Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Publication year range
1.
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
2.
Proc Natl Acad Sci U S A ; 117(35): 21519-21526, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817517

ABSTRACT

The intestinal epithelium is a highly dynamic structure that rejuvenates in response to acute stressors and can undergo alterations in cellular composition as animals age. The microbiota, acting via secreted factors related to indole, appear to regulate the sensitivity of the epithelium to stressors and promote epithelial repair via IL-22 and type I IFN signaling. As animals age, the cellular composition of the intestinal epithelium changes, resulting in a decreased proportion of goblet cells in the colon. We show that colonization of young or geriatric mice with bacteria that secrete indoles and various derivatives or administration of the indole derivative indole-3 aldehyde increases proliferation of epithelial cells and promotes goblet cell differentiation, reversing an effect of aging. To induce goblet cell differentiation, indole acts via the xenobiotic aryl hydrocarbon receptor to increase expression of the cytokine IL-10. However, the effects of indoles on goblet cells do not depend on type I IFN or on IL-22 signaling, pathways responsible for protection against acute stressors. Thus, indoles derived from the commensal microbiota regulate intestinal homeostasis, especially during aging, via mechanisms distinct from those used during responses to acute stressors. Indoles may have utility as an intervention to limit the decline of barrier integrity and the resulting systemic inflammation that occurs with aging.


Subject(s)
Goblet Cells/drug effects , Goblet Cells/microbiology , Indoles/pharmacology , Interleukin-10/metabolism , Microbiota/physiology , Receptors, Aryl Hydrocarbon/metabolism , Aging/metabolism , Animals , Bacteria/metabolism , Cell Differentiation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Goblet Cells/cytology , Goblet Cells/metabolism , Interleukin-10/biosynthesis , Interleukins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mucus/metabolism , Signal Transduction , Interleukin-22
3.
J Fish Dis ; 43(10): 1249-1258, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32830331

ABSTRACT

Groupers are popular aquaculture species in South-East Asia, but their cultivation is affected by infectious disease outbreaks. Mucosa-associated lymphoid tissues provide a first-line defence against pathogens; however, few studies are available relating to cellular or proteomic responses of mucosal immunity in grouper. Skin, gill and intestine were sampled from brown-marbled grouper Epinephelus fuscoguttatus (Forsskål, 1775) at 4 and 96 hr post-infection (hpi) and 7 days post-infection (dpi) following intraperitoneal infection with Vibrio harveyi, and stained with haematoxylin/eosin and Alcian Blue/periodic acid-Schiff. Skin mucus was analysed by 2D-gel electrophoresis, and proteins modulated by the bacterial infection identified. In the infected fish, significant increases in sacciform cells in skin and increased levels of nucleoside diphosphate kinase in mucus were detected at 4 hpi. At 96 hpi, goblet cells containing acidic mucins significantly increased in the intestine, while those containing mixed mucins increased in skin and gills of infected fish. Proteasome subunit alpha type-I and extracellular Cu/Zn superoxide dismutase levels also increased in mucus. Rodlet and mast cells did not appear to respond to the infection. Mucosal tissues of grouper appeared actively involved in response to Vibrio infection. This information may help future research on improving grouper health, production and vaccine development.


Subject(s)
Bass/immunology , Fish Diseases/immunology , Immunity, Mucosal , Vibrio Infections/veterinary , Animals , Bass/microbiology , Fish Diseases/microbiology , Goblet Cells/microbiology , Mucous Membrane/microbiology , Mucous Membrane/pathology , Mucus , Proteome , Vibrio , Vibrio Infections/immunology
4.
Am J Physiol Gastrointest Liver Physiol ; 314(3): G360-G377, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29122749

ABSTRACT

Goblet cells (GCs) are the predominant secretory epithelial cells lining the luminal surface of the mammalian gastrointestinal (GI) tract. Best known for their apical release of mucin 2 (Muc2), which is critical for the formation of the intestinal mucus barrier, GCs have often been overlooked for their active contributions to intestinal protection and host defense. In part, this oversight reflects the limited tools available to study their function but also because GCs have long been viewed as relatively passive players in promoting intestinal homeostasis and host defense. In light of recent studies, this perspective has shifted, as current evidence suggests that Muc2 as well as other GC mediators are actively released into the lumen to defend the host when the GI tract is challenged by noxious stimuli. The ability of GCs to sense and respond to danger signals, such as bacterial pathogens, has recently been linked to inflammasome signaling, potentially intrinsic to the GCs themselves. Moreover, further work suggests that GCs release Muc2, as well as other mediators, to modulate the composition of the gut microbiome, leading to both the expansion as well as the depletion of specific gut microbes. This review will focus on the mechanisms by which GCs actively defend the host from noxious stimuli, as well as describe advanced technologies and new approaches by which their responses can be addressed. Taken together, we will highlight current insights into this understudied, yet critical, aspect of intestinal mucosal protection and its role in promoting gut defense and homeostasis.


Subject(s)
Bacteria/pathogenicity , Bacterial Infections/microbiology , Gastrointestinal Microbiome , Goblet Cells/microbiology , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/microbiology , Animals , Bacteria/growth & development , Bacterial Infections/metabolism , Bacterial Infections/physiopathology , Goblet Cells/metabolism , Homeostasis , Host-Pathogen Interactions , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/physiopathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiopathology , Mucin-2/metabolism , Mucus/metabolism , Signal Transduction
5.
Gut ; 65(7): 1100-9, 2016 07.
Article in English | MEDLINE | ID: mdl-26045138

ABSTRACT

OBJECTIVE: Antibiotic use is associated with an increased risk of developing multiple inflammatory disorders, which in turn are linked to alterations in the intestinal microbiota. How these alterations in the intestinal microbiota translate into an increased risk for inflammatory responses is largely unknown. Here we investigated whether and how antibiotics promote inflammation via the translocation of live native gut commensal bacteria. DESIGN: Oral antibiotics were given to wildtype and induced mutant mouse strains, and the effects on bacterial translocation, inflammatory responses and the susceptibility to colitis were evaluated. The sources of the bacteria and the pathways required for bacterial translocation were evaluated using induced mutant mouse strains, 16s rRNA sequencing to characterise the microbial communities, and in vivo and ex vivo imaging techniques. RESULTS: Oral antibiotics induced the translocation of live native commensal bacteria across the colonic epithelium, promoting inflammatory responses, and predisposing to increased disease in response to coincident injury. Bacterial translocation resulted from decreased microbial signals delivered to colonic goblet cells (GCs), was associated with the formation of colonic GC-associated antigen passages, was abolished when GCs were depleted and required CX3CR1(+) dendritic cells. Bacterial translocation occurred following a single dose of most antibiotics tested, and the predisposition for increased inflammation was only associated with antibiotics inducing bacterial translocation. CONCLUSIONS: These findings reveal an unexpected outcome of antibiotic therapy and suggest that bacterial translocation as a result of alterations in the intestinal microflora may provide a link between increasing antibiotic use and the increased incidence of inflammatory disorders.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Translocation/drug effects , Colon/microbiology , Goblet Cells/metabolism , Goblet Cells/microbiology , Inflammation/microbiology , Lymph Nodes/microbiology , Animals , Bacterial Translocation/immunology , CX3C Chemokine Receptor 1 , Colitis/microbiology , Cytokines/metabolism , Dendritic Cells/chemistry , Disease Susceptibility , Enterococcus faecalis/isolation & purification , Escherichia coli/isolation & purification , Goblet Cells/immunology , Intestine, Small/microbiology , Mesentery , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88 , Receptors, Chemokine/analysis
6.
Int J Med Microbiol ; 306(8): 657-665, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27670078

ABSTRACT

Vibrio cholera survival in an aquatic environment depends on chitin utilization pathway that requires two factors, chitin binding protein and chitinases. The chitinases and the chitin utilization pathway are regulated by a two-component sensor histidine kinase ChiS in V. cholerae. In recent studies these two factors are also shown to be involved in V. cholerae pathogenesis. However, the role played by their upstream regulator ChiS in pathogenesis is yet to be known. In this study, we investigated the activation of ChiS in presence of mucin and its functional role in pathogenesis. We found ChiS is activated in mucin supplemented media. The isogenic chiS mutant (ChiS-) showed less growth compared to the wild type strain (ChiS+) in the presence of mucin supplemented media. The ChiS- strain also showed highly retarded motility as well as mucin layer penetration in vitro. Our result also showed that ChiS was important for adherence and survival in HT-29 cell. These observations indicate that ChiS is activated in presence of intestinal mucin and subsequently switch on the chitin utilization pathway. In animal models, our results also supported the in vitro observation. We found reduced fluid accumulation and colonization during infection with ChiS- strain. We also found ChiS- mutant with reduced expression of ctxA, toxT and tcpA. The cumulative effect of these events made V. cholerae ChiS- strain hypovirulent. Hence, we propose that ChiS plays a vital role in V. cholerae pathogenesis.


Subject(s)
Histidine Kinase/metabolism , Vibrio cholerae/pathogenicity , Virulence Factors/metabolism , Animals , Bacterial Adhesion , Bacterial Proteins/metabolism , Cell Line , Cholera Toxin/metabolism , Culture Media/chemistry , Fimbriae Proteins/metabolism , Gene Deletion , Gene Expression Regulation, Bacterial , Goblet Cells/microbiology , Histidine Kinase/genetics , Humans , Locomotion , Mice , Microbial Viability , Mucins/metabolism , Rabbits , Transcription Factors/metabolism , Transcriptional Activation/drug effects , Vibrio cholerae/growth & development
7.
J Appl Microbiol ; 120(5): 1403-17, 2016 May.
Article in English | MEDLINE | ID: mdl-26868655

ABSTRACT

AIMS: The molecular cross-talk between commensal bacteria and the gut play an important role in the maintenance of the intestinal homeostasis and general health. Here, we studied the impact of a major Gram-positive anaerobic bacterium of the human gut microbiota, that is, Ruminococcus gnavus on the glycosylation pattern and the production of intestinal mucus by the goblet cells. METHODS AND RESULTS: Our results showed that R. gnavus E1 specifically increases the expression and the glycosylation level of the intestinal glyco-conjugates by goblet cells in the colonic mucosa of mono-associated mice with R. gnavus E1 as well as in human HT29-MTX cells. Such an effect was mediated through induction of the level of mRNA encoding for the major intestinal gel-forming mucin such as MUC2 and various glycosyltransferase enzymes. CONCLUSIONS: This study demonstrates for the first time that R. gnavus E1 possess the ability to modulate the glycosylation profile of the glyco-conjugate molecules and mucus in goblet cells. SIGNIFICANCE AND IMPACT OF THE STUDY: Furthermore, we demonstrated that R. gnavus E1 modified specifically the glycosylation pattern and MUC2 expression by means of a small soluble factor of peptidic nature (<3 kDa) and heat stable in the HT29-MTX cell.


Subject(s)
Gastrointestinal Microbiome , Mucins/metabolism , Ruminococcus/physiology , Animals , Colon/metabolism , Colon/microbiology , Glycosylation , Goblet Cells/metabolism , Goblet Cells/microbiology , HT29 Cells , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestines/microbiology , Mice
8.
Carcinogenesis ; 36(11): 1388-96, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26320104

ABSTRACT

Gastrointestinal tumor growth is thought to be promoted by gastrointestinal bacteria and their inflammatory products. We observed that intestine-specific conditional Apc mutant mice (FabplCre;Apc (15lox/+)) developed many more colorectal tumors under conventional than under pathogen-low housing conditions. Shotgun metagenomic sequencing plus quantitative PCR analysis of feces DNA revealed the presence of two bacterial species in conventional mice, absent from pathogen-low mice. One, Helicobacter typhlonius, has not been associated with cancer in man, nor in immune-competent mice. The other species, mucin-degrading Akkermansia muciniphila, is abundantly present in healthy humans, but reduced in patients with inflammatory gastrointestinal diseases and in obese and type 2 diabetic mice. Eradication of H.typhlonius in young conventional mice by antibiotics decreased the number of intestinal tumors. Additional presence of A.muciniphila prior to the antibiotic treatment reduced the tumor number even further. Colonization of pathogen-low FabplCre;Apc (15lox/+) mice with H.typhlonius or A.muciniphila increased the number of intestinal tumors, the thickness of the intestinal mucus layer and A.muciniphila colonization without H.typhlonius increased the density of mucin-producing goblet cells. However, dual colonization with H.typhlonius and A.muciniphila significantly reduced the number of intestinal tumors, the mucus layer thickness and goblet cell density to that of control mice. By global microbiota composition analysis, we found a positive association of A.muciniphila, and of H.typhlonius, and a negative association of unclassified Clostridiales with increased tumor burden. We conclude that A.muciniphila and H.typhlonius can modulate gut microbiota composition and intestinal tumor development in mice.


Subject(s)
Anti-Bacterial Agents/pharmacology , Helicobacter Infections/complications , Helicobacter/drug effects , Intestinal Neoplasms/microbiology , Verrucomicrobia/drug effects , Amoxicillin/pharmacology , Animals , Carcinogenesis , Clarithromycin/pharmacology , Drug Therapy, Combination , Female , Gastrointestinal Microbiome , Goblet Cells/microbiology , Helicobacter Infections/drug therapy , Intestinal Neoplasms/prevention & control , Intestines/microbiology , Intestines/pathology , Male , Metronidazole/pharmacology , Mice, Inbred C57BL , Omeprazole/pharmacology
9.
Infect Immun ; 82(9): 3753-63, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24958710

ABSTRACT

Intestinal epithelial cells (IECs), including secretory goblet cells, form essential physiochemical barriers that separate luminal bacteria from underlying immune cells in the intestinal mucosa. IECs are common targets for enteric bacterial pathogens, with hosts responding to these microbes through innate toll-like receptors that predominantly signal through the MyD88 adaptor protein. In fact, MyD88 signaling confers protection against several enteric bacterial pathogens, including Salmonella enterica serovar Typhimurium and Citrobacter rodentium. Since IECs are considered innately hyporesponsive, it is unclear whether MyD88 signaling within IECs contributes to this protection. We infected mice lacking MyD88 solely in their IECs (IEC-Myd88(-/-)) with S. Typhimurium. Compared to wild-type (WT) mice, infected IEC-Myd88(-/-) mice suffered accelerated tissue damage, exaggerated barrier disruption, and impaired goblet cell responses (Muc2 and RELMß). Immunostaining revealed S. Typhimurium penetrated the IECs of IEC-Myd88(-/-) mice, unlike in WT mice, where they were sequestered to the lumen. When isolated crypts were assayed for their antimicrobial actions, crypts from IEC-Myd88(-/-) mice were severely impaired in their antimicrobial activity against S. Typhimurium. We also examined whether MyD88 signaling in IECs impacted host defense against C. rodentium, with IEC-Myd88(-/-) mice again suffering exaggerated tissue damage, impaired goblet cell responses, and reduced antimicrobial activity against C. rodentium. These results demonstrate that MyD88 signaling within IECs plays an important protective role at early stages of infection, influencing host susceptibility to infection by controlling the ability of the pathogen to reach and survive at the intestinal mucosal surface.


Subject(s)
Anti-Infective Agents/immunology , Colitis/immunology , Goblet Cells/immunology , Intestinal Mucosa/immunology , Myeloid Differentiation Factor 88/immunology , Signal Transduction/immunology , Animals , Citrobacter rodentium/immunology , Colitis/microbiology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Gastroenteritis/immunology , Gastroenteritis/microbiology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Goblet Cells/microbiology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/immunology
10.
Eur J Immunol ; 43(12): 3108-15, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24414823

ABSTRACT

Continuous exposure of intestinal mucosal surfaces to diverse microorganisms and their metabolites reflects the biological necessity for a multifaceted, integrated epithelial and immune cell-mediated regulatory system. The development and function of the host cells responsible for the barrier function of the intestinal surface (e.g., M cells, Paneth cells, goblet cells, and columnar epithelial cells) are strictly regulated through both positive and negative stimulation by the luminal microbiota. Stimulation by damage-associated molecular patterns and commensal bacteria-derived microbe-associated molecular patterns provokes the assembly of inflammasomes, which are involved in maintaining the integrity of the intestinal epithelium. Mucosal immune cells located beneath the epithelium play critical roles in regulating both the mucosal barrier and the relative composition of the luminal microbiota. Innate lymphoid cells and mast cells, in particular, orchestrate the mucosal regulatory system to create a mutually beneficial environment for both the host and the microbiota. Disruption of mucosal homeostasis causes intestinal inflammation such as that seen in inflammatory bowel disease. Here, we review the recent research on the biological interplay among the luminal microbiota, epithelial cells, and mucosal innate immune cells in both healthy and pathological conditions.


Subject(s)
Bacteria/immunology , Goblet Cells/immunology , Homeostasis/immunology , Immunity, Innate/physiology , Immunity, Mucosal/physiology , Paneth Cells/immunology , Animals , Goblet Cells/cytology , Goblet Cells/microbiology , Humans , Inflammation/immunology , Inflammation/microbiology , Lymphocytes/cytology , Lymphocytes/immunology , Mast Cells/cytology , Mast Cells/immunology , Paneth Cells/cytology , Paneth Cells/microbiology
11.
Exp Lung Res ; 40(6): 298-307, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24927409

ABSTRACT

Epidemiologic studies suggest that increased concentrations of airborne spores of Aspergillus fumigatus closely relate to asthma aggravation. Chronic exposure to A. fumigatus aggravates airway inflammation, remodeling, and airway hyperresponsiveness in asthmatic rats. The effects of chronic exposure to A. fumigatus on epidermal growth factor receptor (EGFR) expression in the airway epithelial cells of asthmatic rats remain unclear. This study aimed to investigate the effects of chronic exposure to A. fumigatus on injury and shedding of airway epithelium, goblet cell metaplasia, and EGFR expression in the airway epithelial cells of asthmatic rats. A rat model of chronic asthma was established using ovalbumin (OVA) sensitization and challenge. Rats with chronic asthma were then exposed to long-term inhalation of spores of A. fumigatus, and the dynamic changes in injury and shedding of airway epithelium, goblet cell metaplasia, and EGFR expression were observed and analyzed. Chronic exposure to A. fumigatus could aggravate airway epithelial cell damage, upregulate the expression of EGFR and its ligands EGF and TGF-α, promote goblet cell metaplasia, and increase airway responsiveness in rats with asthma. Chronic exposure to A. fumigatus upregulates the expression of EGFR and its ligands in asthmatic rats. The EGFR pathway may play a role in asthma aggravation induced by exposure to A. fumigatus.


Subject(s)
Aspergillosis/metabolism , Aspergillus fumigatus/metabolism , Asthma/metabolism , Asthma/microbiology , ErbB Receptors/metabolism , Pneumonia/metabolism , Animals , Aspergillosis/microbiology , Aspergillosis/pathology , Asthma/pathology , Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/microbiology , Bronchial Hyperreactivity/pathology , Bronchoalveolar Lavage Fluid , Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Goblet Cells/metabolism , Goblet Cells/microbiology , Goblet Cells/pathology , Male , Metaplasia/metabolism , Metaplasia/microbiology , Ovalbumin/pharmacology , Pneumonia/microbiology , Pneumonia/pathology , Rats , Rats, Wistar
12.
BMC Biol ; 11: 61, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23692866

ABSTRACT

BACKGROUND: The intestinal mucus layer plays a key role in the maintenance of host-microbiota homeostasis. To document the crosstalk between the host and microbiota, we used gnotobiotic models to study the influence of two major commensal bacteria, Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii, on this intestinal mucus layer. B. thetaiotaomicron is known to use polysaccharides from mucus, but its effect on goblet cells has not been addressed so far. F. prausnitzii is of particular physiological importance because it can be considered as a sensor and a marker of human health. We determined whether B. thetaiotaomicron affected goblet cell differentiation, mucin synthesis and glycosylation in the colonic epithelium. We then investigated how F. prausnitzii influenced the colonic epithelial responses to B. thetaiotaomicron. RESULTS: B. thetaiotaomicron, an acetate producer, increased goblet cell differentiation, expression of mucus-related genes and the ratio of sialylated to sulfated mucins in mono-associated rats. B. thetaiotaomicron, therefore, stimulates the secretory lineage, favoring mucus production. When B. thetaiotaomicron was associated with F. prausnitzii, an acetate consumer and a butyrate producer, the effects on goblet cells and mucin glycosylation were diminished. F. prausnitzii, by attenuating the effects of B. thetaiotaomicron on mucus, may help the epithelium to maintain appropriate proportions of different cell types of the secretory lineage. Using a mucus-producing cell line, we showed that acetate up-regulated KLF4, a transcription factor involved in goblet cell differentiation. CONCLUSIONS: B. thetaiotaomicron and F. prausnitzii, which are metabolically complementary, modulate, in vivo, the intestinal mucus barrier by modifying goblet cells and mucin glycosylation. Our study reveals the importance of the balance between two main commensal bacteria in maintaining colonic epithelial homeostasis via their respective effects on mucus.


Subject(s)
Bacteroides/physiology , Colon/microbiology , Goblet Cells/microbiology , Intestinal Mucosa/microbiology , Mucus/metabolism , Polysaccharides/biosynthesis , Ruminococcus/physiology , Acetates/metabolism , Animals , Bacteroides/ultrastructure , Bacteroides Infections/microbiology , Bacteroides Infections/pathology , Cell Differentiation , Colon/metabolism , Colon/pathology , Disease Models, Animal , Gene Expression Regulation , Germ-Free Life , Glycosylation , Goblet Cells/metabolism , Goblet Cells/pathology , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/pathology , HT29 Cells , Host-Pathogen Interactions/genetics , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Kruppel-Like Factor 4 , Mucus/microbiology , Rats , Signal Transduction , Time Factors
13.
Nat Microbiol ; 9(7): 1725-1737, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38858595

ABSTRACT

Pseudomonas aeruginosa, a leading cause of severe hospital-acquired pneumonia, causes infections with up to 50% mortality rates in mechanically ventilated patients. Despite some knowledge of virulence factors involved, it remains unclear how P. aeruginosa disseminates on mucosal surfaces and invades the tissue barrier. Using infection of human respiratory epithelium organoids, here we observed that P. aeruginosa colonization of apical surfaces is promoted by cyclic di-GMP-dependent asymmetric division. Infection with mutant strains revealed that Type 6 Secretion System activities promote preferential invasion of goblet cells. Type 3 Secretion System activity by intracellular bacteria induced goblet cell death and expulsion, leading to epithelial rupture which increased bacterial translocation and dissemination to the basolateral epithelium. These findings show that under physiological conditions, P. aeruginosa uses coordinated activity of a specific combination of virulence factors and behaviours to invade goblet cells and breach the epithelial barrier from within, revealing mechanistic insight into lung infection dynamics.


Subject(s)
Goblet Cells , Pseudomonas Infections , Pseudomonas aeruginosa , Respiratory Mucosa , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/physiology , Goblet Cells/microbiology , Goblet Cells/metabolism , Humans , Respiratory Mucosa/microbiology , Respiratory Mucosa/cytology , Pseudomonas Infections/microbiology , Type VI Secretion Systems/genetics , Type VI Secretion Systems/metabolism , Virulence Factors/metabolism , Virulence Factors/genetics , Type III Secretion Systems/metabolism , Type III Secretion Systems/genetics , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Organoids/microbiology , Bacterial Translocation
14.
Gastroenterology ; 143(3): 708-718.e5, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22796522

ABSTRACT

BACKGROUND & AIMS: Little is known about factors that regulate intestinal epithelial differentiation; microbial recognition receptors such as Toll-like receptor (TLR)4 might be involved. We investigated whether intestinal TLR4 regulates epithelial differentiation and is involved in development of necrotizing enterocolitis (NEC) of the immature intestine. METHODS: Mice with conditional disruption of TLR4 in the intestinal epithelium and TLR4 knockout (TLR4(-/-)) mice were generated by breeding TLR4(loxp/loxp) mice with villin-cre and Ella-cre, respectively. Enterocytes that did not express or overexpressed TLR4 were created by lentiviral or adenoviral transduction. Intestinal organoids were cultured on tissue matrices. Bile acids were measured by colorimetric assays, and microbial composition was determined by 16S pyrosequencing. NEC was induced in 7- to 10-day-old mice by induction of hypoxia twice daily for 4 days. RESULTS: TLR4(-/-) mice and mice with enterocyte-specific deletion of TLR4 were protected from NEC; epithelial differentiation into goblet cells was increased via suppressed Notch signaling in the small intestinal epithelium. TLR4 also regulates differentiation of goblet cells in intestinal organoid and enterocyte cell cultures; differentiation was increased on deletion of TLR4 and restored when TLR4 was expressed ectopically. TLR4 signaling via Notch was increased in intestinal tissue samples from patients with NEC, and numbers of goblet cells were reduced. 16S pyrosequencing revealed that wild-type and TLR4-deficient mice had similar microbial profiles; increased numbers of goblet cells were observed in mice given antibiotics. TLR4 deficiency reduced levels of luminal bile acids in vivo, and addition of bile acids to TLR4-deficient cell cultures prevented differentiation of goblet cells. CONCLUSIONS: TLR4 signaling and Notch are increased in intestinal tissues of patients with NEC and required for induction of NEC in mice. TLR4 prevents goblet cell differentiation, independently of the microbiota. Bile acids might initiate goblet cell development.


Subject(s)
Cell Differentiation , Enterocolitis, Necrotizing/metabolism , Goblet Cells/metabolism , Intestine, Small/metabolism , Toll-Like Receptor 4/metabolism , Animals , Animals, Newborn , Bile Acids and Salts/metabolism , Cell Line , Disease Models, Animal , Enterocolitis, Necrotizing/etiology , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/pathology , Enterocolitis, Necrotizing/prevention & control , Goblet Cells/microbiology , Goblet Cells/pathology , Humans , Hypoxia/complications , Infant Formula , Infant, Newborn , Intestine, Small/microbiology , Intestine, Small/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organoids , RNA Interference , Rats , Receptors, Notch/metabolism , Signal Transduction , Tissue Culture Techniques , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Transfection
15.
Cell Microbiol ; 14(3): 401-15, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22103442

ABSTRACT

The redox-active exotoxin pyocyanin (PCN) can be recovered in 100 µM concentrations in the sputa of bronchiectasis patients chronically infected with Pseudomonas aeruginosa (PA). However, the importance of PCN within bronchiectatic airways colonized by PA remains unrecognized. Recently, we have shown that PCN is required for chronic PA lung infection in mice, and that chronic instillation of PCN induces goblet cell hyperplasia (GCH), pulmonary fibrosis, emphysema and influx of immune cells in mouse airways. Many of these pathological features are strikingly similar to the mouse airways devoid of functional FoxA2, a transcriptional repressor of GCH and mucus biosynthesis. In this study, we postulate that PCN causes and exacerbates GCH and mucus hypersecretion in bronchiectatic airways chronically infected by PA by inactivating FoxA2. We demonstrate that PCN represses the expression of FoxA2 in mouse airways and in bronchial epithelial cells cultured at an air-liquid interface or conventionally, resulting in GCH, increased MUC5B mucin gene expression and mucus hypersecretion. Immunohistochemical and inhibitor studies indicate that PCN upregulates the expression of Stat6 and EGFR, both of which in turn repress the expression of FoxA2. These studies demonstrate that PCN induces GCH and mucus hypersecretion by inactivating FoxA2.


Subject(s)
Goblet Cells/microbiology , Hepatocyte Nuclear Factor 3-beta/genetics , Lung/pathology , Pseudomonas aeruginosa/physiology , Pyocyanine/metabolism , Animals , Cell Line, Tumor , Down-Regulation , ErbB Receptors/metabolism , Goblet Cells/metabolism , Goblet Cells/pathology , Hepatocyte Nuclear Factor 3-beta/metabolism , Host-Pathogen Interactions , Humans , Hyperplasia , Lung/microbiology , Metaplasia , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-5B/genetics , Mucin-5B/metabolism , Mucus/metabolism , Pseudomonas aeruginosa/metabolism , Pyocyanine/pharmacology , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction , Transcriptional Activation
16.
Sci China Life Sci ; 66(9): 2006-2019, 2023 09.
Article in English | MEDLINE | ID: mdl-37340176

ABSTRACT

Iron is an important micronutrient that plays a vital role in host defenses and bacterial pathogenicity. As iron treatments increase the risk of infection by stimulating the growth and virulence of bacterial pathogens, their roles in anti-infection immunity have frequently been underestimated. To estimate whether adequate dietary iron intake would help defend against pathogenic bacterial infection, mice were fed iron-deficient (2 mg kg-1 feed), iron-sufficient (35 mg kg-1 feed), or iron-enriched diet (350 mg kg-1 feed) for 12 weeks, followed by oral infection with Salmonella typhimurium. Our results revealed that dietary iron intake improved mucus layer function and decelerated the invasion of the pathogenic bacteria, Salmonella typhimurium. Positive correlations between serum iron and the number of goblet cells and mucin2 were found in response to total iron intake in mice. Unabsorbed iron in the intestinal tract affected the gut microbiota composition, and the abundance of Bacteroidales, family Muribaculaceae, was positively correlated with their mucin2 expression. However, the results from antibiotic-treated mice showed that the dietary iron-regulated mucin layer function was not microbial-dependent. Furthermore, in vitro studies revealed that ferric citrate directly induced mucin2 expression and promoted the proliferation of goblet cells in both ileal and colonic organoids. Thus, dietary iron intake improves serum iron levels, regulates goblet cell regeneration and mucin layer function, and plays a positive role in the prevention of pathogenic bacteria.


Subject(s)
Goblet Cells , Iron, Dietary , Animals , Mice , Goblet Cells/metabolism , Goblet Cells/microbiology , Goblet Cells/pathology , Iron, Dietary/metabolism , Intestinal Mucosa/metabolism , Salmonella typhimurium/metabolism , Mucins/metabolism , Iron/metabolism , Bacteria/metabolism
17.
Poult Sci ; 91(6): 1335-41, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22582290

ABSTRACT

Clostridial infection of the intestine can result in necrotic enteritis (NE), compromising production and health of poultry. Mucins play a major role in protecting the intestinal epithelium from infection. The relative roles of different mucins in gut pathology following bacterial challenge are unclear. This study was designed to quantify the expression of mucin and mucin-related genes, using intestinal samples from an NE challenge trial where birds were fed diets with or without in-feed antimicrobials. A method for quantifying mucin gene expression was established using a suite of reference genes to normalize expression data. This method was then used to quantify the expression of 11 candidate genes involved in mucin, inflammatory cytokine, or growth factor biosynthesis (IL-18, KGF, TLR4, TFF2, TNF-α, MUC2, MUC4, MUC5ac, MUC5b, MUC13, and MUC16). The only genes that were differentially expressed in the intestine among treatment groups were MUC2, MUC13, and MUC5ac. Expression of MUC2 and MUC13 was depressed by co-challenge with Eimeria spp. and Clostridium perfringens. Antimicrobial treatment prevented an NE-induced decrease in MUC2 expression but did not affect MUC13. The expression of MUC5ac was elevated in birds challenged with Eimeria spp./C. perfringens compared with unchallenged controls and antimicrobial treatment. Changes to MUC gene expression in challenged birds is most likely a consequence of severe necrosis of the jejunal mucosa.


Subject(s)
Chickens , Clostridium Infections/veterinary , Coccidiosis/veterinary , Enteritis/veterinary , Gene Expression Regulation , Mucins/metabolism , Poultry Diseases/immunology , Animals , Clostridium Infections/immunology , Clostridium Infections/microbiology , Clostridium perfringens/physiology , Coccidiosis/immunology , Coccidiosis/parasitology , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Eimeria/physiology , Enteritis/immunology , Enteritis/microbiology , Enteritis/parasitology , Goblet Cells/immunology , Goblet Cells/metabolism , Goblet Cells/microbiology , Goblet Cells/parasitology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/microbiology , Intestines/parasitology , Male , Mucins/genetics , Necrosis/immunology , Necrosis/microbiology , Necrosis/parasitology , Necrosis/veterinary , Poultry Diseases/microbiology , Poultry Diseases/parasitology , Real-Time Polymerase Chain Reaction/veterinary , Sequence Analysis, DNA
18.
Eksp Klin Gastroenterol ; (9): 54-8, 2011.
Article in Russian | MEDLINE | ID: mdl-22629777

ABSTRACT

BACKGROUND AND AIMS: Hypersusceptibility to pathogens, including enterobacteria is the most serious late result of splenectomy. The aim of these studies was to give the morphofunctional characteristics of large bowel and to analyze luminal bacterial ecosystem disturbances as late results of splenectomy. METHODS: Fourty male mice Balb/c with body mass 18-20 g were divided in random manner: control group consisted of ten intact mice, 15 mice in each of two experimental groups subjected to splenectomy. All animal were carried in standard housing conditions and feeding and were upset of the experiment on the 30th (experimental group I) and 60th (experimental group II) days after the splenectomy. For the histological investigation the pieces of colon were taken and processed in standard histological methods. The blood out of cardial cavity was sampled for estimation of the endotoxin level; faeces for the analyzing of luminal bacterial ecosystem disturbances (dysbios) were taken out of the rectum. RESULTS: At the 30th day after splenectomy the pattern of chronic catarrhal colitis was at the histological the stroma by lymphocytes and neutrophils. At the 60th day at the histological slides the increasing of mitotic activity of epithelial cells in crypts of the colon and activation of the inflammative infiltration were observed. The endotoxin level half as large again in comparison with the control one at the 30th day. The I-II stages of dysbiotic disturbances were recognizes by bacterial methods at the 30th day with the following development up the II stage at the 60th day. CONCLUSIONS: The chronic catarrhal colitis with accompanying disturbance of luminal bacterial ecosystem (dysbiotic state of Stages I-II) and increasing og the endotoxin level in the blood are among other late results of splenectomy in mice Balb/c model


Subject(s)
Bacteria , Colitis , Colon/microbiology , Colon/pathology , Splenectomy , Animals , Chronic Disease , Colitis/etiology , Colitis/microbiology , Colitis/pathology , Goblet Cells/microbiology , Goblet Cells/pathology , Lymphocytes/microbiology , Lymphocytes/pathology , Male , Mice , Mice, Inbred BALB C , Neutrophil Infiltration , Neutrophils/microbiology , Neutrophils/pathology
19.
Front Immunol ; 12: 636198, 2021.
Article in English | MEDLINE | ID: mdl-33841417

ABSTRACT

Food allergy is an emerging epidemic, and the underlying mechanisms are not well defined partly due to the lack of robust adjuvant free experimental models of dietary antigen sensitization. As housing mice at thermoneutrality (Tn) - the temperature of metabolic homeostasis (26-30°C) - has been shown to improve modeling various human diseases involved in inflammation, we tested the impact of Tn housing on an experimental model of food sensitization. Here we demonstrate that WT BALB/c mice housed under standard temperature (18-20°C, Ts) conditions translocated the luminal antigens in the small intestine (SI) across the epithelium via goblet cell antigen passages (GAPs). In contrast, food allergy sensitive Il4raF709 mice housed under standard temperature conditions translocated the luminal antigens in the SI across the epithelium via secretory antigen passages (SAPs). Activation of SI antigen passages and oral challenge of Il4raF709 mice with egg allergens at standard temperature predisposed Il4raF709 mice to develop an anaphylactic reaction. Housing Il4raF709 mice at Tn altered systemic type 2 cytokine, IL-4, and the landscape of SI antigen passage patterning (villus and crypt involvement). Activation of SI antigen passages and oral challenge of Il4raF709 mice with egg antigen under Tn conditions led to the robust induction of egg-specific IgE and development of food-induced mast cell activation and hypovolemic shock. Similarly, Tn housing of WT BALB/c mice altered the cellular patterning of SI antigen passage (GAPs to SAPs). Activation of SI antigen passages and the oral challenge of WT BALB/c mice with egg antigen led to systemic reactivity to egg and mast cell activation. Together these data demonstrate that Tn housing alters antigen passage cellular patterning and landscape, and concurrent oral exposure of egg antigens and SAP activation is sufficient to induce oral antigen sensitization.


Subject(s)
Allergens/metabolism , Anaphylaxis/metabolism , Egg Hypersensitivity/metabolism , Egg Proteins/metabolism , Housing, Animal , Intestine, Small/metabolism , Temperature , Administration, Oral , Allergens/administration & dosage , Allergens/immunology , Anaphylaxis/immunology , Anaphylaxis/microbiology , Animals , Disease Models, Animal , Egg Hypersensitivity/immunology , Egg Hypersensitivity/microbiology , Egg Proteins/administration & dosage , Egg Proteins/immunology , Gastrointestinal Microbiome , Goblet Cells/immunology , Goblet Cells/metabolism , Goblet Cells/microbiology , Intestine, Small/immunology , Intestine, Small/microbiology , Mast Cells/immunology , Mast Cells/metabolism , Mice, Inbred BALB C , Mice, Knockout , Permeability , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
20.
Biomed Pharmacother ; 144: 112253, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34607106

ABSTRACT

Iron supplementation is necessary for the treatment of anemia, one of the most frequent complications in inflammatory bowel disease (IBD). However, oral iron supplementation leads to an exacerbation of intestinal inflammation. Gut barrier plays a key role in the pathogenesis of IBD. The aim of this study was to characterize the interrelationship between systemic iron, intestinal barrier and the development of intestinal inflammation in a dextran sulfate sodium (DSS) induced experimental colitis mice model. We found that DSS-treated mice developed severe inflammation of colon, but became much healthy when intraperitoneal injection with iron. Iron supplementation alleviated colonic and systemic inflammation by lower histological scores, restorative morphology of colonic villi, and reduced expression of pro-inflammatory cytokines. Moreover, intraperitoneal supplementation of iron enhanced intestinal barrier function by upregulating the colonic expressions of tight junction proteins, restoring intestinal immune homeostasis by regulating immune cell infiltration and T lymphocyte subsets, and increasing mucous secretion of goblet cells in the colon. High-throughput sequencing of fecal 16 S rRNA showed that iron injection significantly increased the relative abundance of Bacteroidetes, which was suppressed in the gut microbiota of DSS-induced colitis mice. These results provided evidences supporting the protective effects of systemic iron repletion by intraperitoneal injection of iron on intestinal barrier functions. The finding highlights a novel approach for the treatment of IBD with iron injection therapy.


Subject(s)
Colitis/drug therapy , Colon/drug effects , Dietary Supplements , Goblet Cells/drug effects , Intestinal Mucosa/drug effects , Iron-Dextran Complex/administration & dosage , Tight Junction Proteins/metabolism , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Colitis/chemically induced , Colitis/metabolism , Colitis/microbiology , Colon/metabolism , Colon/microbiology , Dextran Sulfate , Disease Models, Animal , Dysbiosis , Gastrointestinal Microbiome/drug effects , Goblet Cells/metabolism , Goblet Cells/microbiology , Injections, Intraperitoneal , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mice, Inbred C57BL , Permeability , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/microbiology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL