Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
iScience ; 27(6): 110093, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38947523

RESUMEN

A diet lacking dietary fibers promotes the expansion of gut microbiota members that can degrade host glycans, such as those on mucins. The microbial foraging on mucin has been associated with disruptions of the gut-protective mucus layer and colonic inflammation. Yet, it remains unclear how the co-utilization of mucin and dietary fibers affects the microbiota composition and metabolic activity. Here, we used 14 dietary fibers and porcine colonic and gastric mucins to study the dynamics of mucin and dietary fiber utilization by the human fecal microbiota in vitro. Combining metaproteome and metabolites analyses revealed the central role of the Bacteroides genus in the utilization of complex fibers together with mucin while Akkermansia muciniphila was the main utilizer of sole porcine colonic mucin but not gastric mucin. This study gives a broad overview of the colonic environment in response to dietary and host glycan availability.

2.
bioRxiv ; 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38405862

RESUMEN

Crohn's disease (CD) is the chronic inflammation of the ileum and colon triggered by bacteria, but insights into molecular perturbations at the bacteria-epithelium interface are limited. We report that membrane mucin MUC17 protects small intestinal enterocytes against commensal and pathogenic bacteria. In non-inflamed CD ileum, reduced MUC17 levels correlated with a compromised glycocalyx, allowing bacterial contact with enterocytes. Muc17 deletion in mice rendered the small intestine prone to atypical infection while maintaining resistance to colitis. The loss of Muc17 resulted in spontaneous deterioration of epithelial homeostasis and extra-intestinal translocation of bacteria. Finally, Muc17-deficient mice harbored specific small intestinal bacterial taxa observed in CD. Our findings highlight MUC17 as an essential line of defense in the small intestine with relevance for early epithelial defects in CD.

3.
Nat Commun ; 14(1): 3652, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37339972

RESUMEN

A key feature in intestinal immunity is the dynamic intestinal barrier, which separates the host from resident and pathogenic microbiota through a mucus gel impregnated with antimicrobial peptides. Using a forward genetic screen, we have found a mutation in Tvp23b, which conferred susceptibility to chemically induced and infectious colitis. Trans-Golgi apparatus membrane protein TVP23 homolog B (TVP23B) is a transmembrane protein conserved from yeast to humans. We found that TVP23B controls the homeostasis of Paneth cells and function of goblet cells, leading to a decrease in antimicrobial peptides and more penetrable mucus layer. TVP23B binds with another Golgi protein, YIPF6, which is similarly critical for intestinal homeostasis. The Golgi proteomes of YIPF6 and TVP23B-deficient colonocytes have a common deficiency of several critical glycosylation enzymes. TVP23B is necessary for the formation of the sterile mucin layer of the intestine and its absence disturbs the balance of host and microbe in vivo.


Asunto(s)
Mucosa Intestinal , Intestinos , Proteínas de la Membrana , Animales , Ratones , Microbioma Gastrointestinal , Glicosilación , Células Caliciformes/metabolismo , Aparato de Golgi/metabolismo , Homeostasis , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Intestinos/metabolismo , Proteínas de la Membrana/metabolismo , Moco , Células de Paneth/metabolismo
4.
Cell Rep ; 42(2): 112084, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36753416

RESUMEN

Intestinal mucus barriers normally prevent microbial infections but are sensitive to diet-dependent changes in the luminal environment. Here we demonstrate that mice fed a Western-style diet (WSD) suffer regiospecific failure of the mucus barrier in the small intestinal jejunum caused by diet-induced mucus aggregation. Mucus barrier disruption due to either WSD exposure or chromosomal Muc2 deletion results in collapse of the commensal jejunal microbiota, which in turn sensitizes mice to atypical jejunal colonization by the enteric pathogen Citrobacter rodentium. We illustrate the jejunal mucus layer as a microbial habitat, and link the regiospecific mucus dependency of the microbiota to distinctive properties of the jejunal niche. Together, our data demonstrate a symbiotic mucus-microbiota relationship that normally prevents jejunal pathogen colonization, but is highly sensitive to disruption by exposure to a WSD.


Asunto(s)
Mucosa Intestinal , Yeyuno , Mucina 2 , Animales , Ratones , Dieta Occidental , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Intestino Delgado , Mucina 2/genética , Mucina 2/metabolismo , Moco , Citrobacter rodentium/fisiología
5.
Nat Commun ; 13(1): 45, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35017479

RESUMEN

The colonic mucus layer is organized as a two-layered system providing a physical barrier against pathogens and simultaneously harboring the commensal flora. The factors contributing to the organization of this gel network are not well understood. In this study, the impact of transglutaminase activity on this architecture was analyzed. Here, we show that transglutaminase TGM3 is the major transglutaminase-isoform expressed and synthesized in the colon. Furthermore, intrinsic extracellular transglutaminase activity in the secreted mucus was demonstrated in vitro and ex vivo. Absence of this acyl-transferase activity resulted in faster degradation of the major mucus component the MUC2 mucin and changed the biochemical properties of mucus. Finally, TGM3-deficient mice showed an early increased susceptibility to Dextran Sodium Sulfate-induced colitis. Here, we report that natural isopeptide cross-linking by TGM3 is important for mucus homeostasis and protection of the colon from inflammation, reducing the risk of colitis.


Asunto(s)
Colon/metabolismo , Moco/metabolismo , Transglutaminasas/metabolismo , Animales , Colitis/etiología , Colitis/metabolismo , Ratones , Mucina 2/metabolismo
6.
Science ; 372(6539)2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33859001

RESUMEN

The intestinal mucus layer, an important element of epithelial protection, is produced by goblet cells. Intestinal goblet cells are assumed to be a homogeneous cell type. In this study, however, we delineated their specific gene and protein expression profiles and identified several distinct goblet cell populations that form two differentiation trajectories. One distinct subtype, the intercrypt goblet cells (icGCs), located at the colonic luminal surface, produced mucus with properties that differed from the mucus secreted by crypt-residing goblet cells. Mice with defective icGCs had increased sensitivity to chemically induced colitis and manifested spontaneous colitis with age. Furthermore, alterations in mucus and reduced numbers of icGCs were observed in patients with both active and remissive ulcerative colitis, which highlights the importance of icGCs in maintaining functional protection of the epithelium.


Asunto(s)
Colon/citología , Células Caliciformes/fisiología , Mucosa Intestinal/citología , Moco/fisiología , Animales , Diferenciación Celular , Colitis/inducido químicamente , Colitis/fisiopatología , Colitis Ulcerosa/patología , Colitis Ulcerosa/fisiopatología , Colon/fisiología , Células Caliciformes/citología , Humanos , Mucosa Intestinal/fisiología , Intestino Delgado/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-ets/genética , Transcriptoma
7.
Methods Mol Biol ; 2259: 167-179, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33687714

RESUMEN

Metaproteomics of host-microbiome interfaces comprises the analysis of complex mixtures of bacteria, archaea, fungi, and viruses in combination with its host cells. Microbial niches can be found all over the host including the skin, oral cavity, and the intestine and are considered to be essential for the homeostasis. The complex interactions between the host and diverse commensal microbiota are poorly characterized while of great interest as dysbiosis is associated with the development of various inflammatory and metabolic diseases. The metaproteomics workflows to study these interfaces are currently being established, and many challenges remain. The major challenge is the large diversity in species composition that make up the microbiota, which results in complex samples that require extended mass spectrometry analysis time. In addition, current database search strategies are not developed to the size of the search space required for unbiased microbial protein identification.Here, we describe a workflow for the proteomics analysis of microbial niches with a focus on intestinal mucus layer. We will cover step-by-step the sample collection, sample preparation, liquid chromatography-mass spectrometry, and data analysis.


Asunto(s)
Bacterias/aislamiento & purificación , Proteínas Bacterianas/análisis , Proteínas Fúngicas/análisis , Hongos/aislamiento & purificación , Microbioma Gastrointestinal , Proteómica/métodos , Animales , Cromatografía Liquida/métodos , Intestinos/microbiología , Espectrometría de Masas/métodos , Ratones , Péptidos/análisis , Flujo de Trabajo
8.
Cell Rep ; 34(7): 108757, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33596425

RESUMEN

The intestine is under constant exposure to chemicals, antigens, and microorganisms from the external environment. Apical aspects of transporting epithelial cells (enterocytes) form a brush-border membrane (BBM), shaped by packed microvilli coated with a dense glycocalyx. We present evidence showing that the glycocalyx forms an epithelial barrier that prevents exogenous molecules and live bacteria from gaining access to BBM. We use a multi-omics approach to investigate the function and regulation of membrane mucins exposed on the BBM during postnatal development of the mouse small intestine. Muc17 is identified as a major membrane mucin in the glycocalyx that is specifically upregulated by IL-22 as part of an epithelial defense repertoire during weaning. High levels of IL-22 at time of weaning reprogram neonatal postmitotic progenitor enterocytes to differentiate into Muc17-expressing enterocytes, as found in the adult intestine during homeostasis. Our findings propose a role for Muc17 in epithelial barrier function in the small intestine.


Asunto(s)
Glicocálix/metabolismo , Interleucinas/metabolismo , Intestino Delgado/metabolismo , Mucinas/metabolismo , Adulto , Animales , Células CHO , Cricetulus , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Destete , Interleucina-22
9.
Anal Biochem ; 597: 113668, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32222540

RESUMEN

In order to demonstrate transglutaminase activity in biological samples immunological as well as glutamine- and amine-donor based assays are commonly used. However, the identification of the transglutaminase reaction product, i. e. the isopeptide cross-linked peptides/proteins or the deamidated protein/peptide are often neglected. This article describes a workflow for the detection of the products of transglutaminase-catalyzed reactions. In particular, possible pitfalls and traps that can arise during the mass spectrometry-based identification of isopeptide cross-links are addressed and characterised on actual samples.


Asunto(s)
Reactivos de Enlaces Cruzados/análisis , Mucina 2/metabolismo , Péptidos/análisis , Transglutaminasas/metabolismo , Biocatálisis , Reactivos de Enlaces Cruzados/metabolismo , Espectrometría de Masas , Mucina 2/química , Péptidos/metabolismo , Transglutaminasas/química
10.
Cell Rep ; 30(4): 1077-1087.e3, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31995731

RESUMEN

The gastrointestinal tract is covered by a single layer of epithelial cells that, together with the mucus layers, protect the underlying tissue from microbial invasion. The epithelium has one of the highest turnover rates in the body. Using stable isotope labeling, high-resolution mass spectrometry, and computational analysis, we report a comprehensive dataset of the turnover of more than 3,000 and the expression of more than 5,000 intestinal epithelial cell proteins, analyzed under conventional and germ-free conditions across five different segments in mouse intestine. The median protein half-life is shorter in the small intestine than in the colon. Differences in protein turnover rates along the intestinal tract can be explained by distinct physiological and immune-related functions between the small and large intestine. An absence of microbiota results in an approximately 1 day longer protein half-life in germ-free animals.


Asunto(s)
Células Epiteliales/metabolismo , Tracto Gastrointestinal/fisiología , Microbiota/fisiología , Moco/metabolismo , Transporte de Proteínas/fisiología , Proteómica/métodos , Animales , Humanos , Ratones , Moco/citología
11.
J Biol Chem ; 294(45): 17075-17089, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31570526

RESUMEN

Calcium-activated chloride channel regulator 1 (CLCA1) is one of the major nonmucin proteins found in intestinal mucus. It is part of a larger family of CLCA proteins that share highly conserved features and domain architectures. The CLCA domain arrangement is similar to proteins belonging to the ADAM (a disintegrin and metalloproteinase) family, known to process extracellular matrix proteins. Therefore, CLCA1 is an interesting candidate in the search for proteases that process intestinal mucus. Here, we investigated CLCA1's biochemical properties both in vitro and in mucus from mouse and human colon biopsy samples. Using immunoblotting with CLCA1-specific antibodies and recombinant proteins, we observed that the CLCA1 C-terminal self-cleavage product forms a disulfide-linked dimer that noncovalently interacts with the N-terminal part of CLCA1, which further interacts to form oligomers. We also characterized a second, more catalytically active, N-terminal product of CLCA1, encompassing the catalytic domain together with its von Willebrand domain type A (VWA). This fragment was unstable but could be identified in freshly prepared mucus. Furthermore, we found that CLCA1 can cleave the N-terminal part of the mucus structural component MUC2. We propose that CLCA1 regulates the structural arrangement of the mucus and thereby takes part in the regulation of mucus processing.


Asunto(s)
Canales de Cloruro/química , Canales de Cloruro/metabolismo , Colon/metabolismo , Mucosa Intestinal/metabolismo , Mucina 2/metabolismo , Multimerización de Proteína , Proteolisis , Secuencia de Aminoácidos , Animales , Humanos , Ratones , Dominios Proteicos , Estructura Cuaternaria de Proteína
13.
Biochem J ; 476(16): 2281-2295, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31387973

RESUMEN

Transmembrane mucin MUC17 is an integral part of the glycocalyx as it covers the brush border membrane of small intestinal enterocytes and presents an extended O-glycosylated mucin domain to the intestinal lumen. Here, we identified two unknown phosphorylated serine residues, S4428 and S4492, in the cytoplasmic tail of human MUC17. We have previously demonstrated that MUC17 is anchored to the apical membrane domain via an interaction with the scaffolding protein PDZK1. S4492, localized in the C-terminal PDZ binding motif of MUC17, was mutated to generate phosphomimetic and phosphodeficient variants of MUC17. Using Caco-2 cells as a model system, we found that induction of an inflammatory state by long-term stimulation with the proinflammatory cytokine TNFα resulted in an increase of MUC17 protein levels and enhanced insertion of MUC17 and its two phospho-variants into apical membranes. Up-regulation and apical insertion of MUC17 was followed by shedding of MUC17-containing vesicles. Transmembrane mucins have previously been shown to play a role in the prevention of bacterial colonization by acting as sheddable decoys for encroaching bacteria. Overexpression and increased presentation at the plasma membrane of wild-type MUC17 and its phosphodeficient variant MUC17 S-4492A protected Caco-2 cells against adhesion of enteropathogenic Escherichia coli, indicating that C-terminal phosphorylation of MUC17 may play a functional role in epithelial cell protection. We propose a new function for MUC17 in inflammation, where MUC17 acts as a second line of defense by preventing attachment of bacteria to the epithelial cell glycocalyx in the small intestine.


Asunto(s)
Adhesión Bacteriana , Escherichia coli Enteropatógena/metabolismo , Glicocálix/metabolismo , Intestino Delgado/metabolismo , Mucinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Sustitución de Aminoácidos , Células CACO-2 , Glicocálix/microbiología , Glicocálix/patología , Células HEK293 , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Intestino Delgado/microbiología , Intestino Delgado/patología , Mucinas/genética , Mutación Missense , Dominios PDZ , Fosforilación/genética , Factor de Necrosis Tumoral alfa/genética
14.
Front Physiol ; 10: 694, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31263421

RESUMEN

Calcium-activated anion secretion is expected to ameliorate cystic fibrosis, a genetic disease that carries an anion secretory defect in exocrine tissues. Human patients and animal models of the disease that present a mild intestinal phenotype have been postulated to bear a compensatory calcium-activated anion secretion in the intestine. TMEM16A is calcium-activated anion channel whose presence in the intestinal epithelium is contradictory. We aim to test the functional expression of TMEM16A using animal models with Cftr and/or Tmem16a intestinal silencing. Expression of TMEM16A was studied in a wild type and intestinal Tmem16a knockout mice by mRNA-seq, mass-spectrometry, q-PCR, Western blotting and immunolocalization. Calcium-activated anion secretion was recorded in the ileum and proximal colon of these animals including intestinal Cftr knockout and double mutants with dual Tmem16a and Cftr intestinal ablation. Mucus homeostasis was studied by immune-analysis of Mucin-2 (Muc2) and survival curves were recorded. Tmem16a transcript was found in intestine. Nevertheless, protein was barely detected in colon samples. Electrophysiological measurements demonstrated that the intestinal deletion of Tmem16a did not change calcium-activated anion secretion induced by carbachol or ATP in ileum and proximal colon. Muc2 architecture was not altered by Tmem16a silencing as was observed when Cftr was deleted from mouse intestine. Tmem16a silencing neither affected animal survival nor modified the lethality observed in the intestinal Cftr-null mouse. Our results demonstrate that TMEM16A function in the murine intestine is not related to electrogenic calcium-activated anion transport and does not affect mucus homeostasis and survival of animals.

15.
Nature ; 572(7770): 474-480, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31330533

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a complex neurodegenerative disorder, in which the clinical manifestations may be influenced by genetic and unknown environmental factors. Here we show that ALS-prone Sod1 transgenic (Sod1-Tg) mice have a pre-symptomatic, vivarium-dependent dysbiosis and altered metabolite configuration, coupled with an exacerbated disease under germ-free conditions or after treatment with broad-spectrum antibiotics. We correlate eleven distinct commensal bacteria at our vivarium with the severity of ALS in mice, and by their individual supplementation into antibiotic-treated Sod1-Tg mice we demonstrate that Akkermansia muciniphila (AM) ameliorates whereas Ruminococcus torques and Parabacteroides distasonis exacerbate the symptoms of ALS. Furthermore, Sod1-Tg mice that are administered AM are found to accumulate AM-associated nicotinamide in the central nervous system, and systemic supplementation of nicotinamide improves motor symptoms and gene expression patterns in the spinal cord of Sod1-Tg mice. In humans, we identify distinct microbiome and metabolite configurations-including reduced levels of nicotinamide systemically and in the cerebrospinal fluid-in a small preliminary study that compares patients with ALS with household controls. We suggest that environmentally driven microbiome-brain interactions may modulate ALS in mice, and we call for similar investigations in the human form of the disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/microbiología , Esclerosis Amiotrófica Lateral/fisiopatología , Microbioma Gastrointestinal/fisiología , Niacinamida/metabolismo , Akkermansia , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Antibacterianos/farmacología , Modelos Animales de Enfermedad , Disbiosis , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Vida Libre de Gérmenes , Humanos , Longevidad , Masculino , Ratones , Ratones Transgénicos , Niacinamida/biosíntesis , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Tasa de Supervivencia , Simbiosis/efectos de los fármacos , Verrucomicrobia/metabolismo , Verrucomicrobia/fisiología
16.
Gut ; 68(12): 2142-2151, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30914450

RESUMEN

OBJECTIVE: The colonic inner mucus layer protects us from pathogens and commensal-induced inflammation, and has been shown to be defective in active UC. The aim of this study was to determine the underlying compositional alterations, their molecular background and potential contribution to UC pathogenesis. DESIGN: In this single-centre case-control study, sigmoid colon biopsies were obtained from patients with UC with ongoing inflammation (n=36) or in remission (n=28), and from 47 patients without colonic disease. Mucus samples were collected from biopsies ex vivo, and their protein composition analysed by nanoliquid chromatography-tandem mass spectrometry. Mucus penetrability and goblet cell responses to microbial stimulus were assessed in a subset of patients. RESULTS: The core mucus proteome was found to consist of a small set of 29 secreted/transmembrane proteins. In active UC, major structural mucus components including the mucin MUC2 (p<0.0001) were reduced, also in non-inflamed segments. Active UC was associated with decreased numbers of sentinel goblet cells and attenuation of the goblet cell secretory response to microbial challenge. Abnormal penetrability of the inner mucus layer was observed in a subset of patients with UC (12/40; 30%). Proteomic alterations in penetrable mucus samples included a reduction of the SLC26A3 apical membrane anion exchanger, which supplies bicarbonate required for colonic mucin barrier formation. CONCLUSION: Core mucus structural components were reduced in active UC. These alterations were associated with attenuation of the goblet cell secretory response to microbial challenge, but occurred independent of local inflammation. Thus, mucus abnormalities are likely to contribute to UC pathogenesis.


Asunto(s)
Colitis Ulcerosa/patología , Colon/patología , Mucosa Intestinal/metabolismo , Mucinas/metabolismo , Moco/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Colitis Ulcerosa/metabolismo , Colon/metabolismo , Colonoscopía , Femenino , Estudios de Seguimiento , Humanos , Mucosa Intestinal/patología , Masculino , Persona de Mediana Edad , Proteómica/métodos , Estudios Retrospectivos , Factores de Tiempo , Adulto Joven
17.
JCI Insight ; 3(17)2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30185674

RESUMEN

The respiratory tract is normally kept essentially free of bacteria by cilia-mediated mucus transport, but in chronic obstructive pulmonary disease (COPD) and cystic fibrosis (CF), bacteria and mucus accumulates instead. To address the mechanisms behind the mucus accumulation, the proteome of bronchoalveolar lavages from COPD patients and mucus collected in an elastase-induced mouse model of COPD was analyzed, revealing similarities with each other and with the protein content in colonic mucus. Moreover, stratified laminated sheets of mucus were observed in airways from patients with CF and COPD and in elastase-exposed mice. On the other hand, the mucus accumulation in the elastase model was reduced in Muc5b-KO mice. While mucus plugs were removed from airways by washing with hypertonic saline in the elastase model, mucus remained adherent to epithelial cells. Bacteria were trapped on this mucus, whereas, in non-elastase-treated mice, bacteria were found on the epithelial cells. We propose that the adherence of mucus to epithelial cells observed in CF, COPD, and the elastase-induced mouse model of COPD separates bacteria from the surface cells and, thus, protects the respiratory epithelium.


Asunto(s)
Bacterias , Células Epiteliales/metabolismo , Moco/microbiología , Moco/fisiología , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Animales , Líquido del Lavado Bronquioalveolar , Fibrosis Quística/complicaciones , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Células Epiteliales/patología , Femenino , Humanos , Pulmón , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucina 5B/genética , Elastasa Pancreática , Pseudomonas aeruginosa , Mucosa Respiratoria
18.
EBioMedicine ; 33: 134-143, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29885864

RESUMEN

Many epithelial surfaces of the body are covered with protective mucus, and disrupted mucus homeostasis is coupled to diseases such as ulcerative colitis, helminth infection, cystic fibrosis, and chronic obstructive lung disease. However, little is known how a balanced mucus system is maintained. By investigating the involvement of proteases in colonic mucus dynamics we identified metalloprotease activity to be a key contributor to mucus expansion. The effect was mediated by calcium-activated chloride channel regulator 1 (CLCA1) as application of recombinant CLCA1 on intestinal mucus in freshly dissected tissue resulted in increased mucus thickness independently of ion and mucus secretion, but dependent on its metallohydrolase activity. Further, CLCA1 modulated mucus dynamics in both human and mouse, and knock-out of CLCA1 in mice was compensated for by cysteine proteases. Our results suggest that CLCA1 is involved in intestinal mucus homeostasis by facilitating processing and removal of mucus to prevent stagnation. In light of our findings, we suggest future studies to investigate if upregulation of CLCA1 in diseases associated with mucus accumulation could facilitate removal of mucus in an attempt to maintain homeostasis.


Asunto(s)
Canales de Cloruro/metabolismo , Colon/metabolismo , Moco/metabolismo , Animales , Canales de Cloruro/genética , Homeostasis , Humanos , Metaloproteasas/metabolismo , Ratones , Ratones Noqueados , Proteolisis , Proteoma/química
19.
Cell Host Microbe ; 23(1): 27-40.e7, 2018 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29276171

RESUMEN

Diet strongly affects gut microbiota composition, and gut bacteria can influence the colonic mucus layer, a physical barrier that separates trillions of gut bacteria from the host. However, the interplay between a Western style diet (WSD), gut microbiota composition, and the intestinal mucus layer is less clear. Here we show that mice fed a WSD have an altered colonic microbiota composition that causes increased penetrability and a reduced growth rate of the inner mucus layer. Both barrier defects can be prevented by transplanting microbiota from chow-fed mice. In addition, we found that administration of Bifidobacterium longum was sufficient to restore mucus growth, whereas administration of the fiber inulin prevented increased mucus penetrability in WSD-fed mice. We hypothesize that the presence of distinct bacteria is crucial for proper mucus function. If confirmed in humans, these findings may help to better understand diseases with an affected mucus layer, such as ulcerative colitis.


Asunto(s)
Bifidobacterium longum/metabolismo , Colon/microbiología , Dieta Occidental/efectos adversos , Fibras de la Dieta/uso terapéutico , Trasplante de Microbiota Fecal , Mucosa Intestinal/microbiología , Animales , Colon/patología , Suplementos Dietéticos , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/patología , Inulina/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/patología
20.
J Clin Oncol ; 36(4): 367-375, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29166170

RESUMEN

Purpose Pancreatic cystic lesions are common incidental findings on imaging, but up to half may be forerunners of pancreatic cancer. Therefore, accurate differential diagnosis is crucial for correct patient management. Unfortunately, currently available diagnostic methods cannot robustly identify premalignant and malignant pancreatic cystic lesions. Methods Cyst fluid samples obtained by routine endoscopic ultrasound-guided aspiration were used for the analyses. In a cohort of 24 patients, eight biomarker candidates for malignant potential and high-grade dysplasia/cancer were identified by an explorative proteomic approach. Subsequently, a quantitative analysis, using 30 heavy-labeled peptides from the biomarkers and parallel reaction monitoring mass spectrometry, was devised, tested in a training cohort of 80, and prospectively evaluated in a validation cohort of 68 patients. End points were surgical pathology diagnosis/clinical follow-up. Diagnostic assessments were blinded to mass spectrometry results. Results The optimal set of markers for detecting malignant potential was a panel of peptides from mucin-5AC and mucin-2, which could discriminate premalignant/malignant lesions from benign with an accuracy of 97% (95% CI, 89% to 99%) in the validation cohort. This result compared favorably with the accuracy of standard analyses: cyst fluid carcinoembryonic antigen (61%; 95% CI, 46% to 74%; P < .001) and cytology (84%; 95% CI, 71% to 92%; P = .02). A combination of proteins mucin-5AC and prostate stem-cell antigen could identify high-grade dysplasia/cancer with an accuracy of 96% (95% CI, 90% to 99%), and detected 95% of malignant/severely dysplastic lesions, compared with 35% and 50% for carcinoembryonic antigen and cytology ( P < .001 and P = .003, respectively). Conclusion Targeted mass spectrometry analysis of just three cyst fluid biomarkers provides highly accurate identification and assessment of cystic precursors to pancreatic adenocarcinoma. Additional studies should determine whether the method can facilitate timely cancer diagnosis, successful intervention, and prevention.


Asunto(s)
Biomarcadores de Tumor/análisis , Espectrometría de Masas , Quiste Pancreático/química , Neoplasias Pancreáticas/química , Lesiones Precancerosas/metabolismo , Proteómica/métodos , Anciano , Antígenos de Neoplasias/análisis , Antígeno Carcinoembrionario/análisis , Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico , Femenino , Proteínas Ligadas a GPI/análisis , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Mucina 5AC/análisis , Mucina 2/análisis , Proteínas de Neoplasias/análisis , Quiste Pancreático/patología , Neoplasias Pancreáticas/patología , Lesiones Precancerosas/patología , Valor Predictivo de las Pruebas , Estudios Prospectivos , Reproducibilidad de los Resultados , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...