Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Immunol ; 196(12): 5130-7, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27194788

RESUMEN

Semapimod, a tetravalent guanylhydrazone, suppresses inflammatory cytokine production and has potential in a variety of inflammatory and autoimmune disorders. The mechanism of action of Semapimod is not well understood. In this study, we demonstrate that in rat IEC-6 intestinal epithelioid cells, Semapimod inhibits activation of p38 MAPK and NF-κB and induction of cyclooxygenase-2 by TLR ligands, but not by IL-1ß or stresses. Semapimod inhibits TLR4 signaling (IC50 ≈0.3 µmol) and acts by desensitizing cells to LPS; it fails to block responses to LPS concentrations of ≥5 µg/ml. Inhibition of TLR signaling by Semapimod is almost instantaneous: the drug is effective when applied simultaneously with LPS. Semapimod blocks cell-surface recruitment of the MyD88 adapter, one of the earliest events in TLR signaling. gp96, the endoplasmic reticulum-localized chaperone of the HSP90 family critically involved in the biogenesis of TLRs, was identified as a target of Semapimod using ATP-desthiobiotin pulldown and mass spectroscopy. Semapimod inhibits ATP-binding and ATPase activities of gp96 in vitro (IC50 ≈0.2-0.4 µmol). On prolonged exposure, Semapimod causes accumulation of TLR4 and TLR9 in perinuclear space, consistent with endoplasmic reticulum retention, an anticipated consequence of impaired gp96 chaperone function. Our data indicate that Semapimod desensitizes TLR signaling via its effect on the TLR chaperone gp96. Fast inhibition by Semapimod is consistent with gp96 participating in high-affinity sensing of TLR ligands in addition to its role as a TLR chaperone.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Hidrazonas/farmacología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Biotina/análogos & derivados , Biotina/metabolismo , Línea Celular , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/inmunología , Retículo Endoplásmico/metabolismo , Enterocitos/inmunología , Células HEK293 , Humanos , Interleucina-1beta/inmunología , Intestinos/citología , Lipopolisacáridos/inmunología , Espectrometría de Masas , Factor 88 de Diferenciación Mieloide/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Ratas , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 9/efectos de los fármacos , Receptor Toll-Like 9/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
Lab Invest ; 93(12): 1265-75, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24126890

RESUMEN

The intestinal barrier becomes compromised during systemic inflammation, leading to the entry of luminal bacteria into the host and gut origin sepsis. Pathogenesis and treatment of inflammatory gut barrier failure is an important problem in critical care. In this study, we examined the role of cyclooxygenase-2 (COX-2), a key enzyme in the production of inflammatory prostanoids, in gut barrier failure during experimental peritonitis in mice. I.p. injection of LPS or cecal ligation and puncture (CLP) increased the levels of COX-2 and its product prostaglandin E2 (PGE2) in the ileal mucosa, caused pathologic sloughing of the intestinal epithelium, increased passage of FITC-dextran and bacterial translocation across the barrier, and increased internalization of the tight junction (TJ)-associated proteins junction-associated molecule-A and zonula occludens-1. Luminal instillation of PGE2 in an isolated ileal loop increased transepithelial passage of FITC-dextran. Low doses (0.5-1 mg/kg), but not a higher dose (5 mg/kg) of the specific COX-2 inhibitor Celecoxib partially ameliorated the inflammatory gut barrier failure. These results demonstrate that high levels of COX-2-derived PGE2 seen in the mucosa during peritonitis contribute to gut barrier failure, presumably by compromising TJs. Low doses of specific COX-2 inhibitors may blunt this effect while preserving the homeostatic function of COX-2-derived prostanoids. Low doses of COX-2 inhibitors may find use as an adjunct barrier-protecting therapy in critically ill patients.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Mucosa Intestinal/efectos de los fármacos , Peritonitis/tratamiento farmacológico , Pirazoles/administración & dosificación , Sulfonamidas/administración & dosificación , Animales , Celecoxib , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Íleon/efectos de los fármacos , Íleon/enzimología , Mucosa Intestinal/enzimología , Ratones , Ratones Endogámicos C57BL , Permeabilidad/efectos de los fármacos
3.
Gut ; 61(10): 1417-25, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22138535

