Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(7)2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38612809

RESUMO

Chorioamnionitis is a risk factor for necrotizing enterocolitis (NEC). Ureaplasma parvum (UP) is clinically the most isolated microorganism in chorioamnionitis, but its pathogenicity remains debated. Chorioamnionitis is associated with ileal barrier changes, but colonic barrier alterations, including those of the mucus barrier, remain under-investigated, despite their importance in NEC pathophysiology. Therefore, in this study, the hypothesis that antenatal UP exposure disturbs colonic mucus barrier integrity, thereby potentially contributing to NEC pathogenesis, was investigated. In an established ovine chorioamnionitis model, lambs were intra-amniotically exposed to UP or saline for 7 d from 122 to 129 d gestational age. Thereafter, colonic mucus layer thickness and functional integrity, underlying mechanisms, including endoplasmic reticulum (ER) stress and redox status, and cellular morphology by transmission electron microscopy were studied. The clinical significance of the experimental findings was verified by examining colon samples from NEC patients and controls. UP-exposed lambs have a thicker but dysfunctional colonic mucus layer in which bacteria-sized beads reach the intestinal epithelium, indicating undesired bacterial contact with the epithelium. This is paralleled by disturbed goblet cell MUC2 folding, pro-apoptotic ER stress and signs of mitochondrial dysfunction in the colonic epithelium. Importantly, the colonic epithelium from human NEC patients showed comparable mitochondrial aberrations, indicating that NEC-associated intestinal barrier injury already occurs during chorioamnionitis. This study underlines the pathogenic potential of UP during pregnancy; it demonstrates that antenatal UP infection leads to severe colonic mucus barrier deficits, providing a mechanistic link between antenatal infections and postnatal NEC development.


Assuntos
Corioamnionite , Infecções por Ureaplasma , Gravidez , Ovinos , Animais , Humanos , Feminino , Recém-Nascido , Infecções por Ureaplasma/complicações , Intestinos , Causalidade , Muco
2.
Nat Commun ; 14(1): 3652, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37339972

RESUMO

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.


Assuntos
Mucosa Intestinal , Intestinos , Proteínas de Membrana , Animais , Camundongos , Microbioma Gastrointestinal , Glicosilação , Células Caliciformes/metabolismo , Complexo de Golgi/metabolismo , Homeostase , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Intestinos/metabolismo , Proteínas de Membrana/metabolismo , Muco , Celulas de Paneth/metabolismo
3.
Cell Rep ; 42(2): 112084, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36753416

RESUMO

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.


Assuntos
Mucosa Intestinal , Jejuno , Mucina-2 , Animais , Camundongos , Dieta Ocidental , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Intestino Delgado , Mucina-2/genética , Mucina-2/metabolismo , Muco , Citrobacter rodentium/fisiologia
4.
Nat Commun ; 13(1): 45, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35017479

RESUMO

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.


Assuntos
Colo/metabolismo , Muco/metabolismo , Transglutaminases/metabolismo , Animais , Colite/etiologia , Colite/metabolismo , Camundongos , Mucina-2/metabolismo
5.
Cell Rep ; 35(1): 108949, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33826887

RESUMO

The colon epithelium is a primary point of interaction with the microbiome and is regenerated by a few rapidly cycling colonic stem cells (CSCs). CSC self-renewal and proliferation are regulated by growth factors and the presence of bacteria. However, the molecular link connecting the diverse inputs that maintain CSC homeostasis remains largely unknown. We report that CSC proliferation is mediated by redox-dependent activation of epidermal growth factor receptor (EGFR) signaling via NADPH oxidase 1 (NOX1). NOX1 expression is CSC specific and is restricted to proliferative CSCs. In the absence of NOX1, CSCs fail to generate ROS and have a reduced proliferation rate. NOX1 expression is regulated by Toll-like receptor activation in response to the microbiota and serves to link CSC proliferation with the presence of bacterial components in the crypt. The TLR-NOX1-EGFR axis is therefore a critical redox signaling node in CSCs facilitating the quiescent-proliferation transition and responds to the microbiome to maintain colon homeostasis.


Assuntos
Colo/citologia , Colo/microbiologia , Receptores ErbB/metabolismo , Microbioma Gastrointestinal , NADPH Oxidase 1/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Receptores Toll-Like/metabolismo , Animais , Bactérias/crescimento & desenvolvimento , Biomarcadores/metabolismo , Proliferação de Células , Contagem de Colônia Microbiana , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/metabolismo
6.
Science ; 372(6539)2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33859001

RESUMO

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.


