Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pflugers Arch ; 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38570355

RESUMEN

Mammalian cells utilize glucose as a primary carbon source to produce energy for most cellular functions. However, the bioenergetic homeostasis of cells can be perturbed by environmental alterations, such as changes in oxygen levels which can be associated with bacterial infection. Reduction in oxygen availability leads to a state of hypoxia, inducing numerous cellular responses that aim to combat this stress. Importantly, hypoxia strongly augments cellular glycolysis in most cell types to compensate for the loss of aerobic respiration. Understanding how this host cell metabolic adaptation to hypoxia impacts the course of bacterial infection will identify new anti-microbial targets. This review will highlight developments in our understanding of glycolytic substrate channeling and spatiotemporal enzymatic organization in response to hypoxia, shedding light on the integral role of the hypoxia-inducible factor (HIF) during host-pathogen interactions. Furthermore, the ability of intracellular and extracellular bacteria (pathogens and commensals alike) to modulate host cellular glucose metabolism will be discussed.

2.
Am J Physiol Gastrointest Liver Physiol ; 324(6): G452-G465, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37070751

RESUMEN

Mucin O-linked glycans are important mediators of host-microbiota-pathogen interactions in the gastrointestinal tract. The major component of intestinal mucus, the MUC2 mucin, is densely glycosylated, with up to 80% of its weight-to-volume ratio represented by O-linked glycans. Glycosylation of secretory gel-forming mucins has an enormous impact on intestinal barrier function, microbial metabolism, and mucus colonization by both pathogenic and commensal microbes. Mucin O-glycans and glycan-derived sugars may be degraded and used as a nutrient source and may regulate microbial gene expression and virulence. Short-chain fatty acids, produced as a by-product of glycan fermentation, can regulate host immunity and goblet cell activity and are important for host-microbe homeostasis. Mucin glycans may also act as microbial binding sites, influencing intestinal colonization and translocation through the mucus gel barrier. Recent findings indicate that alterations to mucin glycosylation impact the susceptibility of mucins to degradation, resulting in altered barrier function and intestinal permeability. Alterations to mucin glycosylation patterns are frequently observed during intestinal infection and inflammation and have been implicated in microbiota dysbiosis and expansion of pathobionts. Recent work has demonstrated that these alterations can play key roles in disease pathogenesis. The precise mechanisms remain obscure. This review highlights the important roles of O-linked glycans in host-microbe interactions and disease pathogenesis in the context of intestinal infections.


Asunto(s)
Microbiota , Mucinas , Humanos , Mucinas/metabolismo , Mucosa Intestinal/metabolismo , Disbiosis , Interacciones Huésped-Patógeno , Homeostasis , Polisacáridos/química , Mucina 2/metabolismo
3.
Eur J Clin Microbiol Infect Dis ; 42(12): 1405-1423, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37897520

RESUMEN

In recent years, multidrug-resistant Acinetobacter baumannii has emerged globally as a major threat to the healthcare system. It is now listed by the World Health Organization as a priority one for the need of new therapeutic agents. A. baumannii has the capacity to develop robust biofilms on biotic and abiotic surfaces. Biofilm development allows these bacteria to resist various environmental stressors, including antibiotics and lack of nutrients or water, which in turn allows the persistence of A. baumannii in the hospital environment and further outbreaks. Investigation into therapeutic alternatives that will act on both biofilm formation and antimicrobial resistance (AMR) is sorely needed. The aim of the present review is to critically discuss the various mechanisms by which AMR and biofilm formation may be co-regulated in A. baumannii in an attempt to shed light on paths towards novel therapeutic opportunities. After discussing the clinical importance of A. baumannii, this critical review highlights biofilm-formation genes that may be associated with the co-regulation of AMR. Particularly worthy of consideration are genes regulating the quorum sensing system AbaI/AbaR, AbOmpA (OmpA protein), Bap (biofilm-associated protein), the two-component regulatory system BfmRS, the PER-1 ß-lactamase, EpsA, and PTK. Finally, this review discusses ongoing experimental therapeutic strategies to fight A. baumannii infections, namely vaccine development, quorum sensing interference, nanoparticles, metal ions, natural products, antimicrobial peptides, and phage therapy. A better understanding of the mechanisms that co-regulate biofilm formation and AMR will help identify new therapeutic targets, as combined approaches may confer synergistic benefits for effective and safer treatments.