RESUMEN

BACKGROUND: Necrotising enterocolitis (NEC) is one of the most common and fatal intestinal disorders in preterm infants. Breast-fed infants are at lower risk for NEC than formula-fed infants, but the protective components in human milk have not been identified. In contrast to formula, human milk contains high amounts of complex glycans. OBJECTIVE: To test the hypothesis that human milk oligosaccharides (HMO) contribute to the protection from NEC. METHODS: Since human intervention studies are unfeasible due to limited availability of HMO, a neonatal rat NEC model was used. Pups were orally gavaged with formula without and with HMO and exposed to hypoxia episodes. Ileum sections were scored blindly for signs of NEC. Two-dimensional chromatography was used to determine the most effective HMO, and sequential exoglycosidase digestions and linkage analysis was used to determine its structure. RESULTS: Compared to formula alone, pooled HMO significantly improved 96-hour survival from 73.1% to 95.0% and reduced pathology scores from 1.98 ± 1.11 to 0.44 ± 0.30 (p<0.001). Within the pooled HMO, a specific isomer of disialyllacto-N-tetraose (DSLNT) was identified to be protective. Galacto-oligosaccharides, currently added to formula to mimic some of the effects of HMO, had no effect. CONCLUSION: HMO reduce NEC in neonatal rats and the effects are highly structure specific. If these results translate to NEC in humans, DSLNT could be used to prevent or treat NEC in formula-fed infants, and its concentration in the mother's milk could serve as a biomarker to identify breast-fed infants at risk of developing this disorder.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Leche Humana/química , Oligosacáridos/uso terapéutico , Animales , Cromatografía Líquida de Alta Presión , Cromatografía por Intercambio Iónico , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/mortalidad , Enterocolitis Necrotizante/patología , Cromatografía de Gases y Espectrometría de Masas , Humanos , Oligosacáridos/análisis , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
4.
Lab Invest ; 91(11): 1668-79, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21788941

RESUMEN

P-glycoprotein (Pgp), a product of the multi-drug resistance gene MDR1a, is a broad specificity efflux ATP cassette transmembrane transporter that is predominantly expressed in epithelial tissues. Because mdr1a(-/-) mice tend to develop spontaneous colitis in bacteria-dependent manner, Pgp is believed to have a role in protection of the intestinal epithelium from luminal bacteria. Here we demonstrate that levels of Pgp in the small intestine of newborn rodents dramatically increase during breastfeeding, but not during formula feeding (FF). In rats and mice, levels of intestinal Pgp peak on days 3-7 and 1-5 of breastfeeding, respectively. The mdr1a(-/-) neonatal mice subjected to FF, hypoxia, and hypothermia have significantly higher incidence and pathology, as well as significantly earlier onset of necrotizing enterocolitis (NEC) than congenic wild type mice. Breast-fed mdr1a(-/-) neonatal mice are also more susceptible to intestinal damage caused by the opportunistic pathogen Cronobacter sakazakii that has been associated with hospital outbreaks of NEC. Breast milk, but not formula, induces Pgp expression in enterocyte cell lines in a dose- and time-dependent manner. High levels of ectopically expressed Pgp protect epithelial cells in vitro from apoptosis induced by C. sakazakii. Taken together, these results show that breast milk-induced expression of Pgp may have a role in the protection of the neonatal intestinal epithelium from injury associated with nascent bacterial colonization.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Enterocolitis Necrotizante/patología , Regulación del Desarrollo de la Expresión Génica/fisiología , Inflamación/patología , Intestino Delgado/microbiología , Leche/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Animales , Animales Recién Nacidos , Western Blotting , Cronobacter sakazakii , Cartilla de ADN/genética , Infecciones por Enterobacteriaceae/metabolismo , Enterocolitis Necrotizante/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Inflamación/metabolismo , Ratones , Ratones Noqueados , Sustitutos de la Leche/farmacología , Plásmidos/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadísticas no Paramétricas , Transfección
5.
J Immunol ; 183(2): 1384-92, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19570823

RESUMEN

Although enterocytes are capable of innate immune responses, the intestinal epithelium is normally tolerant to commensal bacteria. To elucidate the mechanisms of tolerance, we examined the effect of preexposure to LPS on activation of p38, c-Jun, and NF-kappaB in enterocytes by several inflammatory and stress stimuli. Shortly after the initial LPS challenge, enterocytes become tolerant to restimulation with LPS or CpG DNA, but not with IL-17 or UV. The state of tolerance, which lasts 20-26 h, temporally coincides with LPS-induced expression of the anti-inflammatory ubiquitin-editing enzyme A20. Small interfering RNA silencing of A20 prevents tolerance, whereas ectopic expression of A20 blocks responses to LPS and CpG DNA, but not to IL-17 or UV. A20 levels in the epithelium of the small intestine are low at birth and following gut decontamination with antibiotics, but high under conditions of bacterial colonization. In the small intestine of adult rodents, A20 prominently localizes to the luminal interface of villus enterocytes. Lower parts of the crypts display relatively low levels of A20, but relatively high levels of phospho-p38. Gut decontamination with antibiotics reduces the levels of both A20 and phospho-p38. Along with the fact that A20-deficient mice develop severe intestinal inflammation, our results indicate that induction of A20 plays a key role in the tolerance of the intestinal epithelium to TLR ligands and bacteria.


Asunto(s)
Cisteína Endopeptidasas/inmunología , Enterocitos/inmunología , Tolerancia Inmunológica , Péptidos y Proteínas de Señalización Intracelular/inmunología , Lipopolisacáridos/inmunología , Animales , Islas de CpG , Cisteína Endopeptidasas/genética , Gastroenteritis/etiología , Interleucina-17 , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Activación Transcripcional/efectos de los fármacos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
PLoS One ; 14(11): e0216762, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31675374

RESUMEN

Enterococcus faecalis is a ubiquitous intestinal symbiont and common early colonizer of the neonatal gut. Although colonization with E. faecalis has been previously associated with decreased pathology of necrotizing enterocolitis (NEC), these bacteria have been also implicated as opportunistic pathogens. Here we characterized 21 strains of E. faecalis, naturally occurring in 4-day-old rats, for potentially pathogenic properties and ability to colonize the neonatal gut. The strains differed in hemolysis, gelatin liquefaction, antibiotic resistance, biofilm formation, and ability to activate the pro-inflammatory transcription factor NF-κB in cultured enterocytes. Only 3 strains, BB70, 224, and BB24 appreciably colonized the neonatal intestine on day 4 after artificial introduction with the first feeding. The best colonizer, strain BB70, effectively displaced E. faecalis of maternal origin. Whereas BB70 and BB24 significantly increased NEC pathology, strain 224 significantly protected from NEC. Our results show that different strains of E. faecalis may be pathogenic or protective in experimental NEC.


Asunto(s)
Enterococcus faecalis/patogenicidad , Enterocolitis Necrotizante/microbiología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Enterococcus faecalis/clasificación , Enterococcus faecalis/genética , Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/prevención & control , Enterocitos/microbiología , Enterocitos/patología , Femenino , Variación Genética , Humanos , Recién Nacido , Intestinos/microbiología , Intestinos/patología , Fenotipo , Embarazo , Probióticos/uso terapéutico , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Virulencia
7.
PLoS One ; 13(6): e0196710, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29933378

RESUMEN

The use of lactobacilli in prevention of necrotizing enterocolitis (NEC) is hampered by insufficient knowledge about optimal species/strains and effects on intestinal bacterial populations. We therefore sought to identify lactobacilli naturally occurring in postnatal rats and examine their ability to colonize the neonatal intestine and protect from NEC. L. murinus, L. acidophilus, and L. johnsonii were found in 42, 20, and 1 out of 51 4-day old rats, respectively. Higher proportion of L. murinus in microbiota correlated with lower NEC scores. Inoculation with each of the three species during first feeding significantly augmented intestinal populations of lactobacilli four days later, indicating successful colonization. L. murinus, but not L. acidophilus or L. johnsonii, significantly protected against NEC. Thus, lactobacilli protect rats from NEC in a species- or strain-specific manner. Our results may help rationalizing probiotic therapy in NEC.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Microbioma Gastrointestinal , Intestinos/microbiología , Lactobacillus , Probióticos , Animales , Animales Recién Nacidos , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/patología , Intestinos/patología , Ratas , Ratas Sprague-Dawley
8.
PLoS One ; 12(11): e0188211, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29190745

RESUMEN

Necrotizing enterocolitis (NEC) is a significant cause of morbidity and mortality in premature infants; yet its pathogenesis remains poorly understood. To evaluate the role of intestinal bacteria in protection against NEC, we assessed the ability of naturally occurring intestinal colonizer E. coli EC25 to influence composition of intestinal microbiota and NEC pathology in the neonatal rat model. Experimental NEC was induced in neonatal rats by formula feeding/hypoxia, and graded histologically. Bacterial populations were characterized by plating on blood agar, scoring colony classes, and identifying each class by sequencing 16S rDNA. Binding of bacteria to, and induction of apoptosis in IEC-6 enterocytes were examined by plating on blood agar and fluorescent staining for fragmented DNA. E. coli EC 25, which was originally isolated from healthy rats, efficiently colonized the intestine and protected from NEC following introduction to newborn rats with formula at 106 or 108 cfu. Protection did not depend significantly on EC25 inoculum size or load in the intestine, but positively correlated with the fraction of EC25 in the microbiome. Introduction of EC25 did not prevent colonization with other bacteria and did not significantly alter bacterial diversity. EC25 neither induced cultured enterocyte apoptosis, nor protected from apoptosis induced by an enteropathogenic strain of Cronobacter muytjensii. Our results show that E. coli EC25 is a commensal strain that efficiently colonizes the neonatal intestine and protects from NEC.


Asunto(s)
Animales Recién Nacidos , Enterocolitis Necrotizante/prevención & control , Escherichia coli/fisiología , Animales , Apoptosis , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/patología , Enterocitos/patología , Femenino , Microbiota , Embarazo , Ratas , Ratas Sprague-Dawley
9.
J Infect Dis ; 198(4): 586-93, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18588483

RESUMEN

Necrotizing enterocolitis (NEC) is an inflammatory intestinal disorder that affects 2%-5% of all premature infants. Enterobacter sakazakii, a common contaminant of milk-based powdered infant formula, has been implicated as a causative agent of sepsis, meningitis, and NEC in newborn infants, with high mortality rates. However, the role played by E. sakazakii in the pathogenesis of NEC is, to date, not known. Here, we demonstrate for the first time that E. sakazakii can induce clinical and histological NEC in newborn rats. E. sakazakii was found to bind to enterocytes in rat pups at the tips of villi and to intestinal epithelial cells (IEC-6) in culture, with no significant invasion. Exposure to E. sakazakii induced apoptosis and increased the production of interleukin-6 in IEC-6 cells and in the animal model. These data suggest that E. sakazakii could be a potential pathogen that induces NEC and triggers intestinal disease by modulating enterocyte intracellular signaling pathways.


Asunto(s)
Apoptosis/fisiología , Cronobacter sakazakii/patogenicidad , Infecciones por Enterobacteriaceae/fisiopatología , Enterocolitis Necrotizante/patología , Células Epiteliales/patología , Mucosa Intestinal/microbiología , Animales , Cronobacter sakazakii/inmunología , Cronobacter sakazakii/fisiología , Modelos Animales de Enfermedad , Infecciones por Enterobacteriaceae/microbiología , Enterocolitis Necrotizante/microbiología , Enterocitos , Células Epiteliales/microbiología , Ratas
10.
J Immunol ; 176(1): 580-8, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16365453

RESUMEN

Necrotizing enterocolitis (NEC), a severe intestinal inflammation in neonates, occurs following bacterial colonization of the gut. LPS-induced production of inflammatory factors in immature enterocytes may be a factor in NEC. Previously, we described LPS-induced p38 MAPK-dependent expression of cyclooxygenase-2 (COX-2) in rat IEC-6 cells. In this study, we examine COX-2 expression in newborn rat intestinal epithelium and further characterize the mechanisms of COX-2 regulation in enterocytes. Induction of NEC by formula feeding/hypoxia increased phospho-p38 and COX-2 levels in the intestinal mucosa. Celecoxib, a selective COX-2 inhibitor, exacerbated the disease, suggesting a protective role for COX-2. COX-2 was induced in the intestinal epithelium by LPS in vivo and ex vivo. The latter response was attenuated by the p38 inhibitor SB202190, but not by inhibitors of ERK, JNK, or NF-kappaB. In IEC-6 enterocytes, COX-2 was induced by the expression of MAPK kinase 3 EE (MKK3EE), a constitutive activator of p38, but not of activators of ERK or JNK pathways. However, neither MKK3/6 nor MKK4, the known p38 upstream kinases, were activated by LPS. Dominant-negative MKK3 or MKK4 or SB202190 failed to prevent LPS-induced, p38-activating phosphorylation, ruling out important roles of these kinases or p38 autophosphorylation. LPS increased COX-2 and activating phosphorylation of p38 with similar dose-response. Blockade of LPS-induced expression of COX-2-luciferase reporter and destabilization of COX-2 message by SB202190 indicate that p38 regulates COX-2 at transcription and mRNA stability levels. Our data indicate that p38-mediated expression of COX-2 proceeds through a novel upstream pathway and support the role of the neonate's enterocytes as bacterial sensors.


Asunto(s)
Ciclooxigenasa 2/biosíntesis , Enterocitos/inmunología , Lipopolisacáridos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Animales , Animales Recién Nacidos , Western Blotting , Celecoxib , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/enzimología , Enterocolitis Necrotizante/inmunología , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Inhibidores Enzimáticos/farmacología , Microscopía Fluorescente , Pirazoles/farmacología , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfonamidas/farmacología , Transcripción Genética , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA