Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.305
Filter
Add more filters

Publication year range
1.
Cell ; 185(22): 4206-4215.e11, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36206754

ABSTRACT

Mucus protects the epithelial cells of the digestive and respiratory tracts from pathogens and other hazards. Progress in determining the molecular mechanisms of mucus barrier function has been limited by the lack of high-resolution structural information on mucins, the giant, secreted, gel-forming glycoproteins that are the major constituents of mucus. Here, we report how mucin structures we determined enabled the discovery of an unanticipated protective role of mucus: managing the toxic transition metal copper. Using two juxtaposed copper binding sites, one for Cu2+ and the other for Cu1+, the intestinal mucin, MUC2, prevents copper toxicity by blocking futile redox cycling and the squandering of dietary antioxidants, while nevertheless permitting uptake of this important trace metal into cells. These findings emphasize the value of molecular structure in advancing mucosal biology, while introducing mucins, produced in massive quantities to guard extensive mucosal surfaces, as extracellular copper chaperones.


Subject(s)
Copper , Mucins , Mucins/metabolism , Mucin-2 , Copper/analysis , Copper/metabolism , Intestines , Mucus/metabolism , Intestinal Mucosa/metabolism
2.
Cell ; 185(22): 4190-4205.e25, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36243004

ABSTRACT

Neuroepithelial crosstalk is critical for gut physiology. However, the mechanisms by which sensory neurons communicate with epithelial cells to mediate gut barrier protection at homeostasis and during inflammation are not well understood. Here, we find that Nav1.8+CGRP+ nociceptor neurons are juxtaposed with and signal to intestinal goblet cells to drive mucus secretion and gut protection. Nociceptor ablation led to decreased mucus thickness and dysbiosis, while chemogenetic nociceptor activation or capsaicin treatment induced mucus growth. Mouse and human goblet cells expressed Ramp1, receptor for the neuropeptide CGRP. Nociceptors signal via the CGRP-Ramp1 pathway to induce rapid goblet cell emptying and mucus secretion. Notably, commensal microbes activated nociceptors to control homeostatic CGRP release. In the absence of nociceptors or epithelial Ramp1, mice showed increased epithelial stress and susceptibility to colitis. Conversely, CGRP administration protected nociceptor-ablated mice against colitis. Our findings demonstrate a neuron-goblet cell axis that orchestrates gut mucosal barrier protection.


Subject(s)
Colitis , Goblet Cells , Mice , Humans , Animals , Goblet Cells/metabolism , Nociceptors/metabolism , Calcitonin Gene-Related Peptide/metabolism , Colitis/metabolism , Mucus/metabolism , Receptor Activity-Modifying Protein 1/metabolism
3.
Cell ; 185(7): 1172-1188.e28, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35303419

ABSTRACT

Intestinal mucus forms the first line of defense against bacterial invasion while providing nutrition to support microbial symbiosis. How the host controls mucus barrier integrity and commensalism is unclear. We show that terminal sialylation of glycans on intestinal mucus by ST6GALNAC1 (ST6), the dominant sialyltransferase specifically expressed in goblet cells and induced by microbial pathogen-associated molecular patterns, is essential for mucus integrity and protecting against excessive bacterial proteolytic degradation. Glycoproteomic profiling and biochemical analysis of ST6 mutations identified in patients show that decreased sialylation causes defective mucus proteins and congenital inflammatory bowel disease (IBD). Mice harboring a patient ST6 mutation have compromised mucus barriers, dysbiosis, and susceptibility to intestinal inflammation. Based on our understanding of the ST6 regulatory network, we show that treatment with sialylated mucin or a Foxo3 inhibitor can ameliorate IBD.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Sialyltransferases/genetics , Animals , Homeostasis , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Mucus/metabolism , Sialyltransferases/metabolism , Symbiosis
4.
Cell ; 185(20): 3705-3719.e14, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36179667

ABSTRACT

The intestinal microbiota is an important modulator of graft-versus-host disease (GVHD), which often complicates allogeneic hematopoietic stem cell transplantation (allo-HSCT). Broad-spectrum antibiotics such as carbapenems increase the risk for intestinal GVHD, but mechanisms are not well understood. In this study, we found that treatment with meropenem, a commonly used carbapenem, aggravates colonic GVHD in mice via the expansion of Bacteroides thetaiotaomicron (BT). BT has a broad ability to degrade dietary polysaccharides and host mucin glycans. BT in meropenem-treated allogeneic mice demonstrated upregulated expression of enzymes involved in the degradation of mucin glycans. These mice also had thinning of the colonic mucus layer and decreased levels of xylose in colonic luminal contents. Interestingly, oral xylose supplementation significantly prevented thinning of the colonic mucus layer in meropenem-treated mice. Specific nutritional supplementation strategies, including xylose supplementation, may combat antibiotic-mediated microbiome injury to reduce the risk for intestinal GVHD in allo-HSCT patients.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteroides , Carbapenems/pharmacology , Carbapenems/therapeutic use , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Meropenem , Mice , Mucins/metabolism , Mucus/metabolism , Polysaccharides/metabolism , Xylose
5.
Annu Rev Biochem ; 89: 769-793, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32243763

ABSTRACT

Generating the barriers that protect our inner surfaces from bacteria and other challenges requires large glycoproteins called mucins. These come in two types, gel-forming and transmembrane, all characterized by large, highly O-glycosylated mucin domains that are diversely decorated by Golgi glycosyltransferases to become extended rodlike structures. The general functions of mucins on internal epithelial surfaces are to wash away microorganisms and, even more importantly, to build protective barriers. The latter function is most evident in the large intestine, where the inner mucus layer separates the numerous commensal bacteria from the epithelial cells. The host's conversion of MUC2 to the outer mucus layer allows bacteria to degrade the mucin glycans and recover the energy content that is then shared with the host. The molecular nature of the mucins is complex, and how they construct the extracellular complex glycocalyx and mucus is poorly understood and a future biochemical challenge.


Subject(s)
Gastrointestinal Microbiome/physiology , Glycocalyx/chemistry , Glycosyltransferases/chemistry , Goblet Cells/chemistry , Mucins/chemistry , Mucus/chemistry , Animals , Carbohydrate Conformation , Carbohydrate Sequence , Gene Expression , Glycocalyx/metabolism , Glycosylation , Glycosyltransferases/classification , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Goblet Cells/metabolism , Goblet Cells/microbiology , Humans , Mucins/classification , Mucins/genetics , Mucins/metabolism , Mucus/metabolism , Mucus/microbiology , Symbiosis/physiology
6.
Annu Rev Cell Dev Biol ; 34: 189-215, 2018 10 06.
Article in English | MEDLINE | ID: mdl-30296390

ABSTRACT

We review what is currently understood about how the structure of the primary solid component of mucus, the glycoprotein mucin, gives rise to the mechanical and biochemical properties of mucus that are required for it to perform its diverse physiological roles. Macroscale processes such as lubrication require mucus of a certain stiffness and spinnability, which are set by structural features of the mucin network, including the identity and density of cross-links and the degree of glycosylation. At the microscale, these same features affect the mechanical environment experienced by small particles and play a crucial role in establishing an interaction-based filter. Finally, mucin glycans are critical for regulating microbial interactions, serving as receptor binding sites for adhesion, as nutrient sources, and as environmental signals. We conclude by discussing how these structural principles can be used in the design of synthetic mucin-mimetic materials and provide suggestions for directions of future work in this field.


Subject(s)
Glycoproteins/chemistry , Mucin-1/chemistry , Mucus/chemistry , Structure-Activity Relationship , Animals , Glycoproteins/genetics , Glycosylation , Humans , Mucin-1/genetics , Mucus/metabolism , Permeability , Rheology
7.
Annu Rev Biochem ; 84: 947-67, 2015.
Article in English | MEDLINE | ID: mdl-25534639

ABSTRACT

Hagfishes thwart attacks by fish predators by producing liters of defensive slime. The slime is produced when slime gland exudate is released into the predator's mouth, where it deploys in a fraction of a second and clogs the gills. Slime exudate is composed mainly of secretory products from two cell types, gland mucous cells and gland thread cells, which produce the mucous and fibrous components of the slime, respectively. Here, we review what is known about the composition of the slime, morphology of the slime gland, and physiology of the cells that produce the slime. We also discuss several of the mechanisms involved in the deployment of both mucous and thread cells during the transition from thick glandular exudate to ultradilute material. We review biomechanical aspects of the slime, along with recent efforts to produce biomimetic slime thread analogs, and end with a discussion of how hagfish slime may have evolved.


Subject(s)
Hagfishes/chemistry , Hagfishes/physiology , Mucus/metabolism , Animals , Biomimetics , Exocrine Glands/cytology , Exocrine Glands/metabolism , Hagfishes/cytology , Mucus/chemistry
8.
Physiol Rev ; 102(4): 1757-1836, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35001665

ABSTRACT

The mucus clearance system is the dominant mechanical host defense system of the human lung. Mucus is cleared from the lung by cilia and airflow, including both two-phase gas-liquid pumping and cough-dependent mechanisms, and mucus transport rates are heavily dependent on mucus concentration. Importantly, mucus transport rates are accurately predicted by the gel-on-brush model of the mucociliary apparatus from the relative osmotic moduli of the mucus and periciliary-glycocalyceal (PCL-G) layers. The fluid available to hydrate mucus is generated by transepithelial fluid transport. Feedback interactions between mucus concentrations and cilia beating, via purinergic signaling, coordinate Na+ absorptive vs Cl- secretory rates to maintain mucus hydration in health. In disease, mucus becomes hyperconcentrated (dehydrated). Multiple mechanisms derange the ion transport pathways that normally hydrate mucus in muco-obstructive lung diseases, e.g., cystic fibrosis (CF), chronic obstructive pulmonary disease (COPD), non-CF bronchiectasis (NCFB), and primary ciliary dyskinesia (PCD). A key step in muco-obstructive disease pathogenesis is the osmotic compression of the mucus layer onto the airway surface with the formation of adherent mucus plaques and plugs, particularly in distal airways. Mucus plaques create locally hypoxic conditions and produce airflow obstruction, inflammation, infection, and, ultimately, airway wall damage. Therapies to clear adherent mucus with hydrating and mucolytic agents are rational, and strategies to develop these agents are reviewed.


Subject(s)
Cystic Fibrosis , Pulmonary Disease, Chronic Obstructive , Cystic Fibrosis/metabolism , Humans , Lung/metabolism , Mucociliary Clearance , Mucus/metabolism
9.
Cell ; 156(5): 1045-59, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24581500

ABSTRACT

Mucus production by goblet cells of the large intestine serves as a crucial antimicrobial protective mechanism at the interface between the eukaryotic and prokaryotic cells of the mammalian intestinal ecosystem. However, the regulatory pathways involved in goblet cell-induced mucus secretion remain largely unknown. Here, we demonstrate that the NLRP6 inflammasome, a recently described regulator of colonic microbiota composition and biogeographical distribution, is a critical orchestrator of goblet cell mucin granule exocytosis. NLRP6 deficiency leads to defective autophagy in goblet cells and abrogated mucus secretion into the large intestinal lumen. Consequently, NLRP6 inflammasome-deficient mice are unable to clear enteric pathogens from the mucosal surface, rendering them highly susceptible to persistent infection. This study identifies an innate immune regulatory pathway governing goblet cell mucus secretion, linking nonhematopoietic inflammasome signaling to autophagy and highlighting the goblet cell as a critical innate immune player in the control of intestinal host-microbial mutualism. PAPERCLIP:


Subject(s)
Colon/immunology , Goblet Cells/immunology , Inflammasomes/immunology , Intestinal Mucosa/immunology , Receptors, Cell Surface/immunology , Animals , Autophagy , Colitis/immunology , Colitis/microbiology , Colon/microbiology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Goblet Cells/cytology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Mucus/metabolism
10.
Semin Immunol ; 69: 101807, 2023 09.
Article in English | MEDLINE | ID: mdl-37478802

ABSTRACT

A complex mucus network made up of large polymers of the mucin-family glycoprotein MUC2 exists between the large intestinal microbial mass and epithelial and immune cells. This has long been understood as an innate immune defense barrier against the microbiota and other luminal threats that reinforces the barrier function of the epithelium and limits microbiota contact with the tissues. However, past and recent studies have provided new evidence of how critical the mucus network is to act as a 'liaison' between host and microbe to mediate anti-inflammatory, mutualistic interactions with the microbiota and protection from pathogens. This review summarizes historical and recent insights into the formation of the gut mucus network, how the microbes and immune system influence mucus, and in turn, how the mucus influences immune responses to the microbiota.


Subject(s)
Intestinal Mucosa , Microbiota , Humans , Mucin-2 , Mucus , Mucins
11.
PLoS Biol ; 21(8): e3002209, 2023 08.
Article in English | MEDLINE | ID: mdl-37527210

ABSTRACT

The opportunistic pathogen Pseudomonas aeruginosa causes antibiotic-recalcitrant pneumonia by forming biofilms in the respiratory tract. Despite extensive in vitro experimentation, how P. aeruginosa forms biofilms at the airway mucosa is unresolved. To investigate the process of biofilm formation in realistic conditions, we developed AirGels: 3D, optically accessible tissue-engineered human lung models that emulate the airway mucosal environment. AirGels recapitulate important factors that mediate host-pathogen interactions including mucus secretion, flow and air-liquid interface (ALI), while accommodating high-resolution live microscopy. With AirGels, we investigated the contributions of mucus to P. aeruginosa biofilm biogenesis in in vivo-like conditions. We found that P. aeruginosa forms mucus-associated biofilms within hours by contracting luminal mucus early during colonization. Mucus contractions facilitate aggregation, thereby nucleating biofilms. We show that P. aeruginosa actively contracts mucus using retractile filaments called type IV pili. Our results therefore suggest that, while protecting epithelia, mucus constitutes a breeding ground for biofilms.


Subject(s)
Biofilms , Pseudomonas aeruginosa , Humans , Anti-Bacterial Agents/pharmacology , Mucus , Lung
12.
Nature ; 583(7814): 78-82, 2020 07.
Article in English | MEDLINE | ID: mdl-32494011

ABSTRACT

Many animals build complex structures to aid in their survival, but very few are built exclusively from materials that animals create 1,2. In the midwaters of the ocean, mucoid structures are readily secreted by numerous animals, and serve many vital functions3,4. However, little is known about these mucoid structures owing to the challenges of observing them in the deep sea. Among these mucoid forms, the 'houses' of larvaceans are marvels of nature5, and in the ocean twilight zone giant larvaceans secrete and build mucus filtering structures that can reach diameters of more than 1 m6. Here we describe in situ laser-imaging technology7 that reconstructs three-dimensional models of mucus forms. The models provide high-resolution views of giant larvacean houses and elucidate the role that house structure has in food capture and predator avoidance. Now that tools exist to study mucus structures found throughout the ocean, we can shed light on some of nature's most complex forms.


Subject(s)
Aquatic Organisms/metabolism , Mucus/metabolism , Urochordata/anatomy & histology , Urochordata/metabolism , Animals , Carbon Cycle , Feeding Behavior , Food Chain , Imaging, Three-Dimensional/instrumentation , Lasers , Molecular Conformation , Mucus/chemistry , Oceans and Seas , Predatory Behavior , Seawater
13.
PLoS Genet ; 19(6): e1010445, 2023 06.
Article in English | MEDLINE | ID: mdl-37352370

ABSTRACT

Hyper-secretion and/or hyper-concentration of mucus is a defining feature of multiple obstructive lung diseases, including chronic obstructive pulmonary disease (COPD). Mucus itself is composed of a mixture of water, ions, salt and proteins, of which the gel-forming mucins, MUC5AC and MUC5B, are the most abundant. Recent studies have linked the concentrations of these proteins in sputum to COPD phenotypes, including chronic bronchitis (CB) and acute exacerbations (AE). We sought to determine whether common genetic variants influence sputum mucin concentrations and whether these variants are also associated with COPD phenotypes, specifically CB and AE. We performed a GWAS to identify quantitative trait loci for sputum mucin protein concentration (pQTL) in the Sub-Populations and InteRmediate Outcome Measures in COPD Study (SPIROMICS, n = 708 for total mucin, n = 215 for MUC5AC, MUC5B). Subsequently, we tested for associations of mucin pQTL with CB and AE using regression modeling (n = 822-1300). Replication analysis was conducted using data from COPDGene (n = 5740) and by examining results from the UK Biobank. We identified one genome-wide significant pQTL for MUC5AC (rs75401036) and two for MUC5B (rs140324259, rs10001928). The strongest association for MUC5B, with rs140324259 on chromosome 11, explained 14% of variation in sputum MUC5B. Despite being associated with lower MUC5B, the C allele of rs140324259 conferred increased risk of CB (odds ratio (OR) = 1.42; 95% confidence interval (CI): 1.10-1.80) as well as AE ascertained over three years of follow up (OR = 1.41; 95% CI: 1.02-1.94). Associations between rs140324259 and CB or AE did not replicate in COPDGene. However, in the UK Biobank, rs140324259 was associated with phenotypes that define CB, namely chronic mucus production and cough, again with the C allele conferring increased risk. We conclude that sputum MUC5AC and MUC5B concentrations are associated with common genetic variants, and the top locus for MUC5B may influence COPD phenotypes, in particular CB.


Subject(s)
Mucins , Pulmonary Disease, Chronic Obstructive , Humans , Mucins/genetics , Mucins/metabolism , Sputum/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Mucus/metabolism , Phenotype
14.
J Biol Chem ; 300(3): 105675, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38272223

ABSTRACT

The O-glycoprotein Mucin-2 (MUC2) forms the protective colon mucus layer. While animal models have demonstrated the importance of Muc2, few studies have explored human MUC2 in similar depth. Recent studies have revealed that secreted MUC2 is bound to human feces. We hypothesized human fecal MUC2 (HF-MUC2) was accessible for purification and downstream structural and functional characterization. We tested this via histologic and quantitative imaging on human fecal sections; extraction from feces for proteomic and O-glycomic characterization; and functional studies via growth and metabolic assays in vitro. Quantitative imaging of solid fecal sections showed a continuous mucus layer of varying thickness along human fecal sections with barrier functions intact. Lectin profiling showed HF-MUC2 bound several lectins but was weak to absent for Ulex europaeus 1 (α1,2 fucose-binding) and Sambucus nigra agglutinin (α2,6 sialic acid-binding), and did not have obvious b1/b2 barrier layers. HF-MUC2 separated by electrophoresis showed high molecular weight glycoprotein bands (∼1-2 MDa). Proteomics and Western analysis confirmed the enrichment of MUC2 and potential MUC2-associated proteins in HF-MUC2 extracts. MUC2 O-glycomics revealed diverse fucosylation, moderate sialylation, and little sulfation versus porcine colonic MUC2 and murine fecal Muc2. O-glycans were functional and supported the growth of Bacteroides thetaiotaomicron (B. theta) and short-chain fatty acid (SCFA) production in vitro. MUC2 could be similarly analyzed from inflammatory bowel disease stools, which displayed an altered glycomic profile and differential growth and SCFA production by B. theta versus healthy samples. These studies describe a new non-invasive platform for human MUC2 characterization in health and disease.


Subject(s)
Colon , Feces , Proteomics , Animals , Humans , Mice , Colon/metabolism , Glycoproteins/metabolism , Intestinal Mucosa/metabolism , Mucin-2/genetics , Mucin-2/metabolism , Mucus/metabolism , Swine , Male , Mice, Inbred C57BL , Gastrointestinal Microbiome
15.
Eur J Immunol ; 54(1): e2350558, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37855177

ABSTRACT

Airway epithelial cells contribute to a variety of lung diseases including allergic asthma, where IL-4 and IL-13 promote activation of the transcription factor STAT6. This leads to goblet cell hyperplasia and the secretion of effector molecules by epithelial cells. However, the specific effect of activated STAT6 in lung epithelial cells is only partially understood. Here, we created a mouse strain to selectively investigate the role of constitutively active STAT6 in Club cells, a subpopulation of airway epithelial cells. CCSP-Cre_STAT6vt mice and bronchiolar organoids derived from these show an enhanced expression of the chitinase-like protein Chil4 (Ym2) and resistin-like molecules (Relm-α, -ß, -γ). In addition, goblet cells of these mice spontaneously secrete mucus into the bronchi. However, the activated epithelium resulted neither in impaired lung function nor conferred a protective effect against the migrating helminth Nippostrongylus brasiliensis. Moreover, CCSP-Cre_STAT6vt mice showed similar allergic airway inflammation induced by live conidia of the fungus Aspergillus fumigatus and similar recovery after influenza A virus infection compared to control mice. Together these results highlight that STAT6 signaling in Club cells induces the secretion of Relm proteins and mucus without impairing lung function, but this is not sufficient to confer protection against helminth or viral infections.


Subject(s)
Asthma , Resistin , Animals , Mice , Asthma/metabolism , Epithelial Cells/metabolism , Lung , Mucus/metabolism , Resistin/metabolism , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism
16.
Nucleic Acids Res ; 51(10): 5193-5209, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37070602

ABSTRACT

The long non-coding RNA EPR is expressed in epithelial tissues, binds to chromatin and controls distinct biological activities in mouse mammary gland cells. Because of its high expression in the intestine, in this study we have generated a colon-specific conditional targeted deletion (EPR cKO) to evaluate EPR in vivo functions in mice. EPR cKO mice display epithelium hyperproliferation, impaired mucus production and secretion, as well as inflammatory infiltration in the proximal portion of the large intestine. RNA sequencing analysis reveals a rearrangement of the colon crypt transcriptome with strong reduction of goblet cell-specific factors including those involved in the synthesis, assembly, transport and control of mucus proteins. Further, colon mucosa integrity and permeability are impaired in EPR cKO mice, and this results in higher susceptibility to dextran sodium sulfate (DSS)-induced colitis and tumor formation. Human EPR is down-regulated in human cancer cell lines as well as in human cancers, and overexpression of EPR in a colon cancer cell line results in enhanced expression of pro-apoptotic genes. Mechanistically, we show that EPR directly interacts with select genes involved in mucus metabolism whose expression is reduced in EPR cKO mice and that EPR deletion causes tridimensional chromatin organization changes.


Subject(s)
Cell Transformation, Neoplastic , Inflammation , Mucus , RNA, Long Noncoding , Animals , Humans , Mice , Cell Transformation, Neoplastic/immunology , Colon/metabolism , Disease Models, Animal , Inflammation/immunology , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
17.
Proc Natl Acad Sci U S A ; 119(13): e2121731119, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35324331

ABSTRACT

SignificanceIn many lung diseases, increased amounts of and/or abnormal mucus impair mucociliary clearance, a key defense against inhaled and aspirated material. Submucosal glands lining cartilaginous airways secrete mucus strands that are pulled by cilia until they break free from the duct and sweep upward toward the larynx, carrying particulates. In cystic fibrosis (CF) pigs, progressive clearance of insufflated microdisks was repeatedly interrupted as microdisks abruptly recoiled. Aerosolizing a reducing agent to break disulfide bonds linking mucins ruptured mucus strands, freeing them from submucosal gland ducts and allowing cilia to propel them up the airways. These findings highlight the abnormally increased elasticity of CF mucus and suggest that agents that break disulfide bonds might have value in lung diseases with increased mucus.


Subject(s)
Cystic Fibrosis , Mucociliary Clearance , Animals , Disulfides , Mucus , Respiratory Mucosa , Swine
18.
Proc Natl Acad Sci U S A ; 119(27): e2116197119, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35767643

ABSTRACT

The majority of viruses within the gut are obligate bacterial viruses known as bacteriophages (phages). Their bacteriotropism underscores the study of phage ecology in the gut, where they modulate and coevolve with gut bacterial communities. Traditionally, these ecological and evolutionary questions were investigated empirically via in vitro experimental evolution and, more recently, in vivo models were adopted to account for physiologically relevant conditions of the gut. Here, we probed beyond conventional phage-bacteria coevolution to investigate potential tripartite evolutionary interactions between phages, their bacterial hosts, and the mammalian gut mucosa. To capture the role of the mammalian gut, we recapitulated a life-like gut mucosal layer using in vitro lab-on-a-chip devices (to wit, the gut-on-a-chip) and showed that the mucosal environment supports stable phage-bacteria coexistence. Next, we experimentally coevolved lytic phage populations within the gut-on-a-chip devices alongside their bacterial hosts. We found that while phages adapt to the mucosal environment via de novo mutations, genetic recombination was the key evolutionary force in driving mutational fitness. A single mutation in the phage capsid protein Hoc-known to facilitate phage adherence to mucus-caused altered phage binding to fucosylated mucin glycans. We demonstrated that the altered glycan-binding phenotype provided the evolved mutant phage a competitive fitness advantage over its ancestral wild-type phage in the gut-on-a-chip mucosal environment. Collectively, our findings revealed that phages-in addition to their evolutionary relationship with bacteria-are able to evolve in response to a mammalian-derived mucosal environment.


Subject(s)
Bacteria , Bacteriophages , Gastrointestinal Tract , Mucous Membrane , Animals , Bacteria/virology , Bacteriophages/genetics , Bacteriophages/physiology , Capsid Proteins/genetics , Gastrointestinal Tract/virology , Mucous Membrane/virology , Mucus , Mutation , Symbiosis
19.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35046051

ABSTRACT

Submucosal glands (SMGs) protect lungs but can also contribute to disease. For example, in cystic fibrosis (CF), SMGs produce abnormal mucus that disrupts mucociliary transport. CF is an ion transport disease, yet knowledge of the ion transporters expressed by SMG acini, which produce mucus, and SMG ducts that carry it to the airway lumen is limited. Therefore, we isolated SMGs from newborn pigs and used single-cell messenger RNA sequencing, immunohistochemistry, and in situ hybridization to identify cell types, gene expression, and spatial distribution. Cell types and transcript levels were the same in non-CF and CF SMGs, suggesting that loss of epithelial anion secretion rather than an intrinsic cell defect causes CF mucus abnormalities. Gene signatures of acinar mucous and acinar serous cells revealed specialized functions in producing mucins and antimicrobials, respectively. However, surprisingly, these two cell types expressed the same ion transporters and neurohumoral receptors, suggesting the importance of balancing mucin and liquid secretion to produce optimal mucus properties. SMG duct cell transcripts suggest that they secrete HCO3- and Cl-, and thus have some similarity to pancreatic ducts that are also defective in CF. These and additional findings suggest the functions of the SMG acinus and duct and provide a baseline for understanding how environmental and genetic challenges impact their contribution to lung disease.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Mutation , Respiratory Mucosa/metabolism , Acinar Cells/metabolism , Animals , Biomarkers , Cystic Fibrosis/etiology , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression , Gene Knockdown Techniques , Genetic Predisposition to Disease , Mucins/metabolism , Mucociliary Clearance , Mucus/metabolism , Respiratory Mucosa/pathology , Swine
20.
J Allergy Clin Immunol ; 153(5): 1306-1318, 2024 May.
Article in English | MEDLINE | ID: mdl-38181841

ABSTRACT

BACKGROUND: Airway obstruction caused by viscous mucus is an important pathophysiologic characteristic of persistent inflammation, which can result in organ damage. OBJECTIVE: We investigated the hypothesis that the biophysical characteristics of accumulating granulocytes affect the clinical properties of mucus. METHODS: Surgically acquired nasal mucus samples from patients with eosinophilic chronic rhinosinusitis and neutrophil-dominant, noneosinophilic chronic rhinosinusitis were evaluated in terms of computed tomography density, viscosity, water content, wettability, and protein composition. Isolated human eosinophils and neutrophils were stimulated to induce the formation of extracellular traps, followed by the formation of aggregates. The biophysical properties of the aggregated cells were also examined. RESULTS: Mucus from patients with eosinophilic chronic rhinosinusitis had significantly higher computed tomography density, viscosity, dry weight, and hydrophobicity compared to mucus from patients with noneosinophilic chronic rhinosinusitis. The levels of eosinophil-specific proteins in mucus correlated with its physical properties. Eosinophil and neutrophil aggregates showed physical and pathologic characteristics resembling those of mucus. Cotreatment with deoxyribonuclease and heparin, which slenderizes the structure of eosinophil extracellular traps, efficiently induced reductions in the viscosity and hydrophobicity of both eosinophil aggregates and eosinophilic mucus. CONCLUSIONS: The present study elucidated the pathogenesis of mucus stasis in infiltrated granulocyte aggregates from a novel perspective. These findings may contribute to the development of treatment strategies for eosinophilic airway diseases.


Subject(s)
Eosinophils , Extracellular Traps , Mucus , Neutrophils , Rhinitis , Sinusitis , Humans , Sinusitis/immunology , Sinusitis/pathology , Rhinitis/immunology , Rhinitis/pathology , Eosinophils/immunology , Chronic Disease , Neutrophils/immunology , Mucus/metabolism , Male , Female , Adult , Extracellular Traps/immunology , Extracellular Traps/metabolism , Middle Aged , Viscosity , Cell Aggregation , Aged , Nasal Mucosa/immunology , Nasal Mucosa/pathology , Rhinosinusitis
SELECTION OF CITATIONS
SEARCH DETAIL