Asunto(s)
Acinetobacter baumannii , Antibacterianos , Humanos , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana , Biopelículas , Percepción de Quorum , Farmacorresistencia Bacteriana Múltiple/genética
4.
BMC Gastroenterol ; 20(1): 64, 2020 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-32164535

RESUMEN

BACKGROUND: Endoscopic retrograde cholangio-pancreatography (ERCP) is commonly performed in the management of pancreatic and biliary disease. Duodenoscopes are specialized endoscopes used to perform ERCP, and inherent to their design, a high rate of persistent bacterial contamination exists even after automated reprocessing and disinfection. Consequently, in recent years, ERCP has been associated with infection transmission, leading to several fatal patient outbreaks. Due to increasing fears over widespread future duodenoscope-related outbreaks, regulatory bodies have called for alterations in the design of duodenoscopes. A duodenoscope has recently been developed that employs a disposable cap. This novel design theoretically eliminates the mechanism behind persistent bacterial contamination and infection transmission. However, there are no data demonstrating persistent bacterial contamination rates, technical success rates, or clinical outcomes associated with these duodenoscopes. METHODS: A parallel arm randomized controlled trial will be performed for which 520 patients will be recruited. The study population will consist of consecutive patients undergoing ERCP procedures for any indication at a high-volume tertiary care centre in Calgary, Alberta, Canada. Patients will be randomized to an intervention group, that will undergo ERCP with a novel duodenoscope with disposable cap, or to a control group who will undergo ERCP with a traditional duodenoscope. Co-primary outcomes will include persistent bacterial contamination rates (post automated reprocessing) and ERCP technical success rates. Secondary outcomes include clinical success rates, overall and specific early and late adverse event rates, 30-day mortality and healthcare utilization rates, procedure and reprocessing times, and ease of device use. DISCUSSION: The ICECAP trial will answer important questions regarding the use of a novel duodenoscope with disposable cap. Specifically, persistent bacterial contamination, technical performance, and relevant clinical outcomes will be assessed. Given the mortality and morbidity burden associated with ERCP-related infectious outbreaks, the results of this study have the capacity to be impactful at an international level. TRIAL REGISTRATION: This trial was registered on clinicaltrials.gov (NCT04040504) on July 31, 2019.


Asunto(s)
Colangiopancreatografia Retrógrada Endoscópica/instrumentación , Infección Hospitalaria/prevención & control , Duodenoscopios/microbiología , Contaminación de Equipos/prevención & control , Control de Infecciones/métodos , Equipos Desechables , Diseño de Equipo , Humanos
5.
Am J Physiol Gastrointest Liver Physiol ; 314(2): G143-G149, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29025733

RESUMEN

A diverse range of effects of the intestinal microbiota on mucosal defense and injury has become increasingly clear over the past decade. Hydrogen sulfide (H2S) has emerged as an important mediator of many physiological functions, including gastrointestinal mucosal defense and repair. Hydrogen sulfide is produced by gastrointestinal tract tissues and by bacteria residing within the gut and can influence the function of a wide range of cells. The microbiota also appears to be an important target of hydrogen sulfide. H2S donors can modify the gut microbiota, and the gastrointestinal epithelium is a major site of oxidation of microbial-derived H2S. When administered together with nonsteroidal anti-inflammatory drugs, H2S can prevent some of the dysbiosis those drugs induce, possibly contributing to the observed prevention of gastrointestinal damage. Exogenous H2S can also markedly reduce the severity of experimental colitis and plays important roles in modulating epithelial cell-mucus-bacterial interactions in the intestine, contributing to its ability to promote resolution of inflammation and repair of tissue injury. In this paper we review recent studies examining the roles of H2S in mucosal defense, the possibility that H2S can damage the gastrointestinal epithelium, and effects of H2S on the gut microbiota and on mucus and biofilm interactions in the context of intestinal inflammation.


Asunto(s)
Bacterias/metabolismo , Microbioma Gastrointestinal , Sulfuro de Hidrógeno/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Animales , Antibacterianos/toxicidad , Antiinflamatorios no Esteroideos/toxicidad , Bacterias/efectos de los fármacos , Bacterias/crecimiento & desarrollo , Biopelículas/crecimiento & desarrollo , Disbiosis , Microbioma Gastrointestinal/efectos de los fármacos , Interacciones Huésped-Patógeno , Humanos , Sulfuro de Hidrógeno/farmacología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Probióticos/farmacología
6.
Am J Pathol ; 187(11): 2486-2498, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28823873

RESUMEN

The intestinal mucous layer provides a critical host defense against pathogen exposure and epithelial injury, yet little is known about how enteropathogens may circumvent this physiologic barrier. Giardia duodenalis is a small intestinal parasite responsible for diarrheal disease and chronic postinfectious illness. This study reveals a complex interaction at the surface of epithelial cells, between G. duodenalis and the intestinal mucous layer. Here, we reveal mechanisms whereby G. duodenalis evades and disrupts the first line of host defense by degrading human mucin-2 (MUC2), depleting mucin stores and inducing differential gene expression in the mouse small and large intestines. Human colonic biopsy specimens exposed to G. duodenalis were depleted of mucus, and in vivo mice infected with G. duodenalis had a thinner mucous layer and demonstrated differential Muc2 and Muc5ac mucin gene expression. Infection in Muc2-/- mice elevated trophozoite colonization in the small intestine and impaired weight gain. In vitro, human LS174T goblet-like cells were depleted of mucus and had elevated levels of MUC2 mRNA expression after G. duodenalis exposure. Importantly, the cysteine protease inhibitor E64 prevented mucous degradation, mucin depletion, and the increase in MUC2 expression. This article describes a novel role for Giardia's cysteine proteases in pathogenesis and how Giardia's disruptions of the mucous barrier facilitate bacterial translocation that may contribute to the onset and propagation of disease.


Asunto(s)
Células Epiteliales/metabolismo , Giardiasis/genética , Mucinas/genética , Moco/metabolismo , Animales , Traslocación Bacteriana/genética , Proteasas de Cisteína/metabolismo , Femenino , Giardia lamblia/genética , Giardiasis/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Masculino , Ratones , Mucinas/metabolismo
7.
PLoS Pathog ; 11(5): e1004853, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25946018

RESUMEN

Most bacterial infections induce the activation of polymorphonuclear neutrophils (PMNs), enhance their microbicidal function, and promote the survival of these leukocytes for protracted periods of time. Brucella abortus is a stealthy pathogen that evades innate immunity, barely activates PMNs, and resists the killing mechanisms of these phagocytes. Intriguing clinical signs observed during brucellosis are the low numbers of Brucella infected PMNs in the target organs and neutropenia in a proportion of the patients; features that deserve further attention. Here we demonstrate that B. abortus prematurely kills human PMNs in a dose-dependent and cell-specific manner. Death of PMNs is concomitant with the intracellular Brucella lipopolysaccharide (Br-LPS) release within vacuoles. This molecule and its lipid A reproduce the premature cell death of PMNs, a phenomenon associated to the low production of proinflammatory cytokines. Blocking of CD14 but not TLR4 prevents the Br-LPS-induced cell death. The PMNs cell death departs from necrosis, NETosis and classical apoptosis. The mechanism of PMN cell death is linked to the activation of NADPH-oxidase and a modest but steadily increase of ROS mediators. These effectors generate DNA damage, recruitments of check point kinase 1, caspases 5 and to minor extent of caspase 4, RIP1 and Ca++ release. The production of IL-1ß by PMNs was barely stimulated by B. abortus infection or Br-LPS treatment. Likewise, inhibition of caspase 1 did not hamper the Br-LPS induced PMN cell death, suggesting that the inflammasome pathway was not involved. Although activation of caspases 8 and 9 was observed, they did not seem to participate in the initial triggering mechanisms, since inhibition of these caspases scarcely blocked PMN cell death. These findings suggest a mechanism for neutropenia in chronic brucellosis and reveal a novel Brucella-host cross-talk through which B. abortus is able to hinder the innate function of PMN.


Asunto(s)
Brucella abortus/inmunología , Brucelosis/inmunología , Lipopolisacáridos/inmunología , Mortalidad Prematura , Neutrófilos/citología , Brucella abortus/aislamiento & purificación , Muerte Celular , Citocinas/metabolismo , Humanos , Inmunidad Innata/inmunología , Leucocitos/metabolismo
8.
BMC Microbiol ; 17(1): 70, 2017 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-28330466

RESUMEN

BACKGROUND: The nasopharyngeal (NP) microbiota plays an important role in bovine health, comprising a rich and diverse microbial community. The nasopharynx is also the niche for potentially pathogenic agents which are associated with bovine respiratory disease (BRD), a serious and costly illness in feedlot cattle. We used 14 beef heifers from a closed and disease-free herd to assess the dynamics of the NP microbiota of cattle that are transported to a feedlot. Cattle were sampled prior to transport to the feedlot (day 0) and at days 2, 7, and 14. RESULTS: The structure of the NP microbiota changed significantly over the course of the study, with the largest shift occurring between day 0 (prior to transport) and day 2 (P < 0.001). Phylogenetic diversity and richness increased following feedlot placement (day 2; P < 0.05). The genera Pasteurella, Bacillus, and Proteus were enriched at day 0, Streptococcus and Acinetobacter at day 2, Bifidobacterium at day 7, and Mycoplasma at day 14. The functional potential of the NP microbiota was assessed using PICRUSt, revealing that replication and repair, as well as translation pathways, were more relatively abundant in day 14 samples. These differences were driven mostly by Mycoplasma. Although eight cattle were culture-positive for the BRD-associated bacterium Pasteurella multocida at one or more sampling times, none were culture-positive for Mannheimia haemolytica or Histophilus somni. CONCLUSIONS: This study investigated the effect that feedlot placement has on the NP microbiota of beef cattle over a 14-d period. Within two days of transport to the feedlot, the NP microbiota changed significantly, increasing in both phylogenetic diversity and richness. These results demonstrate that there is an abrupt shift in the NP microbiota of cattle after transportation to a feedlot. This may have importance for understanding why cattle are most susceptible to BRD after feedlot placement.


Asunto(s)
Bacterias/clasificación , Bacterias/aislamiento & purificación , Bovinos/microbiología , Microbiota , Nasofaringe/microbiología , Animales , Bacterias/genética , Biodiversidad , Complejo Respiratorio Bovino/microbiología , ADN Bacteriano , Genes Bacterianos , Vivienda para Animales , Microbiota/genética , Filogenia , ARN Ribosómico 16S/genética , Factores de Tiempo
9.
Anaerobe ; 47: 157-164, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28526497

RESUMEN

Biofilms composed of anaerobic bacteria can result in persistent infections and chronic inflammation. Host immune cells have difficulties clearing biofilm-related infections and this can result in tissue damage. Neutrophils are a vital component of the innate immune system and help clear biofilms. The comparative neutrophilic response to biofilms versus planktonic bacteria remains incompletely understood, particularly in the context of mixed infections. The objective of this study was to generate mixed species anaerobic bacterial biofilms composed of two opportunistic pathogens, Fusobacterium necrophorum and Porphyromonas levii, and evaluate neutrophil responses to extracellular fractions from both biofilms and planktonic cell co-cultures of the same bacteria. Purified bovine neutrophils exposed to culture supernatants from mixed species planktonic bacteria showed elevated oxidative activity compared to neutrophils exposed to biofilms composed of the same bacteria. Bacterial lipopolysaccharide plays a significant role in the stimulation of neutrophils; biofilms produced substantially more lipopolysaccharide than planktonic bacteria under these experimental conditions. Removal of lipopolysaccharide significantly reduced neutrophil oxidative response to culture supernatants of planktonic bacteria. Oxidative responses to LPS-removed biofilm supernatants and LPS-removed planktonic cell supernatants were similar. The limited neutrophil response to biofilm bacteria observed in this study supports the reduced ability of the innate immune system to eradicate biofilm-associated infections. Lipopolysaccharide is likely important in neutrophil response; however, the presence of other extracellular, immune modifying molecules in the bacterial media also appears to be important in altering neutrophil function.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Fusobacterium necrophorum/inmunología , Fusobacterium necrophorum/fisiología , Neutrófilos/inmunología , Polisacáridos Bacterianos/metabolismo , Porphyromonas/inmunología , Porphyromonas/fisiología , Animales , Bovinos , Fusobacterium necrophorum/efectos de los fármacos , Interacciones Huésped-Patógeno , Neutrófilos/efectos de los fármacos , Oxidantes/metabolismo , Porphyromonas/efectos de los fármacos
10.
J Nematol ; 49(4): 348-356, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29353922

RESUMEN

The microbiome influences host processes including nutritional availability, development, immunity, and behavioral responses. Caenorhabditis elegans is a powerful model to study molecular mechanisms of host-microbial interactions. Recent efforts have been made to profile the natural microbiome of C. elegans, laying a foundation for mechanistic studies of host-microbiome interactions in this genetically tractable model system. Studies using single-species microbes, multi-microbial systems, and humanized worm-microbiome interaction studies reveal metabolic and microbial-microbial interactions relevant in higher organisms. This article discusses recent developments in modeling the effects of host-microbiome interactions in C. elegans.

11.
Am J Physiol Gastrointest Liver Physiol ; 310(8): G574-85, 2016 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-26744469

RESUMEN

Irritable bowel syndrome (IBS) is the most frequent functional gastrointestinal disorder. It is characterized by abdominal hypersensitivity, leading to discomfort and pain, as well as altered bowel habits. While it is common for IBS to develop following the resolution of infectious gastroenteritis [then termed postinfectious IBS (PI-IBS)], the mechanisms remain incompletely understood. Giardia duodenalis is a cosmopolitan water-borne enteropathogen that causes intestinal malabsorption, diarrhea, and postinfectious complications. Cause-and-effect studies using a human enteropathogen to help investigate the mechanisms of PI-IBS are sorely lacking. In an attempt to establish causality between giardiasis and postinfectious visceral hypersensitivity, this study describes a new model of PI-IBS in neonatal rats infected with G. duodenalis At 50 days postinfection with G. duodenalis (assemblage A or B), long after the parasite was cleared, rats developed visceral hypersensitivity to luminal balloon distension in the jejunum and rectum, activation of the nociceptive signaling pathway (increased c-fos expression), histological modifications (villus atrophy and crypt hyperplasia), and proliferation of mucosal intraepithelial lymphocytes and mast cells in the jejunum, but not in the rectum. G. duodenalis infection also disrupted the intestinal barrier, in vivo and in vitro, which in turn promoted the translocation of commensal bacteria. Giardia-induced bacterial paracellular translocation in vitro correlated with degradation of the tight junction proteins occludin and claudin-4. The extensive observations associated with gut hypersensitivity described here demonstrate that, indeed, in this new model of postgiardiasis IBS, alterations to the gut mucosa and c-fos are consistent with those associated with PI-IBS and, hence, offer avenues for new mechanistic research in the field.


Asunto(s)
Microbioma Gastrointestinal , Giardiasis/complicaciones , Síndrome del Colon Irritable/etiología , Migración Transcelular de la Célula , Animales , Células CACO-2 , Escherichia coli/patogenicidad , Escherichia coli/fisiología , Femenino , Giardiasis/microbiología , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Síndrome del Colon Irritable/microbiología , Síndrome del Colon Irritable/parasitología , Masculino , Nocicepción , Ratas , Ratas Sprague-Dawley , Proteínas de Uniones Estrechas/metabolismo
12.
FASEB J ; 29(4): 1591-602, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25550470

RESUMEN

During a course of colitis, production of the gaseous mediator hydrogen sulfide (H2S) is markedly up-regulated at sites of mucosal damage and contributes significantly to healing and resolution of inflammation. The signaling mechanisms through which H2S promotes resolution of colitis are unknown. We hypothesized that the beneficial effects of H2S in experimental colitis are mediated via stabilization of hypoxia-inducible factor (HIF)-1α. The hapten dinitrobenzene sulfonic acid was used to induce colitis in rats and mice. This resulted in an elevated expression of the H2S-producing enzyme, cystathionine γ-lyase (CSE), and HIF-1α at sites of mucosal ulceration, and the expression of these 2 enzymes followed a similar pattern throughout the course of colitis. This represented a functionally important relationship because the loss of CSE-derived H2S production led to decreased HIF-1α stabilization and exacerbation of colitis. Furthermore, application of an H2S-releasing molecule, diallyl disulfide (DADS), stabilized colonic HIF-1α expression, up-regulated hypoxia-responsive genes, and reduced the severity of disease during peak inflammation. Importantly, the ability of DADS to promote the resolution of colitis was abolished when coadministered with an inhibitor of HIF-1α in vivo (PX-478). DADS was also able to maintain HIF-1α expression at a later point in colitis, when HIF-1α levels would have normally returned to control levels, and to enhance resolution. Finally, we found that HIF-1α stabilization inhibited colonic H2S production and may represent a negative feedback mechanism to prevent prolonged HIF-1α stabilization. Our findings demonstrate an important link between H2S and HIF-1α in the resolution of inflammation and injury during colitis and provide mechanistic insights into the therapeutic value of H2S donors.


Asunto(s)
Colitis/metabolismo , Sulfuro de Hidrógeno/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Compuestos Alílicos/farmacología , Animales , Bencenosulfonatos/toxicidad , Colitis/tratamiento farmacológico , Colitis/patología , Cistationina gamma-Liasa/antagonistas & inhibidores , Cistationina gamma-Liasa/metabolismo , Modelos Animales de Enfermedad , Disulfuros/farmacología , Expresión Génica , Células HT29 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Compuestos de Mostaza/farmacología , Fenilpropionatos/farmacología , Estabilidad Proteica , Ratas , Ratas Wistar , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
13.
J Gastroenterol Hepatol ; 31(9): 1594-603, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26773283

RESUMEN

BACKGROUND AND AIM: Irritable bowel syndrome is characterized by abdominal pain and altered bowel habits and may occur following stressful events or infectious gastroenteritis such as giardiasis. Recent findings revealed a link between cholecystokinin (CCK), neurotrophin synthesis, and intestinal hyperalgesia. The aim was to investigate the role of CCK in visceral hypersensitivity using mouse models challenged with a bout of infection with Giardia lamblia or psychological stress, either alone or in combination. METHODS: Abdominal pain was evaluated by visceromoter response to colorectal distension. Nerve fibers in intestinal tissues were stained using immunohistochemistry (PGP9.5). Human neuroblastoma SH-SY5Y cells incubated with bacterial-free mouse gut supernatant or recombinant CCK-8S were assessed for neurite outgrowth and nerve growth factor (NGF) production. RESULTS: Intestinal hypersensitivity was induced by either stress or Giardia infection, and a trend of increased pain was seen following dual stimuli. Increased CCK levels and PGP9.5 immunoreactivity were found in colonic mucosa of mice following stress and/or infection. Inhibitors to the CCK-A receptor (L-364718) or CCK-B receptor (L-365260) blocked visceral hypersensitivity caused by stress, but not when induced by giardiasis. Nerve fiber elongation and NGF synthesis were observed in SH-SY5Y cells after incubation with colonic supernatants from mice given the dual stimuli, or after treatment with CCK-8S. Increased nerve fiber length by colonic supernatant and CCK-8S was attenuated by L-365260 or neutralizing anti-NGF. CONCLUSIONS: This new model successfully recapitulates intestinal hypernociception induced by stress or Giardia. Colonic CCK contributes to visceral hypersensitivity caused by stress, but not by Giardia, partly via NGF-dependent neurite outgrowth.


Asunto(s)
Colecistoquinina/fisiología , Colon/metabolismo , Hiperalgesia/metabolismo , Proyección Neuronal/fisiología , Dolor Abdominal/etiología , Dolor Abdominal/metabolismo , Dolor Abdominal/patología , Animales , Células Cultivadas , Colecistoquinina/farmacología , Técnicas de Cocultivo , Colon/inervación , Medios de Cultivo Condicionados , Dilatación , Giardia lamblia , Giardiasis/complicaciones , Humanos , Hiperalgesia/etiología , Hiperalgesia/patología , Mucosa Intestinal/inervación , Mucosa Intestinal/metabolismo , Masculino , Ratones Endogámicos C57BL , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/patología , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Factor de Crecimiento Nervioso/metabolismo , Proyección Neuronal/efectos de los fármacos , Proteínas Recombinantes/farmacología , Estrés Psicológico/complicaciones
14.
Infect Immun ; 83(12): 4571-81, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26371123

RESUMEN

Campylobacter jejuni is the most common cause of bacterium-induced gastroenteritis, and while typically self-limiting, C. jejuni infections are associated with postinfectious intestinal disorders, including flares in patients with inflammatory bowel disease and postinfectious irritable bowel syndrome (PI-IBS), via mechanisms that remain obscure. Based on the hypothesis that acute campylobacteriosis may cause pathogenic microbiota dysbiosis, we investigated whether C. jejuni may activate dormant virulence genes in noninvasive Escherichia coli and examined the epithelial pathophysiological consequences of these alterations. Microarray and quantitative real-time PCR analyses revealed that E. coli adhesin, flagellum, and hemolysin gene expression were increased when E. coli was exposed to C. jejuni-conditioned medium. Increased development of bacterial flagella upon exposure to live C. jejuni or C. jejuni-conditioned medium was observed under transmission electron microscopy. Atomic force microscopy demonstrated that the forces of bacterial adhesion to colonic T84 enterocytes, and the work required to rupture this adhesion, were significantly increased in E. coli exposed to C. jejuni-conditioned media. Finally, C. jejuni-modified E. coli disrupted TLR4 gene expression and induced proinflammatory CXCL-8 gene expression in colonic enterocytes. Together, these data suggest that exposure to live C. jejuni, and/or to its secretory-excretory products, may activate latent virulence genes in noninvasive E. coli and that these alterations may directly trigger proinflammatory signaling in intestinal epithelia. These observations shed new light on mechanisms that may contribute, at least in part, to postcampylobacteriosis inflammatory disorders.


Asunto(s)
Campylobacter jejuni/metabolismo , Medios de Cultivo Condicionados/farmacología , Enterocitos/efectos de los fármacos , Interleucina-8/inmunología , Receptor Toll-Like 4/inmunología , Campylobacter jejuni/patogenicidad , Línea Celular , Enterocitos/inmunología , Enterocitos/microbiología , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Escherichia coli/patogenicidad , Flagelos/efectos de los fármacos , Flagelos/genética , Flagelos/metabolismo , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Interleucina-8/agonistas , Interleucina-8/genética , Transducción de Señal , Simbiosis , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética , Virulencia
15.
Am J Physiol Gastrointest Liver Physiol ; 308(6): G550-61, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25573177

RESUMEN

Giardia duodenalis is the most common cause of parasitic diarrhea worldwide and a well-established risk factor for postinfectious irritable bowel syndrome. We hypothesized that Giardia-induced disruptions in host-microbiota interactions may play a role in the pathogenesis of giardiasis and in postgiardiasis disease. Functional changes induced by Giardia in commensal bacteria and the resulting effects on Caenorhabditis elegans were determined. Although Giardia or bacteria alone did not affect worm viability, combining commensal Escherichia coli bacteria with Giardia became lethal to C. elegans. Giardia also induced killing of C. elegans with attenuated Citrobacter rodentium espF and map mutant strains, human microbiota from a healthy donor, and microbiota from inflamed colonic sites of ulcerative colitis patient. In contrast, combinations of Giardia with microbiota from noninflamed sites of the same patient allowed for worm survival. The synergistic lethal effects of Giardia and E. coli required the presence of live bacteria and were associated with the facilitation of bacterial colonization in the C. elegans intestine. Exposure to C. elegans and/or Giardia altered the expression of 172 genes in E. coli. The genes affected by Giardia included hydrogen sulfide biosynthesis (HSB) genes, and deletion of a positive regulator of HSB genes, cysB, was sufficient to kill C. elegans even in the absence of Giardia. Our findings indicate that Giardia induces functional changes in commensal bacteria, possibly making them opportunistic pathogens, and alters host-microbe homeostatic interactions. This report describes the use of a novel in vivo model to assess the toxicity of human microbiota.


Asunto(s)
Caenorhabditis elegans/microbiología , Citrobacter rodentium/patogenicidad , Escherichia coli/patogenicidad , Giardia lamblia/patogenicidad , Intestinos/microbiología , Microbiota , Animales , Estudios de Casos y Controles , Citrobacter rodentium/genética , Citrobacter rodentium/metabolismo , Colitis Ulcerosa/microbiología , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Giardia lamblia/metabolismo , Interacciones Huésped-Patógeno , Humanos , Viabilidad Microbiana , Simbiosis , Factores de Tiempo , Virulencia
16.
Am J Physiol Gastrointest Liver Physiol ; 308(12): G994-1003, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25882612

RESUMEN

The small intestine is a significant site of ulceration and bleeding induced by nonsteroidal anti-inflammatory drugs (NSAIDs). The pathogenesis is poorly understood. The present study explored the roles of bile, bacteria, and enterohepatic circulation to NSAID enteropathy, using both a conventional NSAID (naproxen) and a gastrointestinal-safe naproxen derivative (ATB-346), as well as proton pump inhibitors (PPIs). Rats were treated orally with naproxen or equimolar doses of ATB-346 over a 5-day period, with or without PPI administration, and intestinal damage was quantified. The cytotoxicity of bile from the rats was evaluated in vitro. Biliary excretion of naproxen and ATB-346 was determined. The impact of the NSAIDs and of PPIs on the composition of the intestinal microbiota was examined by deep sequencing of 16s rRNA. Naproxen caused significant intestinal damage and inflammation, whereas ATB-346 did not. Naproxen, but not ATB-346, dose dependently increased the cytotoxicity of bile, and it was further increased by PPI coadministration. Whereas biliary excretion of naproxen was significant in naproxen-treated rats, it was greatly reduced in rats treated with ATB-346. The enteric microbiota of naproxen-treated rats was distinct from that in vehicle- or ATB-346-treated rats, and PPI administration caused significant intestinal dysbiosis. The increase in cytotoxicity of bile induced by naproxen and PPIs may contribute significantly to intestinal ulceration and bleeding. Some of these effects may occur secondary to significant changes in the jejunal microbiota induced by both naproxen and PPIs.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Sulfuro de Hidrógeno/farmacología , Enfermedades Intestinales/tratamiento farmacológico , Naproxeno/análogos & derivados , Inhibidores de la Bomba de Protones/farmacología , Animales , Eliminación Hepatobiliar/fisiología , Inflamación/tratamiento farmacológico , Masculino , Naproxeno/farmacología , ARN Ribosómico 16S/genética , Ratas Wistar
17.
Infect Immun ; 82(7): 2772-87, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24733096

RESUMEN

Giardia duodenalis (syn. G. intestinalis, G. lamblia) infections are a leading cause of waterborne diarrheal disease that can also result in the development of postinfectious functional gastrointestinal disorders via mechanisms that remain unclear. Parasite numbers exceed 10(6) trophozoites per centimeter of gut at the height of an infection. Yet the intestinal mucosa of G. duodenalis-infected individuals is devoid of signs of overt inflammation. G. duodenalis infections can also occur concurrently with infections with other proinflammatory gastrointestinal pathogens. Little is known of whether and how this parasite can attenuate host inflammatory responses induced by other proinflammatory stimuli, such as a gastrointestinal pathogen. Identifying hitherto-unrecognized parasitic immunomodulatory pathways, the present studies demonstrated that G. duodenalis trophozoites attenuate secretion of the potent neutrophil chemoattractant interleukin-8 (CXCL8); these effects were observed in human small intestinal mucosal tissues and from intestinal epithelial monolayers, activated through administration of proinflammatory interleukin-1ß or Salmonella enterica serovar Typhimurium. This attenuation is caused by the secretion of G. duodenalis cathepsin B cysteine proteases that degrade CXCL8 posttranscriptionally. Furthermore, the degradation of CXCL8 via G. duodenalis cathepsin B cysteine proteases attenuates CXCL8-induced chemotaxis of human neutrophils. Taken together, these data demonstrate for the first time that G. duodenalis trophozoite cathepsins are capable of attenuating a component of their host's proinflammatory response induced by a separate proinflammatory stimulus.


Asunto(s)
Quimiotaxis/efectos de los fármacos , Giardia lamblia/enzimología , Interleucina-8/metabolismo , Mucosa Intestinal/metabolismo , Neutrófilos/fisiología , Células Cultivadas , Quimiotaxis/fisiología , Enfermedad de Crohn/metabolismo , Giardia lamblia/genética , Giardia lamblia/metabolismo , Humanos , Interleucina-8/genética , Mucosa Intestinal/parasitología , Neutrófilos/metabolismo , Péptido Hidrolasas , Salmonella typhimurium
18.
Antimicrob Agents Chemother ; 58(8): 4298-307, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24820086

RESUMEN

The accumulation of neutrophils and proinflammatory mediators, such as leukotriene B4 (LTB4), is a classic marker of inflammatory disease. The clearance of apoptotic neutrophils, inhibition of proinflammatory signaling, and production of proresolving lipids (including lipoxins, such as lipoxin A4 [LXA4]) are imperative for resolving inflammation. Tulathromycin (TUL), a macrolide used to treat bovine respiratory disease, confers immunomodulatory benefits via mechanisms that remain unclear. We recently reported the anti-inflammatory properties of TUL in bovine phagocytes in vitro and in Mannheimia haemolytica-challenged calves. The findings demonstrated that this system offers a powerful model for investigating novel mechanisms of pharmacological immunomodulation. In the present study, we examined the effects of TUL in a nonbacterial model of pulmonary inflammation in vivo and characterized its effects on lipid signaling. In bronchoalveolar lavage (BAL) fluid samples from calves challenged with zymosan particles (50 mg), treatment with TUL (2.5 mg/kg of body weight) significantly reduced pulmonary levels of LTB4 and prostaglandin E2 (PGE2). In calcium ionophore (A23187)-stimulated bovine neutrophils, TUL inhibited phospholipase D (PLD), cytosolic phospholipase A2 (PLA2) activity, and the release of LTB4. In contrast, TUL promoted the secretion of LXA4 in resting and A23187-stimulated neutrophils, while levels of its precursor, 15(S)-hydroxyeicosatetraenoic acid [15(S)-HETE], were significantly lower. These findings indicate that TUL directly modulates lipid signaling by inhibiting the production of proinflammatory eicosanoids and promoting the production of proresolving lipoxins.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Dinoprostona/antagonistas & inhibidores , Disacáridos/farmacología , Compuestos Heterocíclicos/farmacología , Leucotrieno B4/antagonistas & inhibidores , Lipoxinas/agonistas , Fosfolipasas A2/metabolismo , Neumonía/tratamiento farmacológico , Animales , Líquido del Lavado Bronquioalveolar/química , Calcimicina/farmacología , Bovinos , Dinoprostona/biosíntesis , Ácidos Hidroxieicosatetraenoicos/metabolismo , Leucotrieno B4/biosíntesis , Lipoxinas/biosíntesis , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/patología , Material Particulado , Neumonía/inducido químicamente , Neumonía/metabolismo , Neumonía/patología , Cultivo Primario de Células , Zimosan
19.
Crit Rev Microbiol ; 40(1): 1-17, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23297858

RESUMEN

Enterocyte turnover along with proper epithelial barrier function are crucial aspects of mucosal defense. Apoptosis is a highly regulated type of programmed cell death that allows for the homeostatic turnover of the epithelial layer. Recent studies have suggested that microbial modulation of enterocyte apoptosis can result in increased epithelial permeability, leading to gastrointestinal pathophysiology. In this review, we highlight key mechanisms and pathways via which various viral, bacterial and parasitic pathogens are able to modulate enterocyte apoptosis. We also discuss how these alterations to enterocyte apoptosis can result in the activation of chronic gastrointestinal disorders, such as allergies, irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD). The role of proteinase-activated receptors in the pathogenesis of modulated apoptosis-induced pathogenesis is also discussed. Newly discovered processes, through which host epithelial cells may have evolved, rescue mechanisms from microbe-induced apoptosis are discussed. Together, these mechanisms are key to our ever-increasing understanding of host-microbe interactions in the gut.


Asunto(s)
Apoptosis , Bacterias/inmunología , Enterocitos/inmunología , Parásitos/inmunología , Virus/inmunología , Animales , Enterocitos/fisiología , Interacciones Huésped-Patógeno , Humanos , Hipersensibilidad/etiología , Hipersensibilidad/inmunología , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/inmunología , Síndrome del Colon Irritable/etiología , Síndrome del Colon Irritable/inmunología , Receptores Proteinasa-Activados/metabolismo
20.
Antimicrob Agents Chemother ; 57(3): 1385-93, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23295921

RESUMEN

Recent evidence indicates that immunomodulation by antibiotics may enhance their clinical efficacy. Specifically, drug-induced leukocyte apoptosis and macrophage efferocytosis have been shown to promote the resolution of inflammation in a variety of disease settings. Tulathromycin is a new macrolide antibiotic for the treatment of bovine respiratory disease. The direct antimicrobial effects of the drug alone do not fully justify its superior clinical efficacy, and we hypothesize that tulathromycin may have immunomodulating properties. We recently reported that tulathromycin promotes apoptosis and inhibits proinflammatory NF-κB signaling in bovine neutrophils. In this study, we investigated the direct and indirect anti-inflammatory effects of tulathromycin in bovine macrophages. The findings indicate that bovine monocyte-derived macrophages and alveolar macrophages readily phagocytose tulathromycin-induced apoptotic neutrophils both in vitro and in the airways of Mannheimia haemolytica-infected calves. Moreover, tulathromycin promotes delayed, concentration-dependent apoptosis, but not necrosis, in bovine macrophages in vitro. Activation of caspase-3 and detection of mono- and oligonucleosomes in bovine monocyte-derived macrophages treated with tulathromycin was observed 12 h posttreatment; pretreatment with a pan-caspase inhibitor (ZVAD) blocked the proapoptotic effects of the drug. Lastly, tulathromycin inhibited the secretion of proinflammatory CXCL-8 in lipopolysaccharide (LPS)-stimulated bovine macrophages; this effect was independent of caspase activation or programmed cell death. Taken together, these immunomodulating effects observed in bovine macrophages help further elucidate the mechanisms through which tulathromycin confers anti-inflammatory and proresolution benefits. Furthermore, these findings offer novel insights on how antibiotics may offer anti-inflammatory benefits by modulating macrophage-mediated events that play a key role in inflammation.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Apoptosis/efectos de los fármacos , Disacáridos/farmacología , Compuestos Heterocíclicos/farmacología , Factores Inmunológicos/farmacología , Interleucina-8/antagonistas & inhibidores , Macrófagos Alveolares/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Neumonía Enzoótica de los Becerros/tratamiento farmacológico , Animales , Apoptosis/inmunología , Caspasa 3/genética , Caspasa 3/metabolismo , Bovinos , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/inmunología , Inflamación/prevención & control , Interleucina-8/biosíntesis , Interleucina-8/inmunología , Lipopolisacáridos/farmacología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Masculino , Mannheimia haemolytica/efectos de los fármacos , Mannheimia haemolytica/crecimiento & desarrollo , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/microbiología , Oligopéptidos/farmacología , Neumonía Enzoótica de los Becerros/inmunología , Neumonía Enzoótica de los Becerros/patología , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA