Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Pediatr Surg Int ; 34(3): 323-330, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29196880

RESUMEN

BACKGROUND: Necrotizing enterocolitis (NEC) is a gastrointestinal disease of complex etiology resulting in devastating systemic inflammation and often death in premature newborns. We previously demonstrated that formula feeding inhibits ileal expression of heat shock protein-70 (Hsp70), a critical stress protein within the intestine. Barrier function for the premature intestine is critical. We sought to determine whether reduced Hsp70 protein expression increases neonatal intestinal permeability. METHODS: Young adult mouse colon cells (YAMC) were utilized to evaluate barrier function as well as intestine from Hsp70-/- pups (KO). Sections of intestine were analyzed by Western blot, immunohistochemistry, and real time PCR. YAMC cells were sub-lethally heated or treated with expressed milk (EM) to induce Hsp70. RESULTS: Immunostaining demonstrates co-localized Hsp70 and tight junction protein zona occludens-1 (ZO-1), suggesting physical interaction to protect tight junction function. The permeability of YAMC monolayers increases following oxidant injury and is partially blocked by Hsp70 induction either by prior heat stress or EM. RT-PCR analysis demonstrated that the Hsp70 isoforms, 70.1 and 70.3, predominate in WT pup; however, Hsp70.2 predominates in the KO pups. While Hsp70 is present in WT milk, it is not present in KO EM. Hsp70 associates with ZO-1 to maintain epithelial barrier function. CONCLUSION: Both induction of Hsp70 and exposure to EM prevent stress-induced increased permeability. Hsp70.2 is present in both WT and KO neonatal intestine, suggesting a crucial role in epithelial integrity. Induction of the Hsp70.2 isoform appears to be mediated by mother's milk. These results suggest that mother's milk feeding modulates Hsp70.2 expression and could attenuate injury leading to NEC. LEVEL OF EVIDENCE: Level III.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Mucosa Intestinal/metabolismo , Leche/metabolismo , Animales , Animales Recién Nacidos , Citoprotección , Proteínas HSP70 de Choque Térmico/genética , Ratones , Permeabilidad , Isoformas de Proteínas , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
2.
J Biol Chem ; 288(48): 34757-66, 2013 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-24129565

RESUMEN

Inflammatory immune responses play an important role in mucosal homeostasis and gut diseases. Nuclear factor κB (NF-κB), central to the proinflammatory cascade, is activated in necrotizing enterocolitis (NEC), a devastating condition of intestinal injury with extensive inflammation in premature infants. TGF-ß is a strong immune suppressor and a factor in breast milk, which has been shown to be protective against NEC. In an NEC animal model, oral administration of the isoform TGF-ß1 activated the downstream effector Smad2 in intestine and significantly reduced NEC incidence. In addition, TGF-ß1 suppressed NF-κB activation, maintained levels of the NF-κB inhibitor IκBα in the intestinal epithelium, and systemically decreased serum levels of IL-6 and IFN-γ. The immature human fetal intestinal epithelial cell line H4 was used as a reductionistic model of the immature enterocyte to investigate mechanism. TGF-ß1 pretreatment inhibited the TNF-α-induced IκBα phosphorylation that targets the IκBα protein for degradation and inhibited NF-κB activation. Chromatin immunoprecipitation (ChIP) assays demonstrated decreased NF-κB binding to the promoters of IL-6, IL-8, and IκBα in response to TNF-α with TGF-ß1 pretreatment. These TGF-ß1 effects appear to be mediated through the canonical Smad pathway as silencing of the TGF-ß central mediator Smad4 resulted in loss of the TGF-ß1 effects. Thus, TGF-ß1 is capable of eliciting anti-inflammatory effects by inhibiting NF-κB specifically in the intestinal epithelium as well as by decreasing systemic IL-6 and IFN-γ levels. Oral administration of TGF-ß1 therefore can potentially be used to protect against gastrointestinal diseases.


Asunto(s)
Enfermedades Gastrointestinales/tratamiento farmacológico , Inflamación/metabolismo , FN-kappa B/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/administración & dosificación , Administración Oral , Animales , Línea Celular , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/patología , Humanos , Inmunidad Innata/genética , Inflamación/tratamiento farmacológico , Inflamación/genética , Interferón gamma/metabolismo , Interleucina-6/metabolismo , Interleucina-8 , Mucosa Intestinal/lesiones , Mucosa Intestinal/metabolismo , Regiones Promotoras Genéticas , Ratas , Transducción de Señal , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
3.
Pediatr Res ; 69(5 Pt 1): 395-400, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21263375

RESUMEN

Preterm infants face many challenges in transitioning from the in utero to extrauterine environment while still immature. Failure of the preterm gut to successfully mature to accommodate bacteria and food substrate leads to significant morbidity such as neonatal necrotizing enterocolitis. The intestinal epithelial barrier plays a critical role in gut protection. Heat shock protein 70 (Hsp70) is an inducible cytoprotective molecule shown to protect the intestinal epithelium in adult models. To investigate the hypothesis that Hsp70 may be important for early protection of the immature intestine, Hsp70 expression was evaluated in intestine of immature rat pups. Data demonstrate that Hsp70 is induced by exposure to mother's milk. Hsp70 is found in mother's milk, and increased Hsp70 transcription is induced by mother's milk. This Hsp70 colocalizes with the tight junction protein ZO-1. Mother's milk-induced Hsp70 may contribute to maintenance of barrier function in the face of oxidant stress. Further understanding of the means by which mother's milk increases Hsp70 in the ileum will allow potential means of strengthening the intestinal barrier in at-risk preterm infants.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Íleon/metabolismo , Mucosa Intestinal/metabolismo , Leche/metabolismo , Animales , Animales Recién Nacidos , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/microbiología , Citoprotección , Modelos Animales de Enfermedad , Proteínas HSP70 de Choque Térmico/genética , Hipoxia/metabolismo , Íleon/microbiología , Íleon/patología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Proteínas de la Membrana/metabolismo , Estrés Oxidativo , Permeabilidad , Fosfoproteínas/metabolismo , ARN Mensajero/metabolismo , Ratas , Uniones Estrechas/metabolismo , Regulación hacia Arriba , Proteína de la Zonula Occludens-1
4.
PLoS One ; 10(4): e0124504, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25928420

RESUMEN

BACKGROUND AND AIMS: It is known that postnatal functional maturation of the small intestine is facilitated by microbial colonization of the gut. Preterm infants exhibit defects in gut maturation, weak innate immunity against intestinal infection and increased susceptibility to inflammatory disorders, all of which may be related to the inappropriate microbial colonization of their immature intestines. The earliest microbes to colonize the preterm infant gut encounter a naïve, immature intestine. Thus this earliest microbiota potentially has the greatest opportunity to fundamentally influence intestinal development and immune function. The aim of this study was to characterize the effect of early microbial colonization on global gene expression in the distal small intestine during postnatal gut development. METHODS: Gnotobiotic mouse models with experimental colonization by early (prior to two weeks of life) intestinal microbiota from preterm human infants were utilized. Microarray analysis was used to assess global gene expression in the intestinal epithelium. RESULTS AND CONCLUSION: Multiple intestinal genes involved in metabolism, cell cycle regulation, cell-cell or cell-extracellular matrix communication, and immune function are developmental- and intestinal microbiota- regulated. Using a humanized gnotobiotic mouse model, we demonstrate that certain early preterm infant microbiota from prior to 2 weeks of life specifically induce increased NF-κB activation and a phenotype of increased inflammation whereas other preterm microbiota specifically induce decreased NF-κB activation. These fundamental differences correlate with altered clinical outcomes and suggest the existence of optimal early microbial communities to improve health outcomes.


Asunto(s)
Inflamación/genética , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Microbiota/genética , Animales , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Vida Libre de Gérmenes , Humanos , Recien Nacido Prematuro , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Embarazo
5.
J Gastrointest Surg ; 6(4): 563-8, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12127122

RESUMEN

Mutations of the adenomatous polyposis coli (APC) gene are implicated early in colorectal tumorigenesis. Restoration of normal APC expression through gene therapy may prevent or reduce intestinal neoplasia. Furthermore, the relationship between colorectal tumors and increased cyclooxygenase-2 (COX-2) activity provides a rationale for the use of selective COX-2 inhibitors such as rofecoxib (Vioxx) to prevent the formation of polyps. This study was performed to determine the effects of liposome-mediated APC gene therapy and a selective COX-2 inhibitor on intestinal neoplasia in vivo. Five-week-old Min mice weaned on a 30% high-fat diet were randomized to receive no treatment (control), APC only, Vioxx only, and APC/Vioxx. APC-treated mice received a plasmid containing the human APC cDNA (pCMV-APC) mixed with a liposome preparation that was administered biweekly. Vioxx was administered at 200 ppm in the high-fat rodent chow. The control mice were treated similarly with a plasmid construct lacking the APC gene. Confirmation of exogenous APC gene expression was determined by Western blot analysis. After 2 months, there was a 54% and 70% reduction in the total number of intestinal polyps after APC and Vioxx treatment, respectively. Combined APC/Vioxx therapy reduced polyp formation by 87%. The reduction of intestinal neoplasia by APC gene replacement and COX-2 inhibition suggests their separate roles in intestinal tumorigenesis. Each modality, both individually and together, may prove therapeutic and therefore contribute to new strategies in the prevention and treatment of colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/terapia , Inhibidores de la Ciclooxigenasa/uso terapéutico , Genes APC , Terapia Genética , Isoenzimas/antagonistas & inhibidores , Lactonas/uso terapéutico , Animales , Neoplasias Colorrectales/genética , Terapia Combinada , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Masculino , Ratones , Prostaglandina-Endoperóxido Sintasas , Sulfonas
6.
PLoS One ; 8(7): e69620, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936061

RESUMEN

Neonatal necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease of preterm infants. Increased intestinal epithelium permeability is an early event in NEC pathogenesis. Autophagy and apoptosis are induced by multiple stress pathways which may impact the intestinal barrier, and they have been associated with pathogenesis of diverse gastrointestinal diseases including inflammatory bowel disease. Using both in vitro and in vivo models, this study investigates autophagy and apoptosis under experimental NEC stresses. Furthermore this study evaluates the effect of erythropoietin (Epo), a component of breast milk previously shown to decrease the incidence of NEC and to preserve intestinal barrier function, on intestinal autophagy and apoptosis. It was found that autophagy and apoptosis are both rapidly up regulated in NEC in vivo as indicated by increased expression of the autophagy markers Beclin 1 and LC3II, and by evidence of apoptosis by TUNEL and cleaved caspase-3 staining. In the rat NEC experimental model, autophagy preceded the onset of apoptosis in intestine. In vitro studies suggested that Epo supplementation significantly decreased both autophagy and apoptosis via the Akt/mTOR signaling pathway and the MAPK/ERK pathway respectively. These results suggest that Epo protects intestinal epithelium from excessive autophagy and apoptosis in experimental NEC.


Asunto(s)
Apoptosis , Autofagia , Enterocolitis Necrotizante/prevención & control , Células Epiteliales/efectos de los fármacos , Eritropoyetina/farmacología , Mucosa Intestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Animales , Animales Recién Nacidos , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Caspasa 3/genética , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/genética , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Eritropoyetina/genética , Eritropoyetina/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica , Humanos , Recién Nacido , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Intestinos/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Leche Humana/química , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
7.
PLoS One ; 8(5): e65108, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23717690

RESUMEN

Balance among the complex interactions of the gut microbial community is important for intestinal health. Probiotic bacteria can improve bacterial balance and have been used to treat gastrointestinal diseases. Neonatal necrotizing enterocolitis (NEC) is a life-threatening inflammatory bowel disorder primarily affecting premature infants. NEC is associated with extensive inflammatory NF-κB signaling activation as well as intestinal barrier disruption. Clinical studies have shown that probiotic administration may protect against NEC, however there are safety concerns associated with the ingestion of large bacterial loads in preterm infants. Bacteria-free conditioned media (CM) from certain probiotic organisms have been shown to retain bioactivity including anti-inflammatory and cytoprotective properties without the risks of live organisms. We hypothesized that the CM from Lactobacillus acidophilus (La), Bifidobacterium infantis (Bi), and Lactobacillus plantarum (Lp), used separately or together would protect against NEC. A rodent model with intestinal injury similar to NEC was used to study the effect of CM from Lp, La/Bi, and La/Bi/Lp on the pathophysiology of NEC. All the CM suppressed NF-κB activation via preserved IκBα expression and this protected IκBα was associated with decreased liver activity of the proteasome, which is the degrading machinery for IκBα. These CM effects also caused decreases in intestinal production of the pro-inflammatory cytokine TNF-α, a downstream target of the NF-κB pathway. Combined La/Bi and La/Bi/Lp CM in addition protected intestinal barrier function by maintaining tight junction protein ZO-1 levels and localization at the tight junction. Double combined La/Bi CM significantly reduced intestinal injury incidence from 43% to 28% and triple combined La/Bi/Lp CM further reduced intestinal injury incidence to 20%. Thus, this study demonstrates different protective mechanisms and synergistic bioactivity of the CM from different organisms in ameliorating NEC-like intestinal injury in an animal model.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Enterocolitis Necrotizante/prevención & control , Probióticos/administración & dosificación , Animales , Animales Recién Nacidos , Enterocolitis Necrotizante/metabolismo , Femenino , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Intestinos/microbiología , Intestinos/patología , FN-kappa B/metabolismo , Embarazo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratas , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Proteína de la Zonula Occludens-1/metabolismo
8.
Inflamm Bowel Dis ; 15(10): 1537-47, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19373789

RESUMEN

BACKGROUND: Bacteria play a role in inflammatory bowel disease and other forms of intestinal inflammation. Although much attention has focused on the search for a pathogen or inciting inflammatory bacteria, another possibility is a lack of beneficial bacteria that normally confer anti-inflammatory properties in the gut. The purpose of this study was to determine whether normal commensal bacteria could inhibit inflammatory pathways important in intestinal inflammation. METHODS: Conditioned media from Lactobacillus plantarum (Lp-CM) and other gut bacteria was used to treat intestinal epithelial cell (YAMC) and macrophage (RAW 264.7) or primary culture murine dendritic cells. NF-kappaB was activated through TNF-Receptor, MyD88-dependent and -independent pathways and effects of Lp-CM on the NF-kappaB pathway were assessed. NF-kappaB binding activity was measured using ELISA and EMSA. 1kappaB expression was assessed by Western blot analysis, and proteasome activity determined using fluorescence-based proteasome assays. MCP-1 release was determined by ELISA. RESULTS: Lp-CM inhibited NF-kappaB binding activity, degradation of IkappaBalpha and the chymotrypsin-like activity of the proteasome. Moreover, Lp-CM directly inhibited the activity of purified mouse proteasomes. This effect was specific, since conditioned media from other bacteria had no inhibitory effect. Unlike other proteasome inhibitors, Lp-CM was not toxic in cell death assays. Lp-CM inhibited MCP-1 release in all cell types tested. CONCLUSIONS: These studies confirm, and provide a mechanism for, the anti-inflammatory effects of the probiotic and commensal bacterium Lactobacillus plantarum. The use of bacteria-free Lp-CM provides a novel strategy for treatment of intestinal inflammation which would eliminate the risk of bacteremia reported with conventional probiotics.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Mucosa Intestinal/metabolismo , Lactobacillus plantarum , FN-kappa B/antagonistas & inhibidores , Probióticos/farmacología , Inhibidores de Proteasoma , Animales , Western Blotting , Células Cultivadas , Quimiocina CCL2/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/microbiología , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Femenino , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Intestinos/inmunología , Intestinos/microbiología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratas , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Ubiquitina/metabolismo
9.
EMBO Rep ; 8(6): 563-8, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17468738

RESUMEN

The steroid hormone receptors are characterized by binding to relatively rigid, inflexible endogenous steroid ligands. Other members of the nuclear receptor superfamily bind to conformationally flexible lipids and show a corresponding degree of elasticity in the ligand-binding pocket. Here, we report the X-ray crystal structure of the oestrogen receptor alpha (ERalpha) bound to an oestradiol derivative with a prosthetic group, ortho- trifluoromethlyphenylvinyl, which binds in a novel extended pocket in the ligand-binding domain. Unlike ER antagonists with bulky side groups, this derivative is enclosed in the ligand-binding pocket, and acts as a potent agonist. This work shows that steroid hormone receptors can interact with a wider array of pharmacophores than previously thought through structural plasticity in the ligand-binding pocket.


Asunto(s)
Estradiol/metabolismo , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/metabolismo , Línea Celular Tumoral , Cristalografía por Rayos X , Estradiol/química , Humanos , Ligandos , Modelos Moleculares , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
10.
J Biol Chem ; 281(26): 17909-19, 2006 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-16648639

RESUMEN

Estrogen receptors alpha (ERalpha) and beta (ERbeta) have distinct functions and differential expression in certain tissues. These differences have stimulated the search for subtype-selective ligands. Therapeutically, such ligands offer the potential to target specific tissues or pathways regulated by one receptor subtype without affecting the other. As reagents, they can be utilized to probe the physiological functions of the ER subtypes to provide information complementary to that obtained from knock-out animals. A fluorescence resonance energy transfer-based assay was used to screen a 10,000-compound chemical library for ER agonists. From the screen, we identified a family of ERbeta-selective agonists whose members contain bulky oxabicyclic scaffolds in place of the planar scaffolds common to most ER ligands. These agonists are 10-50-fold selective for ERbeta in competitive binding assays and up to 60-fold selective in transactivation assays. The weak uterotrophic activity of these ligands in immature rats and their ability to stimulate expression of an ERbeta regulated gene in human U2OS osteosarcoma cells provides more physiological evidence of their ERbeta-selective nature. To provide insight into the molecular mechanisms of their activity and selectivity, we determined the crystal structures of the ERalpha ligand-binding domain (LBD) and a peptide from the glucocorticoid receptor-interacting protein 1 (GRIP1) coactivator complexed with the ligands OBCP-3M, OBCP-2M, and OBCP-1M. These structures illustrate how the bicyclic scaffolds of these ligands are accommodated in the flexible ligand-binding pocket of ER. A comparison of these structures with existing ER structures suggests that the ERbeta selectivity of OBCP ligands can be attributed to a combination of their interactions with Met-336 in ERbeta and Met-421 in ERalpha. These bicyclic ligands show promise as lead compounds that can target ERbeta. In addition, our understanding of the molecular determinants of their subtype selectivity provides a useful starting point for developing other ER modulators belonging to this relatively new structural class.


Asunto(s)
Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/agonistas , Receptor beta de Estrógeno/metabolismo , Animales , Sitios de Unión , Línea Celular Tumoral , Cristalografía , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/química , Receptor beta de Estrógeno/genética , Femenino , Genisteína/química , Genisteína/metabolismo , Inhibidores de Crecimiento/química , Inhibidores de Crecimiento/metabolismo , Humanos , Ligandos , Metionina/metabolismo , Osteosarcoma , Fenol/química , Fenol/metabolismo , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/metabolismo , Transfección , Útero/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA