Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
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.
J Biol Chem ; 294(45): 17105-17116, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31570524

ABSTRACT

Mucin 5B (MUC5B) has an essential role in mucociliary clearance that protects the pulmonary airways. Accordingly, knowledge of MUC5B structure and its interactions with itself and other proteins is critical to better understand airway mucus biology and improve the management of lung diseases such as asthma, cystic fibrosis, and chronic obstructive pulmonary disease (COPD). The role of an N-terminal multimerization domain in the supramolecular organization of MUC5B has been previously described, but less is known about its C-terminal dimerization domain. Here, using cryogenic electron microscopy (cryo-EM) and small-angle X-ray scattering (SAXS) analyses of recombinant disulfide-linked dimeric MUC5B dimerization domain we identified an asymmetric, elongated twisted structure, with a double globular base. We found that the dimerization domain is more resistant to disruption than the multimerization domain suggesting the twisted structure of the dimerization domain confers additional stability to MUC5B polymers. Size-exclusion chromatography-multiangle light scattering (SEC-MALS), SPR-based biophysical analyses and microscale thermophoresis of the dimerization domain disclosed no further assembly, but did reveal reversible, calcium-dependent interactions between the dimerization and multimerization domains that were most active at acidic pH, suggesting that these domains have a role in MUC5B intragranular organization. In summary, our results suggest a role for the C-terminal dimerization domain of MUC5B in compaction of mucin chains during granular packaging via interactions with the N-terminal multimerization domain. Our findings further suggest that the less stable multimerization domain provides a potential target for mucin depolymerization to remove mucus plugs in COPD and other lung pathologies.


Subject(s)
Intracellular Space/metabolism , Mucin-5B/chemistry , Mucin-5B/metabolism , Protein Multimerization , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Models, Molecular , Protein Domains , Protein Stability , Protein Structure, Quaternary
3.
Am J Physiol Lung Cell Mol Physiol ; 317(4): L498-L509, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31389736

ABSTRACT

Muco-obstructive lung diseases (MOLDs), like cystic fibrosis and chronic obstructive pulmonary disease, affect a spectrum of subjects globally. In MOLDs, the airway mucus becomes hyperconcentrated, increasing osmotic and viscoelastic moduli and impairing mucus clearance. MOLD research requires relevant sources of healthy airway mucus for experimental manipulation and analysis. Mucus collected from endotracheal tubes (ETT) may represent such a source with benefits, e.g., in vivo production, over canonical sample types such as sputum or human bronchial epithelial (HBE) mucus. Ionic and biochemical compositions of ETT mucus from healthy human subjects were characterized and a stock of pooled ETT samples generated. Pooled ETT mucus exhibited concentration-dependent rheologic properties that agreed across spatial scales with reported individual ETT samples and HBE mucus. We suggest that the practical benefits compared with other sample types make ETT mucus potentially useful for MOLD research.


Subject(s)
Mucus/chemistry , Potassium/analysis , Rheology/methods , Sodium/analysis , Trachea/chemistry , Adult , Aged , Aged, 80 and over , Cations, Monovalent , Female , Healthy Volunteers , Humans , Intubation, Intratracheal , Male , Middle Aged , Polysaccharides/classification , Polysaccharides/isolation & purification , Potassium/metabolism , Proteins/classification , Proteins/isolation & purification , Sodium/metabolism , Sputum/chemistry , Trachea/physiology
4.
Am J Respir Crit Care Med ; 199(6): 715-727, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30352166

ABSTRACT

RATIONALE: MUC5AC and MUC5B are the predominant gel-forming mucins in the mucus layer of human airways. Each mucin has distinct functions and site-specific expression. However, the regional distribution of expression and cell types that secrete each mucin in normal/healthy human airways are not fully understood. OBJECTIVES: To characterize the regional distribution of MUC5B and MUC5AC in normal/healthy human airways and assess which cell types produce these mucins, referenced to the club cell secretory protein (CCSP). METHODS: Multiple airway regions from 16 nonsmoker lungs without a history of lung disease were studied. MUC5AC, MUC5B, and CCSP expression/colocalization were assessed by RNA in situ hybridization and immunohistochemistry in five lungs with histologically healthy airways. Droplet digital PCR and cell cultures were performed for absolute quantification of MUC5AC/5B ratios and protein secretion, respectively. MEASUREMENTS AND MAIN RESULTS: Submucosal glands expressed MUC5B, but not MUC5AC. However, MUC5B was also extensively expressed in superficial epithelia throughout the airways except for the terminal bronchioles. Morphometric calculations revealed that the distal airway superficial epithelium was the predominant site for MUC5B expression, whereas MUC5AC expression was concentrated in proximal, cartilaginous airways. RNA in situ hybridization revealed MUC5AC and MUC5B were colocalized with CCSP-positive secretory cells in proximal superficial epithelia, whereas MUC5B and CCSP-copositive cells dominated distal regions. CONCLUSIONS: In normal/healthy human airways, MUC5B is the dominant secretory mucin in the superficial epithelium and glands, with distal airways being a major site of expression. MUC5B and MUC5AC expression is a property of CCSP-positive secretory cells in superficial airway epithelia.


Subject(s)
Lung/diagnostic imaging , Lung/physiology , Mucin 5AC/analysis , Mucin-5B/analysis , Protein Transport/physiology , Respiratory Physiological Phenomena , Humans
6.
J Biol Chem ; 288(20): 14636-14646, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23546879

ABSTRACT

The colonic epithelial surface is protected by an inner mucus layer that the commensal microflora cannot penetrate. We previously demonstrated that Entamoeba histolytica secretes a protease capable of dissolving this layer that is required for parasite penetration. Here, we asked whether there are bacteria that can secrete similar proteases. We screened bacterial culture supernatants for such activity using recombinant fragments of the MUC2 mucin, the major structural component, and the only gel-forming mucin in the colonic mucus. MUC2 has two central heavily O-glycosylated mucin domains that are protease-resistant and has cysteine-rich N and C termini responsible for polymerization. Culture supernatants of Porphyromonas gingivalis, a bacterium that secretes proteases responsible for periodontitis, cleaved the MUC2 C-terminal region, whereas the N-terminal region was unaffected. The active enzyme was isolated and identified as Arg-gingipain B (RgpB). Two cleavage sites were localized to IR↓TT and NR↓QA. IR↓TT cleavage will disrupt the MUC2 polymers. Because this site has two potential O-glycosylation sites, we tested whether recombinant GalNAc-transferases (GalNAc-Ts) could glycosylate a synthetic peptide covering the IRTT sequence. Only GalNAc-T3 was able to glycosylate the second Thr in IRTT, rendering the sequence resistant to cleavage by RgpB. Furthermore, when GalNAc-T3 was expressed in CHO cells expressing the MUC2 C terminus, the second threonine was glycosylated, and the protein became resistant to RgpB cleavage. These findings suggest that bacteria can produce proteases capable of dissolving the inner protective mucus layer by specific cleavages in the MUC2 mucin and that this cleavage can be modulated by site-specific O-glycosylation.


Subject(s)
Adhesins, Bacterial/metabolism , Cysteine Endopeptidases/metabolism , Mucin-2/metabolism , Porphyromonas gingivalis/enzymology , Amino Acid Sequence , Animals , CHO Cells , Chromatography , Colitis/microbiology , Colon/metabolism , Cricetinae , Epithelium/metabolism , Gingipain Cysteine Endopeptidases , Glycosylation , Humans , Mass Spectrometry , Molecular Sequence Data
7.
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
8.
J Immunol ; 182(1): 216-24, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109152

ABSTRACT

With a 5-year survival rate of <5%, pancreatic cancer is one of the most rapidly fatal malignancies. Current protocols for the treatment of pancreas cancer are not as effective as we desire. In this study, we show that a novel Mucin-1 (MUC1)-based vaccine in combination with a cyclooxygenase-2 inhibitor (celecoxib), and low-dose chemotherapy (gemcitabine) was effective in preventing the progression of preneoplastic intraepithelial lesions to invasive pancreatic ductal adenocarcinomas. The study was conducted in an appropriate triple transgenic model of spontaneous pancreatic cancer induced by the KRAS(G12D) mutation and that expresses human MUC1 as a self molecule. The combination treatment elicited robust antitumor cellular and humoral immune responses and was associated with increased apoptosis in the tumor. The mechanism for the increased immune response was attributed to the down-regulation of circulating prostaglandin E(2) and indoleamine 2, 3,-dioxygenase enzymatic activity, as well as decreased levels of T regulatory and myeloid suppressor cells within the tumor microenvironment. The preclinical data provide the rationale to design clinical trials with a combination of MUC1-based vaccine, celecoxib, and gemcitabine for the treatment of pancreatic cancer.


Subject(s)
Adenocarcinoma/enzymology , Adenocarcinoma/prevention & control , Cancer Vaccines/administration & dosage , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/prevention & control , Cyclooxygenase 2 Inhibitors/administration & dosage , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/prevention & control , Adenocarcinoma/pathology , Animals , Antibodies/blood , Cancer Vaccines/immunology , Carcinoma, Pancreatic Ductal/pathology , Celecoxib , Cyclooxygenase 2/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease Progression , Drug Evaluation, Preclinical , Drug Therapy, Combination , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mice , Mice, Transgenic , Mucin-1/administration & dosage , Mucin-1/immunology , Pancreatic Neoplasms/pathology , Pyrazoles/administration & dosage , Sulfonamides/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Gemcitabine
9.
J Immunol ; 181(5): 3116-25, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18713982

ABSTRACT

MUC1, a membrane tethered mucin glycoprotein, is overexpressed and aberrantly glycosylated in >80% of human ductal pancreatic adenocarcinoma. However, the role of MUC1 in pancreatic cancer has been elusive, partly due to the lack of an appropriate model. We report the characterization of a novel mouse model that expresses human MUC1 as a self molecule (PDA.MUC1 mice). Pancreatic tumors arise in an appropriate MUC1-tolerant background within an immune-competent host. Significant enhancement in the development of pancreatic intraepithelial preneoplastic lesions and progression to adenocarcinoma is observed in PDA.MUC1 mice, possibly due to increased proliferation. Tumors from PDA.MUC1 mice express higher levels of cyclooxygenase-2 and IDO compared with PDA mice lacking MUC1, especially during early stages of tumor development. The increased proinflammatory milieu correlates with an increased percentage of regulatory T cells and myeloid suppressor cells in the pancreatic tumor and tumor draining lymph nodes. Data shows that during pancreatic cancer progression, MUC1-mediated mechanisms enhance the onset and progression of the disease, which in turn regulate the immune responses. Thus, the mouse model is ideally suited for testing novel chemopreventive and therapeutic strategies against pancreatic cancer.


Subject(s)
Adenocarcinoma , Immune Tolerance , Mucin-1/physiology , Pancreatic Neoplasms/pathology , Animals , Cyclooxygenase 2/genetics , Disease Progression , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Mice , Mice, Transgenic , Models, Animal , Mucin-1/genetics , Pancreatic Neoplasms/immunology , Tumor Escape , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...