cytology , Humans , Josamycin/pharmacology , Lipopolysaccharides/metabolism , Mucin 5AC/chemistry , Rats , Trachea/drug effects
19.
Chemistry ; 17(8): 2393-404, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21264968

ABSTRACT

Despite the growing importance of mucin core O-glycosylation in many biological processes including the protection of epithelial cell surfaces, the immune response, cell adhesion, inflammation, and tumorigenesis/metastasis, the regulation mechanism and conformational significance of the multiple introduction of α-GalNAc residues by UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferases (ppGalNAcTs) remains unclear. Here we report an efficient approach by combining MS and NMR spectroscopy that allows for the identification of O-glycosylation site(s) and the effect of O-glycosylation on the peptide backbone structures during enzymatic mucin domain assembly by using an isoform UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferase-T2 (ppGalNAcT2) in vitro. An electron-capture dissociation device in a linear radio-frequency quadrupole ion trap (RFQ-ECD) combined with a time-of-flight (TOF) mass spectrometer was employed for the identification of Thr/Ser residues occupied by α-GalNAc branching among multiple and potential O-glycosylation sites in the tandem repeats of human mucin glycoproteins MUC4 (Thr-Ser-Ser-Ala-Ser-Thr-Gly-His-Ala-Thr-Pro-Leu-Pro-Val-Thr-Asp) and MUC5AC (Pro-Thr-Thr-Val-Gly-Ser-Thr-Thr-Val-Gly). In the present study, O-glycosylation was initiated specifically at Thr10 in naked MUC4 peptide and additional introduction of α-GalNAc proceeded preferentially but randomly at three other Thr residues to afford densely glycosylated MUC4 containing six α-GalNAc residues at Thr1, Ser2, Ser5, Thr6, Thr10, and Thr15. On the contrary, O-glycosylation of naked MUC5AC peptide occurred predominantly at consecutive Thr residues and led to MUC5AC with four α-GalNAc residues at Thr2, Thr3, Thr7, and Thr8. The solution structures determined by NMR spectroscopic studies elicited that the preferential introduction of α-GalNAc at Thr10 of MUC4 stabilizes specifically a ß-like extended backbone structure at this area, whereas other synthetic models with a single α-GalNAc residue at Thr1, Thr6, or Thr15 did not exhibit any converged three-dimensional structure at the proximal peptide moiety. Such conformational impact on the underlying peptides was proved to be remarkable in the glycosylation at the consecutive Thr residues of MUC5AC.


Subject(s)
Glycopeptides/chemistry , Mucin 5AC/chemistry , Mucin-4/chemistry , Mucins/chemistry , N-Acetylgalactosaminyltransferases/metabolism , Amino Acid Sequence , Glycopeptides/metabolism , Glycosylation , Humans , Models, Molecular , Mucins/chemical synthesis , Mucins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Serine/chemistry , Threonine/chemistry
20.
J Biomed Biotechnol ; 2011: 934757, 2011.
Article in English | MEDLINE | ID: mdl-21197415

ABSTRACT

A serum ELISA using a monoclonal antibody that detects a MUC5AC-related antigen (NPC-1C antigen) expressed by pancreatic and colorectal cancer was developed. The NPC-1C antibody reacts with specific epitopes expressed by tumor-associated MUC5AC that does not appear on MUC5AC from normal tissues. Based on observations of a highly specific antibody, we tested the ELISA to differentiate serum from healthy blood donors compared to serum from patients with colorectal or pancreatic cancer. Additionally, patient tumor tissue was stained to examine the expression pattern of MUC5AC-related antigen in pancreatic and colorectal cancers. The results indicate the NPC-1C antibody ELISA distinguished serum of cancer patients from normal donors with very good sensitivity and specificity. Most patient's tumor biopsy exhibited NPC-1C antibody reactivity, indicating that tumor-associated MUC5AC antigen from tumor is shed into blood, where it can be detected by the NPC-1C antibody ELISA. This serum test provides a new tool to aid in the diagnosis of these cancers and immune monitoring of cancer treatment regimens.


Subject(s)
Biomarkers, Tumor/blood , Enzyme-Linked Immunosorbent Assay/methods , Mucin 5AC/classification , Antibodies, Monoclonal/chemistry , Colorectal Neoplasms/blood , Humans , Immunohistochemistry , Mucin 5AC/blood , Mucin 5AC/chemistry , Pancreatic Neoplasms/blood
SELECTION OF CITATIONS
SEARCH DETAIL