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.
Proc Natl Acad Sci U S A ; 120(28): e2301115120, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37399418

RESUMO

Enteric bacterial pathogens pose significant threats to human health; however, the mechanisms by which they infect the mammalian gut in the face of daunting host defenses and an established microbiota remain poorly defined. For the attaching and effacing (A/E) bacterial family member and murine pathogen Citrobacter rodentium, its virulence strategy likely involves metabolic adaptation to the host's intestinal luminal environment, as a necessary precursor to reach and infect the mucosal surface. Suspecting this adaptation involved the intestinal mucus layer, we found that C. rodentium was able to catabolize sialic acid, a monosaccharide derived from mucins, and utilize it as its sole carbon source for growth. Moreover, C. rodentium also sensed and displayed chemotactic activity toward sialic acid. These activities were abolished when the nanT gene, encoding a sialic acid transporter, was deleted (ΔnanT). Correspondingly, the ΔnanT C. rodentium strain was significantly impaired in its ability to colonize the murine intestine. Intriguingly, sialic acid was also found to induce the secretion of two autotransporter proteins, Pic and EspC, which possess mucinolytic and host-adherent properties. As a result, sialic acid enhanced the ability of C. rodentium to degrade intestinal mucus (through Pic), as well as to adhere to intestinal epithelial cells (through EspC). We thus demonstrate that sialic acid, a monosaccharide constituent of the intestinal mucus layer, functions as an important nutrient and a key signal for an A/E bacterial pathogen to escape the colonic lumen and directly infect its host's intestinal mucosa.


Assuntos
Citrobacter rodentium , Infecções por Enterobacteriaceae , Animais , Camundongos , Bactérias , Citrobacter , Infecções por Enterobacteriaceae/microbiologia , Mucosa Intestinal/microbiologia , Mamíferos , Monossacarídeos , Ácido N-Acetilneuramínico
2.
Cell Mol Gastroenterol Hepatol ; 15(2): 425-438, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36244647

RESUMO

BACKGROUND & AIMS: Increased intestinal permeability is seen in a variety of inflammatory conditions such as enteric infections and inflammatory bowel disease. Because barrier function can provide a key biomarker of disease severity, it often is assayed in animal models. A common methodology involves gavaging mice with fluorescein isothiocyanate-conjugated dextran (FITC-D), followed by cardiac puncture to assay plasma fluorescence on a spectrophotometer. Although the FITC-D method is relatively simple, its sensitivity is limited and enables only a single measurement because the test requires killing the subject. Herein, we describe a novel flow cytometry-based method of intestinal permeability measurement based on detection of orally gavaged ovalbumin (OVA) that leaks out of the gut. Our approach uses minute blood volumes collected from the tail vein, permitting repeated testing of the same subject at multiple time points. By comparing this assay against the gold standard FITC-D method, we show the expanded utility of our OVA assay in measuring intestinal permeability. METHODS: We directly compared our OVA assay against the FITC-D assay by co-administering both probes orally to the same animals and subsequently using their respective methodologies to measure intestinal permeability by detecting probe levels in the plasma. Permeability was assessed in mice genetically deficient in intestinal mucus production or glycosylation. In addition, wild-type mice undergoing dextran sodium sulfate-induced colitis or infected by the enteric bacterial pathogen Citrobacter rodentium also were tested. RESULTS: The OVA assay showed very high efficacy in all animal models of intestinal barrier dysfunction tested. Besides identifying intestinal barrier dysfunction in mice with impaired mucin glycosylation, the assay also allowed for repeated tracking of intestinal permeability within the same animal over time, providing data that cannot be easily acquired with other currently applied methods. CONCLUSIONS: The OVA assay is a highly sensitive and effective method of measuring intestinal permeability in mouse models of barrier dysfunction and experimental colitis.