Assuntos
Colo/citologia , Células Caliciformes/fisiologia , Mucosa Intestinal/citologia , Muco/fisiologia , Animais , Diferenciação Celular , Colite/induzido quimicamente , Colite/fisiopatologia , Colite Ulcerativa/patologia , Colite Ulcerativa/fisiopatologia , Colo/fisiologia , Células Caliciformes/citologia , Humanos , Mucosa Intestinal/fisiologia , Intestino Delgado/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-ets/genética , Transcriptoma
7.
Science ; 370(6515): 402-403, 2020 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-33093095
8.
J Biol Chem ; 295(46): 15712-15726, 2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-32900852

RESUMO

The intestinal mucus layer is a physical barrier separating the tremendous number of gut bacteria from the host epithelium. Defects in the mucus layer have been linked to metabolic diseases, but previous studies predominantly investigated mucus function during high-caloric/low-fiber dietary interventions, thus making it difficult to separate effects mediated directly through diet quality from potential obesity-dependent effects. As such, we decided to examine mucus function in mouse models with metabolic disease to distinguish these factors. Here we show that, in contrast to their lean littermates, genetically obese (ob/ob) mice have a defective inner colonic mucus layer that is characterized by increased penetrability and a reduced mucus growth rate. Exploiting the coprophagic behavior of mice, we next co-housed ob/ob and lean mice to investigate if the gut microbiota contributed to these phenotypes. Co-housing rescued the defect of the mucus growth rate, whereas mucus penetrability displayed an intermediate phenotype in both mouse groups. Of note, non-obese diabetic mice with high blood glucose levels displayed a healthy colonic mucus barrier, indicating that the mucus defect is obesity- rather than glucose-mediated. Thus, our data suggest that the gut microbiota community of obesity-prone mice may regulate obesity-associated defects in the colonic mucosal barrier, even in the presence of dietary fiber.


Assuntos
Microbioma Gastrointestinal , Mucosa Intestinal/metabolismo , Obesidade/patologia , Animais , Glicemia/análise , Colo/metabolismo , Colo/microbiologia , Colo/patologia , Feminino , Glucose/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Obesos , Obesidade/genética , Fenótipo
9.
J Exp Med ; 216(11): 2602-2618, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31420376

RESUMO

The inner mucus layer (IML) is a critical barrier that protects the colonic epithelium from luminal threats and inflammatory bowel disease. Innate immune signaling is thought to regulate IML formation via goblet cell Nlrp6 inflammasome activity that controls secretion of the mucus structural component Muc2. We report that isolated colonic goblet cells express components of several inflammasomes; however, analysis of IML properties in multiple inflammasome-deficient mice, including littermate-controlled Nlrp6-/- , detect a functional IML barrier in all strains. Analysis of mice lacking inflammasome substrate cytokines identifies a defective IML in Il18-/- mice, but this phenotype is ultimately traced to a microbiota-driven, Il18-independent effect. Analysis of phenotypic transfer between IML-deficient and IML-intact mice finds that the Bacteroidales family S24-7 (Muribaculaceae) and genus Adlercrutzia consistently positively covary with IML barrier function. Together, our results demonstrate that baseline IML formation and function is independent of inflammasome activity and highlights the role of the microbiota in determining IML barrier function.


Assuntos
Colo/imunologia , Células Caliciformes/imunologia , Inflamassomos/imunologia , Mucosa Intestinal/imunologia , Muco/imunologia , Receptores de Superfície Celular/imunologia , Animais , Colo/metabolismo , Colo/microbiologia , Microbioma Gastrointestinal/imunologia , Células Caliciformes/metabolismo , Células Caliciformes/microbiologia , Inflamassomos/genética , Inflamassomos/metabolismo , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Interleucina-18/genética , Interleucina-18/imunologia , Interleucina-18/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mucina-2/imunologia , Mucina-2/metabolismo , Muco/metabolismo , Muco/microbiologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/imunologia
10.
Infect Immun ; 87(5)2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30833331

RESUMO

Gastrointestinal (GI) colonization of 2-day-old (P2) rat pups with Escherichia coli K1 results in translocation of the colonizing bacteria across the small intestine, bacteremia, and invasion of the meninges, with animals frequently succumbing to lethal infection. Infection, but not colonization, is strongly age dependent; pups become progressively less susceptible to infection over the P2-to-P9 period. Colonization leads to strong downregulation of the gene encoding trefoil factor 2 (Tff2), preventing maturation of the protective mucus barrier in the small intestine. Trefoil factors promote mucosal homeostasis. We investigated the contribution of Tff2 to protection of the neonatal rat from E. coli K1 bacteremia and tissue invasion. Deletion of tff2, using clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9, sensitized P9 pups to E. coli K1 bacteremia. There were no differences between tff2-/- homozygotes and the wild type with regard to the dynamics of GI colonization. Loss of the capacity to elaborate Tff2 did not impact GI tract integrity or the thickness of the small-intestinal mucus layer but, in contrast to P9 wild-type pups, enabled E. coli K1 bacteria to gain access to epithelial surfaces in the distal region of the small intestine and exploit an intracellular route across the epithelial monolayer to enter the blood circulation via the mesenteric lymphatic system. Although primarily associated with the mammalian gastric mucosa, we conclude that loss of Tff2 in the developing neonatal small intestine enables the opportunistic neonatal pathogen E. coli K1 to enter the compromised mucus layer in the distal small intestine prior to systemic invasion and infection.


Assuntos
Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Escherichia coli/patogenicidade , Imunidade Inata/imunologia , Sepse Neonatal/imunologia , Fator Trefoil-2/imunologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Humanos , Ratos
11.
EBioMedicine ; 33: 134-143, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29885864

RESUMO

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.


Assuntos
Canais de Cloreto/metabolismo , Colo/metabolismo , Muco/metabolismo , Animais , Canais de Cloreto/genética , Homeostase , Humanos , Metaloproteases/metabolismo , Camundongos , Camundongos Knockout , Proteólise , Proteoma/química
12.
Cell Host Microbe ; 23(1): 27-40.e7, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29276171

RESUMO

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.


Assuntos
Bifidobacterium longum/metabolismo , Colo/microbiologia , Dieta Ocidental/efeitos adversos , Fibras na Dieta/uso terapêutico , Transplante de Microbiota Fecal , Mucosa Intestinal/microbiologia , Animais , Colo/patologia , Suplementos Nutricionais , Microbioma Gastrointestinal/fisiologia , Mucosa Intestinal/patologia , Inulina/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/patologia
13.
Sci Rep ; 7(1): 83, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28250440

RESUMO

The strong age dependency of neonatal systemic infection with Escherichia coli K1 can be replicated in the neonatal rat. Gastrointestinal (GI) colonization of two-day-old (P2) rats leads to invasion of the blood within 48 h of initiation of colonization; pups become progressively less susceptible to infection over the P2-P9 period. We show that, in animals colonized at P2 but not at P9, E. coli K1 bacteria gain access to the enterocyte surface in the mid-region of the small intestine and translocate through the epithelial cell monolayer by an intracellular pathway to the submucosa. In this region of the GI tract, the protective mucus barrier is poorly developed but matures to full thickness over P2-P9, coincident with the development of resistance to invasion. At P9, E. coli K1 bacteria are physically separated from villi by the mucus layer and their numbers controlled by mucus-embedded antimicrobial peptides, preventing invasion of host tissues.


Assuntos
Infecções por Escherichia coli/microbiologia , Escherichia coli/fisiologia , Mucosa Intestinal/crescimento & desenvolvimento , Intestino Delgado/citologia , Animais , Animais Recém-Nascidos , Sangue/microbiologia , Células Cultivadas , Modelos Animais de Doenças , Mucosa Intestinal/microbiologia , Intestino Delgado/crescimento & desenvolvimento , Intestino Delgado/microbiologia , Especificidade de Órgãos , Celulas de Paneth/citologia , Celulas de Paneth/microbiologia , Ratos
14.
Proc Natl Acad Sci U S A ; 113(48): 13833-13838, 2016 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-27849619

RESUMO

The distal colon functions as a bioreactor and harbors an enormous amount of bacteria in a mutualistic relationship with the host. The microbiota have to be kept at a safe distance to prevent inflammation, something that is achieved by a dense inner mucus layer that lines the epithelial cells. The large polymeric nets made up by the heavily O-glycosylated MUC2 mucin forms this physical barrier. Proteomic analyses of mucus have identified the lectin-like protein ZG16 (zymogen granulae protein 16) as an abundant mucus component. To elucidate the function of ZG16, we generated recombinant ZG16 and studied Zg16-/- mice. ZG16 bound to and aggregated Gram-positive bacteria via binding to the bacterial cell wall peptidoglycan. Zg16-/- mice have a distal colon mucus layer with normal thickness, but with bacteria closer to the epithelium. Using distal colon explants mounted in a horizontal perfusion chamber we demonstrated that treatment of bacteria with recombinant ZG16 hindered bacterial penetration into the mucus. The inner colon mucus of Zg16-/- animals had a higher load of Gram-positive bacteria and showed bacteria with higher motility in the mucus close to the host epithelium compared with cohoused littermate Zg16+/+ The more penetrable Zg16-/- mucus allowed Gram-positive bacteria to translocate to systemic tissues. Viable bacteria were found in spleen and were associated with increased abdominal fat pad mass in Zg16-/- animals. The function of ZG16 reveals a mechanism for keeping bacteria further away from the host colon epithelium.


Assuntos
Bactérias Gram-Positivas/genética , Lectinas/genética , Proteínas de Membrana/genética , Proteômica , Animais , Colo/metabolismo , Colo/microbiologia , Sistema Digestório/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Glicosilação , Bactérias Gram-Positivas/metabolismo , Interações Hospedeiro-Patógeno/genética , Lectinas/metabolismo , Camundongos , Camundongos Knockout , Muco/metabolismo , Muco/microbiologia , Simbiose/genética
15.
Science ; 352(6293): 1535-42, 2016 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-27339979

RESUMO

Innate immune signaling pathways contribute to the protection of host tissue when bacterially challenged. Colonic goblet cells are responsible for generating the two mucus layers that physically separate the luminal microbiota from the host epithelium. Analysis of colonic tissues from multiple mouse strains allowed us to identify a "sentinel" goblet cell (senGC) localized to the colonic crypt entrance. This cell nonspecifically endocytoses and reacts to the TLR2/1, TLR4, and TLR5 ligands by activating the Nlrp6 inflammasome downstream of TLR- and MyD88-dependent Nox/Duox reactive oxygen species synthesis. This triggers calcium ion-dependent compound exocytosis of Muc2 mucin from the senGC and generates an intercellular gap junction signal; in turn, this signal induces Muc2 secretion from adjacent goblet cells in the upper crypt, which expels bacteria. Thus, senGCs guard and protect the colonic crypt from bacterial intruders that have penetrated the inner mucus layer.


Assuntos
Colo/imunologia , Colo/microbiologia , Microbioma Gastrointestinal/imunologia , Células Caliciformes/imunologia , Mucina-2/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Bactérias/imunologia , Cálcio/metabolismo , Endocitose/imunologia , Imunidade Inata , Inflamassomos/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Superfície Celular/genética , Transdução de Sinais , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptor 5 Toll-Like/metabolismo
16.
PLoS One ; 10(7): e0131991, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26162072

RESUMO

The secreted, goblet cell-derived protein Clca1 (chloride channel regulator, calcium-activated-1) has been linked to diseases with mucus overproduction, including asthma and cystic fibrosis. In the intestine Clca1 is found in the mucus with an abundance and expression pattern similar to Muc2, the major structural mucus component. We hypothesized that Clca1 is required for the synthesis, structure or barrier function of intestinal mucus and therefore compared wild type and Clca1-deficient mice under naive and at various time points of DSS (dextran sodium sulfate)-challenged conditions. The mucus phenotype in Clca1-deficient compared to wild type mice was systematically characterized by assessment of the mucus protein composition using proteomics, immunofluorescence and expression analysis of selected mucin genes on mRNA level. Mucus barrier integrity was assessed in-vivo by analysis of bacterial penetration into the mucus and translocation into sentinel organs combined analysis of the fecal microbiota and ex-vivo by assessment of mucus penetrability using beads. All of these assays revealed no relevant differences between wild type and Clca1-deficient mice under steady state or DSS-challenged conditions in mouse colon. Clca1 is not required for mucus synthesis, structure and barrier function in the murine colon.


Assuntos
Canais de Cloreto/fisiologia , Colite/metabolismo , Colo/metabolismo , Muco/metabolismo , Animais , Colite/induzido quimicamente , Colite/patologia , Colo/microbiologia , Colo/patologia , Sulfato de Dextrana , Fezes/microbiologia , Expressão Gênica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microbiota , Mucina-2/metabolismo , Permeabilidade , RNA Ribossômico 16S/genética
17.
J Vis Exp ; (92): e52018, 2014 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-25408299

RESUMO

Investigation of the interactions between animal host and bacterial pathogen is only meaningful if the infection model employed replicates the principal features of the natural infection. This protocol describes procedures for the establishment and evaluation of systemic infection due to neuropathogenic Escherichia coli K1 in the neonatal rat. Colonization of the gastrointestinal tract leads to dissemination of the pathogen along the gut-lymph-blood-brain course of infection and the model displays strong age dependency. A strain of E. coli O18:K1 with enhanced virulence for the neonatal rat produces exceptionally high rates of colonization, translocation to the blood compartment and invasion of the meninges following transit through the choroid plexus. As in the human host, penetration of the central nervous system is accompanied by local inflammation and an invariably lethal outcome. The model is of proven utility for studies of the mechanism of pathogenesis, for evaluation of therapeutic interventions and for assessment of bacterial virulence.


Assuntos
Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Escherichia coli/patogenicidade , Animais , Animais Recém-Nascidos , Plexo Corióideo/microbiologia , Feminino , Ratos , Virulência
18.
Immunol Rev ; 260(1): 8-20, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24942678

RESUMO

The gastrointestinal tract is covered by mucus that has different properties in the stomach, small intestine, and colon. The large highly glycosylated gel-forming mucins MUC2 and MUC5AC are the major components of the mucus in the intestine and stomach, respectively. In the small intestine, mucus limits the number of bacteria that can reach the epithelium and the Peyer's patches. In the large intestine, the inner mucus layer separates the commensal bacteria from the host epithelium. The outer colonic mucus layer is the natural habitat for the commensal bacteria. The intestinal goblet cells secrete not only the MUC2 mucin but also a number of typical mucus components: CLCA1, FCGBP, AGR2, ZG16, and TFF3. The goblet cells have recently been shown to have a novel gate-keeping role for the presentation of oral antigens to the immune system. Goblet cells deliver small intestinal luminal material to the lamina propria dendritic cells of the tolerogenic CD103(+) type. In addition to the gel-forming mucins, the transmembrane mucins MUC3, MUC12, and MUC17 form the enterocyte glycocalyx that can reach about a micrometer out from the brush border. The MUC17 mucin can shuttle from a surface to an intracellular vesicle localization, suggesting that enterocytes might control and report epithelial microbial challenge. There is communication not only from the epithelial cells to the immune system but also in the opposite direction. One example of this is IL10 that can affect and improve the properties of the inner colonic mucus layer. The mucus and epithelial cells of the gastrointestinal tract are the primary gate keepers and controllers of bacterial interactions with the host immune system, but our understanding of this relationship is still in its infancy.


Assuntos
Enterócitos/fisiologia , Trato Gastrointestinal/imunologia , Células Caliciformes/fisiologia , Mucinas/fisiologia , Mucosa/imunologia , Muco/fisiologia , Animais , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Humanos , Sistema Imunitário , Mucosa/metabolismo , Mucosa/microbiologia , Muco/química , Muco/microbiologia , Nódulos Linfáticos Agregados/imunologia
19.
Infect Immun ; 81(9): 3264-75, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23798529

RESUMO

Two-day-old (P2), but not 9-day-old (P9), rat pups are susceptible to systemic infection following gastrointestinal colonization by Escherichia coli K1. Age dependency reflects the capacity of colonizing K1 to translocate from gastrointestinal (GI) tract to blood. A complex GI microbiota developed by P2, showed little variation over P2 to P9, and did not prevent stable K1 colonization. Substantial developmental expression was observed over P2 to P9, including upregulation of genes encoding components of the small intestinal (α-defensins Defa24 and Defa-rs1) and colonic (trefoil factor Tff2) mucus barrier. K1 colonization modulated expression of these peptides: developmental expression of Tff2 was dysregulated in P2 tissues and was accompanied by a decrease in mucin Muc2. Conversely, α-defensin genes were upregulated in P9 tissues. We propose that incomplete development of the mucus barrier during early neonatal life and the capacity of colonizing K1 to interfere with mucus barrier maturation provide opportunities for neuropathogen translocation into the bloodstream.


Assuntos
Escherichia coli/crescimento & desenvolvimento , Escherichia coli/imunologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Animais , Animais Recém-Nascidos , Colo/imunologia , Colo/metabolismo , Colo/microbiologia , Trato Gastrointestinal/metabolismo , Imunidade Inata/imunologia , Intestino Delgado/metabolismo , Microbiota/genética , Microbiota/imunologia , Mucinas/genética , Mucinas/imunologia , Mucinas/metabolismo , Muco/imunologia , Muco/metabolismo , Muco/microbiologia , Peptídeos/genética , Peptídeos/imunologia , Peptídeos/metabolismo , Ratos , Fator Trefoil-2 , Regulação para Cima/genética , Regulação para Cima/imunologia , alfa-Defensinas/genética , alfa-Defensinas/imunologia , alfa-Defensinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...