Assuntos
Colite , Dextranos , Camundongos , Animais , Dextranos/efeitos adversos , Mucosa Intestinal , Citometria de Fluxo , Fluoresceína-5-Isotiocianato/efeitos adversos , Colite/induzido quimicamente , Modelos Animais de Doenças , Permeabilidade
3.
PLoS Pathog ; 17(8): e1009719, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34352037

RESUMO

Reducing food intake is a common host response to infection, yet it remains unclear whether fasting is detrimental or beneficial to an infected host. Despite the gastrointestinal tract being the primary site of nutrient uptake and a common route for infection, studies have yet to examine how fasting alters the host's response to an enteric infection. To test this, mice were fasted before and during oral infection with the invasive bacterium Salmonella enterica serovar Typhimurium. Fasting dramatically interrupted infection and subsequent gastroenteritis by suppressing Salmonella's SPI-1 virulence program, preventing invasion of the gut epithelium. Virulence suppression depended on the gut microbiota, as Salmonella's invasion of the epithelium proceeded in fasting gnotobiotic mice. Despite Salmonella's restored virulence within the intestines of gnotobiotic mice, fasting downregulated pro-inflammatory signaling, greatly reducing intestinal pathology. Our study highlights how food intake controls the complex relationship between host, pathogen and gut microbiota during an enteric infection.


Assuntos
Bactérias/crescimento & desenvolvimento , Jejum , Gastroenterite/prevenção & controle , Inflamação/prevenção & controle , Intestinos/imunologia , NF-kappa B/antagonistas & inibidores , Salmonelose Animal/imunologia , Salmonella typhimurium/fisiologia , Animais , Bactérias/imunologia , Bactérias/metabolismo , Feminino , Gastroenterite/imunologia , Gastroenterite/microbiologia , Microbioma Gastrointestinal , Inflamação/imunologia , Inflamação/microbiologia , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Salmonelose Animal/complicações , Salmonelose Animal/microbiologia , Salmonelose Animal/patologia
4.
Sci Rep ; 11(1): 8206, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33859245

RESUMO

Intestinal epithelial cells (IEC) reside in close proximity to the gut microbiota and are hypo-responsive to bacterial products, likely to prevent maladaptive inflammatory responses. This is in part due to their strong expression of Single Ig IL-1 related receptor (SIGIRR), a negative regulator of interleukin (IL)-1 and toll-like receptor signaling. IL-37 is an anti-inflammatory cytokine that inhibits innate signaling in diverse cells by signaling through SIGIRR. Despite the strong expression of SIGIRR by IEC, few studies have examined whether IL-37 can suppress their innate immune signaling. We characterized innate immune responses of human and murine colonoids to bacteria (FliC, LPS) and host (IL-1ß) products and the role of IL-37/SIGIRR in regulating these responses. We demonstrated that human colonoids responded only to FliC, but not to LPS or IL-1ß. While colonoids derived from different donors displayed significant inter-individual variability in the magnitude of their innate responses to FliC stimulation, all colonoids released a variety of chemokines. Interestingly, IL-37 attenuated these responses through inhibition of p38 and NFκB signaling pathways. We determined that this suppression by IL-37 was SIGIRR dependent, in murine organoids. Along with species-specific differences in IEC innate responses, we show that IL-37 can promote IEC hypo-responsiveness by suppressing inflammatory signaling.


Assuntos
Colo/imunologia , Imunidade Inata/genética , Interleucina-1/fisiologia , Organoides/imunologia , Adulto , Animais , Células Cultivadas , Criança , Colo/metabolismo , Colo/patologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Organoides/metabolismo , Organoides/patologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Adulto Jovem
5.
Cell Host Microbe ; 28(6): 771-773, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33301714

RESUMO

To colonize the nutritionally competitive environment of the gut, invading bacteria must utilize unique metabolites. Two reports in this issue of Cell Host & Microbe describe how Citrobacter rodentium and Escherichia coli leverage colonocyte derived H2O2 to drive two distinct respiratory pathways that support their proliferation in the mouse intestine.


Assuntos
Escherichia coli , Peróxido de Hidrogênio , Animais , Citrobacter rodentium , Colo , Inflamação , Camundongos , Espécies Reativas de Oxigênio , Respiração , Microbiologia do Solo
6.
Infect Immun ; 88(7)2020 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-32284374

RESUMO

Recent studies have determined that inflammasome signaling plays an important role in driving intestinal epithelial cell (IEC) responses to bacterial infections, such as Salmonella enterica serovar Typhimurium. There are two primary inflammasome pathways, canonical (involving caspase-1) and noncanonical (involving caspase-4 and -5 in humans and caspase-11 in mice). Prior studies identified the canonical inflammasome as the major pathway leading to interleukin-18 (IL-18) release and restriction of S Typhimurium replication in the mouse cecum. In contrast, the human C2Bbe1 colorectal carcinoma cell line expresses little caspase-1 but instead utilizes caspase-4 to respond to S Typhimurium infection. Intestinal enteroid culture has enabled long-term propagation of untransformed IECs from multiple species, including mouse and human. Capitalizing on this technology, we used a genetic approach to directly compare the relative importance of different inflammatory caspases in untransformed mouse and human IECs and transformed human IECs upon S Typhimurium infection in vitro We show that caspase-1 is important for restricting intracellular S Typhimurium replication and initiating IL-18 secretion in mouse IECs but is dispensable in human IECs. In contrast, restriction of intracellular S Typhimurium and production of IL-18 are dependent on caspase-4 in both transformed and untransformed human IECs. Notably, cytosolic replication in untransformed cells from both species was less pronounced than in transformed human cells, suggesting that transformation may impact additional pathways that restrict S Typhimurium replication. Taken together, these data highlight the differences between mouse and human IECs and the utility of studying transformed and untransformed cells in parallel.


Assuntos
Inflamassomos/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Infecções por Salmonella/metabolismo , Infecções por Salmonella/microbiologia , Salmonella enterica/fisiologia , Animais , Biomarcadores , Caspases/metabolismo , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Humanos , Mucosa Intestinal/patologia , Camundongos , Infecções por Salmonella/genética
7.
PLoS Pathog ; 16(4): e1008498, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32282854

RESUMO

We investigated the role of the inflammasome effector caspases-1 and -11 during Salmonella enterica serovar Typhimurium infection of murine intestinal epithelial cells (IECs). Salmonella burdens were significantly greater in the intestines of caspase-1/11 deficient (Casp1/11-/-), Casp1-/- and Casp11-/- mice, as compared to wildtype mice. To determine if this reflected IEC-intrinsic inflammasomes, enteroid monolayers were derived and infected with Salmonella. Casp11-/- and wildtype monolayers responded similarly, whereas Casp1-/- and Casp1/11-/- monolayers carried significantly increased intracellular burdens, concomitant with marked decreases in IEC shedding and death. Pretreatment with IFN-γ to mimic inflammation increased caspase-11 levels and IEC death, and reduced Salmonella burdens in Casp1-/- monolayers, while high intracellular burdens and limited cell shedding persisted in Casp1/11-/- monolayers. Thus caspase-1 regulates inflammasome responses in IECs at baseline, while proinflammatory activation of IECs reveals a compensatory role for caspase-11. These results demonstrate the importance of IEC-intrinsic canonical and non-canonical inflammasomes in host defense against Salmonella.


Assuntos
Caspase 1/imunologia , Caspases Iniciadoras/imunologia , Inflamassomos/imunologia , Intestinos/enzimologia , Intestinos/imunologia , Infecções por Salmonella/enzimologia , Salmonella typhimurium/imunologia , Animais , Células Epiteliais/enzimologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Feminino , Imunidade nas Mucosas , Inflamassomos/metabolismo , Interferon gama/imunologia , Mucosa Intestinal/enzimologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Intestinos/microbiologia , Lipopolissacarídeos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Salmonella/imunologia , Salmonella typhimurium/patogenicidade
9.
Trends Immunol ; 39(9): 677-696, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29716793

RESUMO

The gastrointestinal (GI) tract represents a unique challenge to the mammalian immune system. It must tolerate the presence of the luminal microbiota and thus not respond to their products, but still protect the intestinal mucosa from potentially harmful dietary antigens and invading pathogens. The intestinal epithelium, composed of a single layer of cells, is crucial for preserving gut homeostasis and acts both as a physical barrier and as a coordinating hub for immune defense and crosstalk between bacteria and immune cells. We highlight here recent findings regarding communication between microbes and intestinal epithelial cells (IECs), as well as the immune mechanisms employed by distinct IEC subsets to promote homeostasis, emphasizing the central and active role that these cells play in host enteric defense.


Assuntos
Imunidade nas Mucosas , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Animais , Antígenos/imunologia , Comunicação Celular , Disbiose , Células Epiteliais/metabolismo , Microbioma Gastrointestinal/imunologia , Homeostase , Interações Hospedeiro-Patógeno/imunologia , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Imunidade Inata , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Transdução de Sinais
10.
Am J Physiol Gastrointest Liver Physiol ; 314(3): G360-G377, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29122749

RESUMO

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.


Assuntos
Bactérias/patogenicidade , Infecções Bacterianas/microbiologia , Microbioma Gastrointestinal , Células Caliciformes/microbiologia , Doenças Inflamatórias Intestinais/microbiologia , Mucosa Intestinal/microbiologia , Animais , Bactérias/crescimento & desenvolvimento , Infecções Bacterianas/metabolismo , Infecções Bacterianas/fisiopatologia , Células Caliciformes/metabolismo , Homeostase , Interações Hospedeiro-Patógeno , Humanos , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/fisiopatologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiopatologia , Mucina-2/metabolismo , Muco/metabolismo , Transdução de Sinais
11.
Sci Rep ; 7: 45274, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28349941

RESUMO

Breast milk has many beneficial properties and unusual characteristics including a unique fat component, termed milk fat globule membrane (MFGM). While breast milk yields important developmental benefits, there are situations where it is unavailable resulting in a need for formula feeding. Most formulas do not contain MFGM, but derive their lipids from vegetable sources, which differ greatly in size and composition. Here we tested the effects of MFGM supplementation on intestinal development and the microbiome as well as its potential to protect against Clostridium difficile induced colitis. The pup-in-a-cup model was used to deliver either control or MFGM supplemented formula to rats from 5 to 15 days of age; with mother's milk (MM) reared animals used as controls. While CTL formula yielded significant deficits in intestinal development as compared to MM littermates, addition of MFGM to formula restored intestinal growth, Paneth and goblet cell numbers, and tight junction protein patterns to that of MM pups. Moreover, the gut microbiota of MFGM and MM pups displayed greater similarities than CTL, and proved protective against C. difficile toxin induced inflammation. Our study thus demonstrates that addition of MFGM to formula promotes development of the intestinal epithelium and microbiome and protects against inflammation.


Assuntos
Microbioma Gastrointestinal , Intestinos/efeitos dos fármacos , Lipídeos de Membrana/farmacologia , Leite/química , Animais , Suplementos Nutricionais , Células Epiteliais/química , Células Epiteliais/metabolismo , Feminino , Humanos , Intestinos/crescimento & desenvolvimento , Intestinos/microbiologia , Masculino , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/metabolismo , Lipídeos de Membrana/administração & dosagem , Lipídeos de Membrana/análise , Ratos , Ratos Sprague-Dawley
12.
Cell Microbiol ; 19(4)2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28117938

RESUMO

Although much research has focused on defining the actions of caspase-1 containing canonical inflammasomes in promoting host defense, noncanonical inflammasomes have received comparatively little attention. Exciting new concepts have recently emerged detailing their atypical mechanism of activation, importance in defending against cytosolic Gram-negative pathogens, and role in innate immune defenses of nonmyeloid cells, which has revamped interest in the study of noncanonical inflammmasomes. Here, we will discuss these latest findings about caspase-4, -5, and -11 containing inflammasomes in the context of their role in pathogen elimination in mice and humans.


Assuntos
Infecções por Bactérias Gram-Negativas/imunologia , Inflamassomos/fisiologia , Animais , Caspases/metabolismo , Infecções por Bactérias Gram-Negativas/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Transdução de Sinais/imunologia
13.
Curr Top Microbiol Immunol ; 397: 43-67, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27460804

RESUMO

Inflammasomes are macromolecular cytoplasmic complexes that act as signaling platforms for the activation of inflammatory caspases. Their activation triggers the processing and secretion of the pro-inflammatory cytokines IL-1ß and IL-18, as well as the induction of a specialized form of inflammatory cell death termed pyroptosis. Here, we review the mechanisms of inflammasome activation triggered by the intracellular pathogen Salmonella enterica serovar Typhimurium. We highlight the different inflammasome subfamilies utilized by macrophages, neutrophils, dendritic cells, and intestinal epithelial cells response to a Salmonella infection as well as the Salmonella ligands that trigger each inflammasome's formation. We also discuss the evasion strategies utilized by Salmonella to avoid inflammasome detection. Overall, inflammasomes play a key and multilayered role at distinct stages of host cell defense against Salmonella infection.


Assuntos
Inflamassomos/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Animais , Interações Hospedeiro-Patógeno , Humanos , Inflamassomos/genética , Macrófagos/imunologia , Macrófagos/microbiologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/genética , Salmonella typhimurium/fisiologia
14.
PLoS Pathog ; 11(8): e1005107, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26295949

RESUMO

Efficient acquisition of extracellular nutrients is essential for bacterial pathogenesis, however the identities and mechanisms for transport of many of these substrates remain unclear. Here, we investigate the predicted iron-binding transporter AfuABC and its role in bacterial pathogenesis in vivo. By crystallographic, biophysical and in vivo approaches, we show that AfuABC is in fact a cyclic hexose/heptose-phosphate transporter with high selectivity and specificity for a set of ubiquitous metabolites (glucose-6-phosphate, fructose-6-phosphate and sedoheptulose-7-phosphate). AfuABC is conserved across a wide range of bacterial genera, including the enteric pathogens EHEC O157:H7 and its murine-specific relative Citrobacter rodentium, where it lies adjacent to genes implicated in sugar sensing and acquisition. C. rodentium ΔafuA was significantly impaired in an in vivo murine competitive assay as well as its ability to transmit infection from an afflicted to a naïve murine host. Sugar-phosphates were present in normal and infected intestinal mucus and stool samples, indicating that these metabolites are available within the intestinal lumen for enteric bacteria to import during infection. Our study shows that AfuABC-dependent uptake of sugar-phosphates plays a critical role during enteric bacterial infection and uncovers previously unrecognized roles for these metabolites as important contributors to successful pathogenesis.


Assuntos
Metabolismo dos Carboidratos/fisiologia , Infecções por Enterobacteriaceae/metabolismo , Infecções por Enterobacteriaceae/transmissão , Intestinos/microbiologia , Animais , Transporte Biológico Ativo/fisiologia , Calorimetria , Cromatografia Líquida , Citrobacter rodentium , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Mutagênese Sítio-Dirigida , Fosforilação , Filogenia , Espectrometria de Massas em Tandem
15.
Cell Host Microbe ; 16(2): 249-256, 2014 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-25121752

RESUMO

Inflammasome-mediated host defenses have been extensively studied in innate immune cells. Whether inflammasomes function for innate defense in intestinal epithelial cells, which represent the first line of defense against enteric pathogens, remains unknown. We observed enhanced Salmonella enterica serovar Typhimurium colonization in the intestinal epithelium of caspase-11-deficient mice, but not at systemic sites. In polarized epithelial monolayers, siRNA-mediated depletion of caspase-4, a human ortholog of caspase-11, also led to increased bacterial colonization. Decreased rates of pyroptotic cell death, a host defense mechanism that extrudes S. Typhimurium-infected cells from the polarized epithelium, accounted for increased pathogen burdens. The caspase-4 inflammasome also governs activation of the proinflammatory cytokine, interleukin (IL)-18, in response to intracellular (S. Typhimurium) and extracellular (enteropathogenic Escherichia coli) enteric pathogens, via intracellular LPS sensing. Therefore, an epithelial cell-intrinsic noncanonical inflammasome plays a critical role in antimicrobial defense at the intestinal mucosal surface.


Assuntos
Caspases Iniciadoras/metabolismo , Caspases/metabolismo , Infecções por Escherichia coli/enzimologia , Inflamassomos/fisiologia , Infecções por Salmonella/enzimologia , Animais , Linhagem Celular Tumoral , Escherichia coli Enteropatogênica/imunologia , Ativação Enzimática , Infecções por Escherichia coli/imunologia , Gastroenterite/enzimologia , Gastroenterite/microbiologia , Humanos , Interleucina-18/metabolismo , Mucosa Intestinal/enzimologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Lipopolissacarídeos/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Salmonella/imunologia , Salmonella enterica/imunologia
16.
PLoS Pathog ; 10(7): e1004264, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25033044

RESUMO

Campylobacter jejuni is a major source of foodborne illness in the developed world, and a common cause of clinical gastroenteritis. Exactly how C. jejuni colonizes its host's intestines and causes disease is poorly understood. Although it causes severe diarrhea and gastroenteritis in humans, C. jejuni typically dwells as a commensal microbe within the intestines of most animals, including birds, where its colonization is asymptomatic. Pretreatment of C57BL/6 mice with the antibiotic vancomycin facilitated intestinal C. jejuni colonization, albeit with minimal pathology. In contrast, vancomycin pretreatment of mice deficient in SIGIRR (Sigirr(-/-)), a negative regulator of MyD88-dependent signaling led to heavy and widespread C. jejuni colonization, accompanied by severe gastroenteritis involving strongly elevated transcription of Th1/Th17 cytokines. C. jejuni heavily colonized the cecal and colonic crypts of Sigirr(-/-) mice, adhering to, as well as invading intestinal epithelial cells. This infectivity was dependent on established C. jejuni pathogenicity factors, capsular polysaccharides (kpsM) and motility/flagella (flaA). We also explored the basis for the inflammatory response elicited by C. jejuni in Sigirr(-/-) mice, focusing on the roles played by Toll-like receptors (TLR) 2 and 4, as these innate receptors were strongly stimulated by C. jejuni. Despite heavy colonization, Tlr4(-/-)/Sigirr(-/-) mice were largely unresponsive to infection by C. jejuni, whereas Tlr2(-/-)/Sigirr(-/-) mice developed exaggerated inflammation and pathology. This indicates that TLR4 signaling underlies the majority of the enteritis seen in this model, whereas TLR2 signaling had a protective role, acting to promote mucosal integrity. Furthermore, we found that loss of the C. jejuni capsule led to increased TLR4 activation and exaggerated inflammation and gastroenteritis. Together, these results validate the use of Sigirr(-/-) mice as an exciting and relevant animal model for studying the pathogenesis and innate immune responses to C. jejuni.


Assuntos
Infecções por Campylobacter/imunologia , Campylobacter jejuni/imunologia , Gastroenterite/imunologia , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Cápsulas Bacterianas/imunologia , Infecções por Campylobacter/genética , Infecções por Campylobacter/patologia , Modelos Animais de Doenças , Gastroenterite/genética , Gastroenterite/microbiologia , Gastroenterite/patologia , Camundongos , Camundongos Knockout , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/imunologia , Transdução de Sinais/genética , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética
17.
Infect Immun ; 82(9): 3753-63, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24958710

RESUMO

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.


Assuntos
Anti-Infecciosos/imunologia , Colite/imunologia , Células Caliciformes/imunologia , Mucosa Intestinal/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais/imunologia , Animais , Citrobacter rodentium/imunologia , Colite/microbiologia , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/microbiologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Gastroenterite/imunologia , Gastroenterite/microbiologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Células Caliciformes/microbiologia , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Salmonella/imunologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/imunologia
18.
Artigo em Inglês | MEDLINE | ID: mdl-22919603

RESUMO

Bacteriophages are estimated to be the most abundant entities on earth and can be found in every niche where their bacterial hosts reside. The initial interaction between phages and Campylobacter jejuni, a common colonizer of poultry intestines and a major source of foodborne bacterial gastroenteritis in humans, is not well understood. Recently, we isolated and characterized a phage F336 resistant variant of C. jejuni NCTC11168 called 11168R. Comparisons of 11168R with the wildtype lead to the identification of a novel phage receptor, the phase variable O-methyl phosphoramidate (MeOPN) moiety of the C. jejuni capsular polysaccharide (CPS). In this study we demonstrate that the 11168R strain has gained cross-resistance to four other phages in our collection (F198, F287, F303, and F326). The reduced plaquing efficiencies suggested that MeOPN is recognized as a receptor by several phages infecting C. jejuni. To further explore the role of CPS modifications in C. jejuni phage recognition and infectivity, we tested the ability of F198, F287, F303, F326, and F336 to infect different CPS variants of NCTC11168, including defined CPS mutants. These strains were characterized by high-resolution magic angle spinning NMR spectroscopy. We found that in addition to MeOPN, the phase variable 3-O-Me and 6-O-Me groups of the NCTC11168 CPS structure may influence the plaquing efficiencies of the phages. Furthermore, co-infection of chickens with both C. jejuni NCTC11168 and phage F336 resulted in selection of resistant C. jejuni bacteria, which either lack MeOPN or gain 6-O-Me groups on their surface, demonstrating that resistance can be acquired in vivo. In summary, we have shown that phase variable CPS structures modulate phage infectivity in C. jejuni and suggest that the constant phage predation in the avian gut selects for changes in these structures leading to a continuing phage-host co-evolution.


Assuntos
Bacteriófagos/fisiologia , Infecções por Campylobacter/microbiologia , Campylobacter jejuni/química , Campylobacter jejuni/virologia , Galinhas/microbiologia , Polissacarídeos Bacterianos/metabolismo , Animais , Mutação , Polissacarídeos Bacterianos/genética , Receptores Virais/genética , Receptores Virais/metabolismo , Ensaio de Placa Viral , Internalização do Vírus
19.
J Cereb Blood Flow Metab ; 28(9): 1613-23, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18506197

RESUMO

We hypothesized that magnetic resonance magnetization transfer (MT) imaging would be sensitive for detecting cerebral ischemic injury in white matter of neonatal brain. We compared the progression of changes in T(2) and the MT ratio (MTR) after cerebral hypoxic-ischemic insults of differing severity in neonatal rats. Magnetization transfer imaging parameters were first optimized, and then MTR and T(2) maps were acquired at various times after a mild (rather selective white matter) or substantial insult produced by unilateral cerebral hypoxia-ischemia. Depending on insult severity, time after insult, and region (e.g., subcortical white matter or cortex), cerebral hypoxia-ischemia produced reductions in MTR and an increase in T(2). The exception was acutely at 1 to 5 h at which time points MTR was reduced ipsilaterally in white matter, whereas T(2) was not affected significantly. Progression of imaging changes differed in rats grouped according to whether gross damage was present after chronic recovery. Behavioral changes were generally associated with chronic reductions in MTR and gross brain damage. Magnetization transfer imaging was capable of early detection of hypoxic-ischemic injury and particularly sensitive for identifying the progression of cerebral injury in white matter. Magnetization transfer ratio has potential for assisting with early diagnosis and treatment assessment for infants affected by perinatal hypoxia-ischemia.


Assuntos
Hipóxia-Isquemia Encefálica/diagnóstico , Imageamento por Ressonância Magnética/métodos , Animais , Animais Recém-Nascidos , Diagnóstico por Imagem , Fibras Nervosas Mielinizadas , Ratos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...