Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 325(1): G23-G41, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37120853

RESUMEN

Necrotizing enterocolitis (NEC) is the leading cause of morbidity and mortality in premature infants. One of the most devastating complications of NEC is the development of NEC-induced brain injury, which manifests as impaired cognition that persists beyond infancy and which represents a proinflammatory activation of the gut-brain axis. Given that oral administration of the human milk oligosaccharides (HMOs) 2'-fucosyllactose (2'-FL) and 6'-sialyslactose (6'-SL) significantly reduced intestinal inflammation in mice, we hypothesized that oral administration of these HMOs would reduce NEC-induced brain injury and sought to determine the mechanisms involved. We now show that the administration of either 2'-FL or 6'-SL significantly attenuated NEC-induced brain injury, reversed myelin loss in the corpus callosum and midbrain of newborn mice, and prevented the impaired cognition observed in mice with NEC-induced brain injury. In seeking to define the mechanisms involved, 2'-FL or 6'-SL administration resulted in a restoration of the blood-brain barrier in newborn mice and also had a direct anti-inflammatory effect on the brain as revealed through the study of brain organoids. Metabolites of 2'-FL were detected in the infant mouse brain by nuclear magnetic resonance (NMR), whereas intact 2'-FL was not. Strikingly, the beneficial effects of 2'-FL or 6'-SL against NEC-induced brain injury required the release of the neurotrophic factor brain-derived neurotrophic factor (BDNF), as mice lacking BDNF were not protected by these HMOs from the development of NEC-induced brain injury. Taken in aggregate, these findings reveal that the HMOs 2'-FL and 6'-SL interrupt the gut-brain inflammatory axis and reduce the risk of NEC-induced brain injury.NEW & NOTEWORTHY This study reveals that the administration of human milk oligosaccharides, which are present in human breast milk, can interfere with the proinflammatory gut-brain axis and prevent neuroinflammation in the setting of necrotizing enterocolitis, a major intestinal disorder seen in premature infants.


Asunto(s)
Lesiones Encefálicas , Disfunción Cognitiva , Enterocolitis Necrotizante , Humanos , Recién Nacido , Lactante , Femenino , Animales , Ratones , Leche Humana/metabolismo , Factor Neurotrófico Derivado del Encéfalo , Enfermedades Neuroinflamatorias , Enterocolitis Necrotizante/etiología , Oligosacáridos/farmacología , Oligosacáridos/uso terapéutico , Oligosacáridos/análisis , Disfunción Cognitiva/prevención & control , Disfunción Cognitiva/complicaciones , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/metabolismo
2.
Am J Physiol Gastrointest Liver Physiol ; 323(3): G265-G282, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35819175

RESUMEN

Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in premature infants and is steadily rising in frequency. Patients who develop NEC have a very high mortality, illustrating the importance of developing novel prevention or treatment approaches. We and others have shown that NEC arises in part from exaggerated signaling via the bacterial receptor, Toll-like receptor 4 (TLR4) on the intestinal epithelium, leading to widespread intestinal inflammation and intestinal ischemia. Strategies that limit the extent of TLR4 signaling, including the administration of amniotic fluid, can reduce NEC development in mouse and piglet models. We now seek to test the hypothesis that a secretome derived from amnion-derived cells can prevent or treat NEC in preclinical models of this disease via a process involving TLR4 inhibition. In support of this hypothesis, we show that the administration of this secretome, named ST266, to mice or piglets can prevent and treat experimental NEC. The protective effects of ST266 occurred in the presence of marked TLR4 inhibition in the intestinal epithelium of cultured epithelial cells, intestinal organoids, and human intestinal samples ex vivo, independent of epidermal growth factor. Strikingly, RNA-seq analysis of the intestinal epithelium in mice reveals that the ST266 upregulates critical genes associated with gut remodeling, intestinal immunity, gut differentiation. and energy metabolism. These findings show that the amnion-derived secretome ST266 can prevent and treat NEC, suggesting the possibility of novel therapeutic approaches for patients with this devastating disease.NEW & NOTEWORTHY This work provides hope for children who develop NEC, a devastating disease of premature infants that is often fatal, by revealing that the secreted product of amniotic progenitor cells (called ST266) can prevent or treat NEC in mice, piglet, and "NEC-in-a-dish" models of this disease. Mechanistically, ST266 prevented bacterial signaling, and a detailed transcriptomic analysis revealed effects on gut differentiation, immunity, and metabolism. Thus, an amniotic secretome may offer novel approaches for NEC.


Asunto(s)
Enterocolitis Necrotizante , Células Madre Multipotentes , Secretoma , Amnios/citología , Animales , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/prevención & control , Mucosa Intestinal/metabolismo , Ratones , Células Madre Multipotentes/metabolismo , Porcinos , Receptor Toll-Like 4/metabolismo
3.
Br J Nutr ; 128(6): 1050-1063, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34632971

RESUMEN

Necrotising enterocolitis (NEC) is a devastating gastrointestinal disease of prematurity that typically develops after the administration of infant formula, suggesting a link between nutritional components and disease development. One of the most significant complications that develops in patients with NEC is severe lung injury. We have previously shown that the administration of a nutritional formula that is enriched in pre-digested Triacylglyceride that do not require lipase action can significantly reduce the severity of NEC in a mouse model. We now hypothesise that this 'pre-digested fat (PDF) system' may reduce NEC-associated lung injury. In support of this hypothesis, we now show that rearing newborn mice on a nutritional formula based on the 'PDF system' promotes lung development, as evidenced by increased tight junctions and surfactant protein expression. Mice that were administered this 'PDF system' were significantly less vulnerable to the development of NEC-induced lung inflammation, and the administration of the 'PDF system' conferred lung protection. In seeking to define the mechanisms involved, the administration of the 'PDF system' significantly enhanced lung maturation and reduced the production of reactive oxygen species (ROS). These findings suggest that the PDF system protects the development of NEC-induced lung injury through effects on lung maturation and reduced ROS in the lung and also increases lung maturation in non-NEC mice.


Asunto(s)
Enterocolitis Necrotizante , Lesión Pulmonar , Animales , Ratones , Enterocolitis Necrotizante/etiología , Animales Recién Nacidos , Especies Reactivas de Oxígeno , Lesión Pulmonar/complicaciones , Lesión Pulmonar/metabolismo , Alimentos Formulados , Modelos Animales de Enfermedad
4.
Pediatr Res ; 89(1): 91-101, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32221473

RESUMEN

BACKGROUND: Necrotizing enterocolitis (NEC) develops through exaggerated toll-like receptor 4 (TLR4) signaling in the intestinal epithelium. Breast milk is rich in non-digestible oligosaccharides and prevents NEC through unclear mechanisms. We now hypothesize that the human milk oligosaccharides 2'-fucosyllactose (2'-FL) and 6'-sialyllactose (6'-SL) can reduce NEC through inhibition of TLR4 signaling. METHODS: NEC was induced in newborn mice and premature piglets and infant formula was supplemented with 2'-FL, 6'-SL, or lactose. Intestinal tissue was obtained at surgical resection. HMO inhibition of TLR4 was assessed in IEC-6 enterocytes, mice, and human tissue explants and via in silico modeling. RESULTS: Supplementation of infant formula with either 2'-FL and/or 6'-SL, but not the parent sugar lactose, reduced NEC in mice and piglets via reduced apoptosis, inflammation, weight loss, and histological appearance. Mechanistically, both 2'-FL and 6'-SL, but not lactose, reduced TLR4-mediated nuclear factor kappa light-chain enhancer of activated B cells (NF-kB) inflammatory signaling in the mouse and human intestine. Strikingly, in silico modeling revealed 2'-FL and 6'-SL, but not lactose, to dock into the binding pocket of the TLR4-MD2 complex, explaining their ability to inhibit TLR4 signaling. CONCLUSIONS: 2'-FL and 6'-SL, but not lactose, prevent NEC in mice and piglet models and attenuate NEC inflammation in the human ileum, in part through TLR4 inhibition. IMPACT: Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants that occurs in the setting of bacterial colonization of the gut and administration of formula feeds and activation by the innate immune receptor toll-like receptor 4 (TLR4). Breast milk prevents NEC through unclear mechanisms. We now show that breast milk-enriched human milk oligosaccharides (HMOs) that are derived from lactose prevent NEC through inhibition of TLR4. The human milk oligosaccharides 2'-FL and 6'-SL, but not the backbone sugar lactose, prevent NEC in mice and piglets. 2'-FL and 6'-SL but not lactose inhibited TLR4 signaling in cultured enterocytes, in enteroids derived from mouse intestine, and in human intestinal explants obtained at the time of surgical resection for patients with NEC. In seeking the mechanisms involved, 2'-FL and 6'-SL but not lactose were found to directly bind to TLR4, explaining the inhibition and protection against NEC. These findings may impact clinical practice by suggesting that administration of HMOs could serve as a preventive strategy for premature infants at risk for NEC development.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Íleon/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Lactosa/análogos & derivados , Leche Humana/química , Receptor Toll-Like 4/antagonistas & inhibidores , Trisacáridos/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Humanos , Íleon/inmunología , Íleon/metabolismo , Íleon/patología , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Lactosa/aislamiento & purificación , Lactosa/farmacología , Ratones , Simulación del Acoplamiento Molecular , Transducción de Señal , Sus scrofa , Receptor Toll-Like 4/metabolismo , Trisacáridos/aislamiento & purificación , Pérdida de Peso/efectos de los fármacos
5.
J Immunol ; 203(11): 3000-3012, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31645418

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) is a potent negative regulator capable of restraining overactivation of the renin-angiotensin system, which contributes to exuberant inflammation after bacterial infection. However, the mechanism through which ACE2 modulates this inflammatory response is not well understood. Accumulating evidence indicates that infectious insults perturb ACE2 activity, allowing for uncontrolled inflammation. In the current study, we demonstrate that pulmonary ACE2 levels are dynamically varied during bacterial lung infection, and the fluctuation is critical in determining the severity of bacterial pneumonia. Specifically, we found that a pre-existing and persistent deficiency of active ACE2 led to excessive neutrophil accumulation in mouse lungs subjected to bacterial infection, resulting in a hyperinflammatory response and lung damage. In contrast, pre-existing and persistent increased ACE2 activity reduces neutrophil infiltration and compromises host defense, leading to overwhelming bacterial infection. Further, we found that the interruption of pulmonary ACE2 restitution in the model of bacterial lung infection delays the recovery process from neutrophilic lung inflammation. We observed the beneficial effects of recombinant ACE2 when administered to bacterially infected mouse lungs following an initial inflammatory response. In seeking to elucidate the mechanisms involved, we discovered that ACE2 inhibits neutrophil infiltration and lung inflammation by limiting IL-17 signaling by reducing the activity of the STAT3 pathway. The results suggest that the alteration of active ACE2 is not only a consequence of bacterial lung infection but also a critical component of host defense through modulation of the innate immune response to bacterial lung infection by regulating neutrophil influx.


Asunto(s)
Inflamación/inmunología , Neutrófilos/inmunología , Peptidil-Dipeptidasa A/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Enzima Convertidora de Angiotensina 2 , Animales , Modelos Animales de Enfermedad , Femenino , Imidazoles/administración & dosificación , Imidazoles/farmacología , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Inflamación/tratamiento farmacológico , Inflamación/patología , Leucina/administración & dosificación , Leucina/análogos & derivados , Leucina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pruebas de Sensibilidad Microbiana , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Peptidil-Dipeptidasa A/deficiencia , Peptidil-Dipeptidasa A/genética , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/efectos de los fármacos
6.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L17-L31, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28935640

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) is a terminal carboxypeptidase with important functions in the renin-angiotensin system and plays a critical role in inflammatory lung diseases. ACE2 cleaves single-terminal residues from several bioactive peptides such as angiotensin II. However, few of its substrates in the respiratory tract have been identified, and the mechanism underlying the role of ACE2 in inflammatory lung disease has not been fully characterized. In an effort to identify biological targets of ACE2 in the lung, we tested its effects on des-Arg9 bradykinin (DABK) in airway epithelial cells on the basis of the hypothesis that DABK is a biological substrate of ACE2 in the lung and ACE2 plays an important role in the pathogenesis of acute lung inflammation partly through modulating DABK/bradykinin receptor B1 (BKB1R) axis signaling. We found that loss of ACE2 function in mouse lung in the setting of endotoxin inhalation led to activation of the DABK/BKB1R axis, release of proinflammatory chemokines such as C-X-C motif chemokine 5 (CXCL5), macrophage inflammatory protein-2 (MIP2), C-X-C motif chemokine 1 (KC), and TNF-α from airway epithelia, increased neutrophil infiltration, and exaggerated lung inflammation and injury. These results indicate that a reduction in pulmonary ACE2 activity contributes to the pathogenesis of lung inflammation, in part because of an impaired ability to inhibit DABK/BKB1R axis-mediated signaling, resulting in more prompt onset of neutrophil infiltration and more severe inflammation in the lung. Our study identifies a biological substrate of ACE2 within the airways, as well as a potential new therapeutic target for inflammatory diseases.


Asunto(s)
Bradiquinina/análogos & derivados , Lipopolisacáridos/toxicidad , Infiltración Neutrófila/inmunología , Peptidil-Dipeptidasa A/fisiología , Neumonía/inmunología , Receptor de Bradiquinina B1/metabolismo , Tráquea/inmunología , Enzima Convertidora de Angiotensina 2 , Animales , Antiinflamatorios , Bradiquinina/farmacología , Células Cultivadas , Quimiocina CXCL5/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/efectos de los fármacos , Neumonía/inducido químicamente , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Tráquea/efectos de los fármacos , Tráquea/patología
7.
Br J Nutr ; 120(6): 665-680, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30176959

RESUMEN

Necrotising enterocolitis (NEC) is a devastating disease that typically affects formula-fed premature infants, suggesting that dietary components may influence disease pathogenesis. TAG are the major fat components of infant formula, and their digestion requires pancreatic lipases, which may be naturally deficient in premature neonates. We hypothesise that NEC develops partly from the accumulation of incompletely digested long-chain TAG-containing unsaturated fatty acids within the intestinal epithelial cells, leading to oxidative stress and enterocyte damage. We further hypothesise that the administration of a formula that contains reduced TAG ('pre-digested fat') that do not require lipase action may reduce NEC severity. To test these hypotheses, we induced NEC in neonatal mice using three different fat formulations, namely 'standard fat', 'pre-digested fat' or 'very low fat', and determined that mice fed 'standard fat' developed severe NEC, which was significantly reduced in mice fed 'pre-digested fat' or 'very low fat'. The expression level of the critical fat-digesting enzyme carboxyl ester lipase was significantly lower in the newborn compared with older pups, leading to impaired fat digestion. The accumulation of mal-digested fat resulted in the significant accumulation of fat droplets within the intestinal epithelium of the distal ileum, resulting in the generation of reactive oxygen species and intestinal inflammation. Strikingly, these changes were prevented in pups fed 'pre-digested fat' or 'very low fat' formulas. These findings suggest that nutritional formula containing a pre-digested fat system may overcome the natural lipase deficiency of the premature gut, and serve as a novel approach to prevent NEC.


Asunto(s)
Dieta , Grasas de la Dieta/farmacología , Digestión , Enterocolitis Necrotizante/metabolismo , Fórmulas Infantiles/química , Mucosa Intestinal/efectos de los fármacos , Triglicéridos/farmacología , Animales , Animales Recién Nacidos , Grasas de la Dieta/metabolismo , Enterocolitis Necrotizante/etiología , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Enterocitos/patología , Ácidos Grasos Insaturados/metabolismo , Alimentos Formulados , Humanos , Íleon/efectos de los fármacos , Íleon/metabolismo , Fenómenos Fisiológicos Nutricionales del Lactante , Recién Nacido , Inflamación/etiología , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Lipasa/metabolismo , Ratones , Estrés Oxidativo , Índice de Severidad de la Enfermedad , Triglicéridos/metabolismo
8.
J Immunol ; 197(3): 859-71, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27307558

RESUMEN

We seek to define the mechanisms leading to the development of lung disease in the setting of neonatal necrotizing enterocolitis (NEC), a life-threatening gastrointestinal disease of premature infants characterized by the sudden onset of intestinal necrosis. NEC development in mice requires activation of the LPS receptor TLR4 on the intestinal epithelium, through its effects on modulating epithelial injury and repair. Although NEC-associated lung injury is more severe than the lung injury that occurs in premature infants without NEC, the mechanisms leading to its development remain unknown. In this study, we now show that TLR4 expression in the lung gradually increases during postnatal development, and that mice and humans with NEC-associated lung inflammation express higher levels of pulmonary TLR4 than do age-matched controls. NEC in wild-type newborn mice resulted in significant pulmonary injury that was prevented by deletion of TLR4 from the pulmonary epithelium, indicating a role for pulmonary TLR4 in lung injury development. Mechanistically, intestinal epithelial TLR4 activation induced high-mobility group box 1 release from the intestine, which activated pulmonary epithelial TLR4, leading to the induction of the neutrophil recruiting CXCL5 and the influx of proinflammatory neutrophils to the lung. Strikingly, the aerosolized administration of a novel carbohydrate TLR4 inhibitor prevented CXCL5 upregulation and blocked NEC-induced lung injury in mice. These findings illustrate the critical role of pulmonary TLR4 in the development of NEC-associated lung injury, and they suggest that inhibition of this innate immune receptor in the neonatal lung may prevent this devastating complication of NEC.


Asunto(s)
Enterocolitis Necrotizante/complicaciones , Lesión Pulmonar/etiología , Mucosa Respiratoria/metabolismo , Receptor Toll-Like 4/biosíntesis , Animales , Animales Recién Nacidos , Quimiocina CXCL5/metabolismo , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Proteína HMGB1/metabolismo , Humanos , Inmunohistoquímica , Recién Nacido , Lesión Pulmonar/inmunología , Lesión Pulmonar/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Mucosa Respiratoria/inmunología
10.
Br J Nutr ; 116(7): 1175-1187, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27609061

RESUMEN

Necrotising enterocolitis (NEC) is a common disease in premature infants characterised by intestinal ischaemia and necrosis. The only effective preventative strategy against NEC is the administration of breast milk, although the protective mechanisms remain unknown. We hypothesise that an abundant human milk oligosaccharide (HMO) in breast milk, 2'-fucosyllactose (2'FL), protects against NEC by enhancing intestinal mucosal blood flow, and we sought to determine the mechanisms underlying this protection. Administration of HMO-2'FL protected against NEC in neonatal wild-type mice, resulted in a decrease in pro-inflammatory markers and preserved the small intestinal mucosal architecture. These protective effects occurred via restoration of intestinal perfusion through up-regulation of the vasodilatory molecule endothelial nitric oxide synthase (eNOS), as administration of HMO-2'FL to eNOS-deficient mice or to mice that received eNOS inhibitors did not protect against NEC, and by 16S analysis HMO-2'FL affected the microbiota of the neonatal mouse gut, although these changes do not seem to be the primary mechanism of protection. Induction of eNOS by HMO-2'FL was also observed in cultured endothelial cells, providing a link between eNOS and HMO in the endothelium. These data demonstrate that HMO-2'FL protects against NEC in part through maintaining mesenteric perfusion via increased eNOS expression, and suggest that the 2'FL found in human milk may be mediating some of the protective benefits of breast milk in the clinical setting against NEC.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Enfermedades del Prematuro/fisiopatología , Leche Humana/química , Circulación Esplácnica/efectos de los fármacos , Trisacáridos/administración & dosificación , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/fisiopatología , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Recién Nacido , Mucosa Intestinal/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/efectos de los fármacos , Óxido Nítrico/análisis , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/fisiología
11.
Proc Natl Acad Sci U S A ; 110(23): 9451-6, 2013 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-23650378

RESUMEN

Necrotizing enterocolitis (NEC) is a devastating disease of premature infants characterized by severe intestinal necrosis and for which breast milk represents the most effective protective strategy. Previous studies have revealed a critical role for the lipopolysaccharide receptor toll-like receptor 4 (TLR4) in NEC development through its induction of mucosal injury, yet the reasons for which intestinal ischemia in NEC occurs in the first place remain unknown. We hypothesize that TLR4 signaling within the endothelium plays an essential role in NEC development by regulating perfusion to the small intestine via the vasodilatory molecule endothelial nitric oxide synthase (eNOS). Using a unique mouse system in which we selectively deleted TLR4 from the endothelium, we now show that endothelial TLR4 activation is required for NEC development and that endothelial TLR4 activation impairs intestinal perfusion without effects on other organs and reduces eNOS expression via activation of myeloid differentiation primary response gene 88. NEC severity was significantly increased in eNOS(-/-) mice and decreased upon administration of the phosphodiesterase inhibitor sildenafil, which augments eNOS function. Strikingly, compared with formula, human and mouse breast milk were enriched in sodium nitrate--a precursor for enteral generation of nitrite and nitric oxide--and repletion of formula with sodium nitrate/nitrite restored intestinal perfusion, reversed the deleterious effects of endothelial TLR4 signaling, and reduced NEC severity. These data identify that endothelial TLR4 critically regulates intestinal perfusion leading to NEC and reveal that the protective properties of breast milk involve enhanced intestinal microcirculatory integrity via augmentation of nitrate-nitrite-NO signaling.


Asunto(s)
Enterocolitis Necrotizante/etiología , Mucosa Intestinal/irrigación sanguínea , Microcirculación/fisiología , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/metabolismo , Fórmulas Infantiles/química , Fórmulas Infantiles/farmacología , Ratones , Ratones Noqueados , Microcirculación/efectos de los fármacos , Microscopía Confocal , Leche Humana/química , Nitratos/análisis , Nitratos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Nitritos/metabolismo , Piperazinas/farmacología , Piperazinas/uso terapéutico , Purinas/farmacología , Purinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Citrato de Sildenafil , Sulfonas/farmacología , Sulfonas/uso terapéutico , Receptor Toll-Like 4/deficiencia
12.
J Biol Chem ; 289(14): 9584-99, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24519940

RESUMEN

The cellular cues that regulate the apoptosis of intestinal stem cells (ISCs) remain incompletely understood, yet may play a role in diseases characterized by ISC loss including necrotizing enterocolitis (NEC). Toll-like receptor-4 (TLR4) was recently found to be expressed on ISCs, where its activation leads to ISC apoptosis through mechanisms that remain incompletely explained. We now hypothesize that TLR4 induces endoplasmic reticulum (ER) stress within ISCs, leading to their apoptosis in NEC pathogenesis, and that high ER stress within the premature intestine predisposes to NEC development. Using transgenic mice and cultured enteroids, we now demonstrate that TLR4 induces ER stress within Lgr5 (leucine-rich repeat-containing G-protein-coupled receptor 5)-positive ISCs, resulting in crypt apoptosis. TLR4 signaling within crypts was required, because crypt ER stress and apoptosis occurred in TLR4(ΔIEC-OVER) mice expressing TLR4 only within intestinal crypts and epithelium, but not TLR4(ΔIEC) mice lacking intestinal TLR4. TLR4-mediated ER stress and apoptosis of ISCs required PERK (protein kinase-related PKR-like ER kinase), CHOP (C/EBP homologous protein), and MyD88 (myeloid differentiation primary response gene 88), but not ATF6 (activating transcription factor 6) or XBP1 (X-box-binding protein 1). Human and mouse NEC showed high crypt ER stress and apoptosis, whereas genetic inhibition of PERK or CHOP attenuated ER stress, crypt apoptosis, and NEC severity. Strikingly, using intragastric delivery into fetal mouse intestine, prevention of ER stress reduced TLR4-mediated ISC apoptosis and mucosal disruption. These findings identify a novel link between TLR4-induced ER stress and ISC apoptosis in NEC pathogenesis and suggest that increased ER stress within the premature bowel predisposes to NEC development.


Asunto(s)
Estrés del Retículo Endoplásmico , Enterocolitis Necrotizante/metabolismo , Mucosa Intestinal/metabolismo , Células Madre/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Transcripción Activador 6/genética , Factor de Transcripción Activador 6/metabolismo , Animales , Apoptosis/genética , Enterocolitis Necrotizante/genética , Enterocolitis Necrotizante/patología , Células HEK293 , Humanos , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Células Madre/patología , Receptor Toll-Like 4/genética , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
13.
J Immunol ; 190(7): 3541-51, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23455503

RESUMEN

Necrotizing enterocolitis (NEC) develops in response to elevated TLR4 signaling in the newborn intestinal epithelium and is characterized by TLR4-mediated inhibition of enterocyte migration and reduced mucosal healing. The downstream processes by which TLR4 impairs mucosal healing remain incompletely understood. In other systems, TLR4 induces autophagy, an adaptive response to cellular stress. We now hypothesize that TLR4 induces autophagy in enterocytes and that TLR4-induced autophagy plays a critical role in NEC development. Using mice selectively lacking TLR4 in enterocytes (TLR4(ΔIEC)) and in TLR4-deficient cultured enterocytes, we now show that TLR4 activation induces autophagy in enterocytes. Immature mouse and human intestine showed increased expression of autophagy genes compared with full-term controls, and NEC development in both mouse and human was associated with increased enterocyte autophagy. Importantly, using mice in which we selectively deleted the autophagy gene ATG7 from the intestinal epithelium (ATG7(ΔIEC)), the induction of autophagy was determined to be required for and not merely a consequence of NEC, because ATG7(ΔIEC) mice were protected from NEC development. In defining the mechanisms involved, TLR4-induced autophagy led to impaired enterocyte migration both in vitro and in vivo, which in cultured enterocytes required the induction of RhoA-mediated stress fibers. These findings depart from current dogma in the field by identifying a unique effect of TLR4-induced autophagy within the intestinal epithelium in the pathogenesis of NEC and identify that the negative consequences of autophagy on enterocyte migration play an essential role in its development.


Asunto(s)
Autofagia , Movimiento Celular , Enterocolitis Necrotizante/etiología , Enterocitos/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Autofagia/genética , Línea Celular , Movimiento Celular/genética , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/patología , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Transgénicos , Receptor Toll-Like 4/genética , Proteínas de Unión al GTP rho/metabolismo
14.
Tetrahedron Lett ; 56(23): 3097-3100, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-26236050

RESUMEN

The low-molecular weight isopropyl 2-acetamido-α-glucoside 16 (C34) inhibits toll-like receptor 4 (TLR4) in enterocytes and macrophages in vitro, and reduces systemic inflammation in mouse models of endotoxemia and necrotizing enterocolitis. We used a copper(II)-mediated solvolysis of anomeric oxazolines and an acid-mediated conversion of ß-glucosamine and ß-galactosamine pentaacetates to generate analogs of 16 at the anomeric carbon and at C-4 of the pyranose ring. These compounds were evaluated for their influence on TLR4-mediated inflammatory signaling in cultured enterocytes and monocytes. Their efficacy was confirmed using a NF-kB-luciferase reporter mouse, thus establishing the first structure-activity relationship (SAR) study in this series and identifying the more efficacious isopropyl 2-acetamido-α-galactoside 17.

15.
Proc Natl Acad Sci U S A ; 109(28): 11330-5, 2012 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-22733781

RESUMEN

The fetal intestinal mucosa is characterized by elevated Toll-like receptor 4 (TLR4) expression, which can lead to the development of necrotizing enterocolitis (NEC)--a devastating inflammatory disease of the premature intestine--upon exposure to microbes. To define endogenous strategies that could reduce TLR4 signaling, we hypothesized that amniotic fluid can inhibit TLR4 signaling within the fetal intestine and attenuate experimental NEC, and we sought to determine the mechanisms involved. We show here that microinjection of amniotic fluid into the fetal (embryonic day 18.5) gastrointestinal tract reduced LPS-mediated signaling within the fetal intestinal mucosa. Amniotic fluid is abundant in EGF, which we show is required for its inhibitory effects on TLR4 signaling via peroxisome proliferator-activated receptor, because inhibition of EGF receptor (EGFR) with cetuximab or EGF-depleted amniotic fluid blocked the inhibitory effects of amniotic fluid on TLR4, whereas amniotic fluid did not prevent TLR4 signaling in EGFR- or peroxisome proliferator-activated receptor γ-deficient enterocytes or in mice deficient in intestinal epithelial EGFR, and purified EGF attenuated the exaggerated intestinal mucosal TLR4 signaling in wild-type mice. Moreover, amniotic fluid-mediated TLR4 inhibition reduced the severity of NEC in mice through EGFR activation. Strikingly, NEC development in both mice and humans was associated with reduced EGFR expression that was restored upon the administration of amniotic fluid in mice or recovery from NEC in humans, suggesting that a lack of amniotic fluid-mediated EGFR signaling could predispose to NEC. These findings may explain the unique susceptibility of premature infants to the development of NEC and offer therapeutic approaches to this devastating disease.


Asunto(s)
Líquido Amniótico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mucosa Intestinal/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular , Enterocolitis Necrotizante/metabolismo , Enterocitos/metabolismo , Receptores ErbB/metabolismo , Humanos , Recién Nacido , Mucosa Intestinal/embriología , Intestinos/embriología , Ratones , Microscopía Confocal/métodos , Transducción de Señal , Factores de Tiempo
16.
J Immunol ; 188(9): 4543-57, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22461698

RESUMEN

Necrotizing enterocolitis (NEC) is the leading cause of gastrointestinal-related mortality in premature infants, and it develops under conditions of exaggerated TLR4 signaling in the newborn intestinal epithelium. Because NEC does not develop spontaneously, despite the presence of seemingly tonic stimulation of intestinal TLR4, we hypothesized that mechanisms must exist to constrain TLR4 signaling that become diminished during NEC pathogenesis and focused on the intracellular stress response protein and chaperone heat shock protein-70 (Hsp70). We demonstrate that the induction of intracellular Hsp70 in enterocytes dramatically reduced TLR4 signaling, as assessed by LPS-induced NF-κB translocation, cytokine expression, and apoptosis. These findings were confirmed in vivo, using mice that either globally lacked Hsp70 or overexpressed Hsp70 within the intestinal epithelium. TLR4 activation itself significantly increased Hsp70 expression in enterocytes, which provided a mechanism of autoinhibition of TLR4 signaling in enterocytes. In seeking to define the mechanisms involved, intracellular Hsp70-mediated inhibition of TLR4 signaling required both its substrate-binding EEVD domain and association with the cochaperone CHIP, resulting in ubiquitination and proteasomal degradation of TLR4. The expression of Hsp70 in the intestinal epithelium was significantly decreased in murine and human NEC compared with healthy controls, suggesting that loss of Hsp70 protection from TLR4 could lead to NEC. In support of this, intestinal Hsp70 overexpression in mice and pharmacologic upregulation of Hsp70 reversed TLR4-induced cytokines and enterocyte apoptosis, as well as prevented and treated experimental NEC. Thus, a novel TLR4 regulatory pathway exists within the newborn gut involving Hsp70 that may be pharmacologically activated to limit NEC severity.


Asunto(s)
Proteínas HSP70 de Choque Térmico/inmunología , Mucosa Intestinal/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Femenino , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Recién Nacido , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Lipopolisacáridos/farmacología , Masculino , Ratones , FN-kappa B/inmunología , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Estructura Terciaria de Proteína , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos , Ubiquitinación/inmunología
17.
Cell Mol Gastroenterol Hepatol ; 18(1): 53-70, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38438014

RESUMEN

BACKGROUND & AIMS: The abdominal discomfort experienced by patients with colitis may be attributable in part to the presence of small intestinal dysmotility, yet mechanisms linking colonic inflammation with small-bowel motility remain largely unexplored. We hypothesize that colitis results in small intestinal hypomotility owing to a loss of enteroendocrine cells (EECs) within the small intestine that can be rescued using serotonergic-modulating agents. METHODS: Male C57BL/6J mice, as well as mice that overexpress (EECOVER) or lack (EECDEL) NeuroD1+ enteroendocrine cells, were exposed to dextran sulfate sodium (DSS) colitis (2.5% or 5% for 7 days) and small intestinal motility was assessed by 70-kilodalton fluorescein isothiocyanate-dextran fluorescence transit. EEC number and differentiation were evaluated by immunohistochemistry, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling staining, and quantitative reverse-transcriptase polymerase chain reaction. Mice were treated with the 5-hydroxytryptamine receptor 4 agonist prucalopride (5 mg/kg orally, daily) to restore serotonin signaling. RESULTS: DSS-induced colitis was associated with a significant small-bowel hypomotility that developed in the absence of significant inflammation in the small intestine and was associated with a significant reduction in EEC density. EEC loss occurred in conjunction with alterations in the expression of key serotonin synthesis and transporter genes, including Tph1, Ddc, and Slc6a4. Importantly, mice overexpressing EECs revealed improved small intestinal motility, whereas mice lacking EECs had worse intestinal motility when exposed to DSS. Finally, treatment of DSS-exposed mice with the 5-hydroxytryptamine receptor 4 agonist prucalopride restored small intestinal motility and attenuated colitis. CONCLUSIONS: Experimental DSS colitis induces significant small-bowel dysmotility in mice owing to enteroendocrine loss that can be reversed by genetic modulation of EEC or administering serotonin analogs, suggesting novel therapeutic approaches for patients with symptomatic colitis.


Asunto(s)
Colitis , Sulfato de Dextran , Células Enteroendocrinas , Motilidad Gastrointestinal , Intestino Delgado , Animales , Células Enteroendocrinas/metabolismo , Ratones , Colitis/patología , Colitis/inducido químicamente , Colitis/complicaciones , Masculino , Motilidad Gastrointestinal/efectos de los fármacos , Intestino Delgado/patología , Intestino Delgado/efectos de los fármacos , Sulfato de Dextran/toxicidad , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Serotonina/metabolismo , Benzofuranos
18.
J Biol Chem ; 287(44): 37296-308, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-22955282

RESUMEN

Factors regulating the proliferation and apoptosis of intestinal stem cells (ISCs) remain incompletely understood. Because ISCs exist among microbial ligands, immune receptors such as toll-like receptor 4 (TLR4) could play a role. We now hypothesize that ISCs express TLR4 and that the activation of TLR4 directly on the intestinal stem cells regulates their ability to proliferate or to undergo apoptosis. Using flow cytometry and fluorescent in situ hybridization for the intestinal stem cell marker Lgr5, we demonstrate that TLR4 is expressed on the Lgr5-positive intestinal stem cells. TLR4 activation reduced proliferation and increased apoptosis in ISCs both in vivo and in ISC organoids, a finding not observed in mice lacking TLR4 in the Lgr5-positive ISCs, confirming the in vivo significance of this effect. To define molecular mechanisms involved, TLR4 inhibited ISC proliferation and increased apoptosis via the p53-up-regulated modulator of apoptosis (PUMA), as TLR4 did not affect crypt proliferation or apoptosis in organoids or mice lacking PUMA. In vivo effects of TLR4 on ISCs required TIR-domain-containing adapter-inducing interferon-ß (TRIF) but were independent of myeloid-differentiation primary response-gene 88 (MYD88) and TNFα. Physiological relevance was suggested, as TLR4 activation in necrotizing enterocolitis led to reduced proliferation and increased apoptosis of the intestinal crypts in a manner that could be reversed by inhibition of PUMA, both globally or restricted to the intestinal epithelium. These findings illustrate that TLR4 is expressed on ISCs where it regulates their proliferation and apoptosis through activation of PUMA and that TLR4 regulation of ISCs contributes to the pathogenesis of necrotizing enterocolitis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Proliferación Celular , Mucosa Intestinal/patología , Células Madre/metabolismo , Receptor Toll-Like 4/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/fisiología , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Técnicas de Inactivación de Genes , Íleon/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Ratas , Receptores Acoplados a Proteínas G/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal , Células Madre/inmunología , Células Madre/fisiología , Receptor Toll-Like 4/genética , Activación Transcripcional , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología
19.
Gastroenterology ; 143(3): 708-718.e5, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22796522

RESUMEN

BACKGROUND & AIMS: Little is known about factors that regulate intestinal epithelial differentiation; microbial recognition receptors such as Toll-like receptor (TLR)4 might be involved. We investigated whether intestinal TLR4 regulates epithelial differentiation and is involved in development of necrotizing enterocolitis (NEC) of the immature intestine. METHODS: Mice with conditional disruption of TLR4 in the intestinal epithelium and TLR4 knockout (TLR4(-/-)) mice were generated by breeding TLR4(loxp/loxp) mice with villin-cre and Ella-cre, respectively. Enterocytes that did not express or overexpressed TLR4 were created by lentiviral or adenoviral transduction. Intestinal organoids were cultured on tissue matrices. Bile acids were measured by colorimetric assays, and microbial composition was determined by 16S pyrosequencing. NEC was induced in 7- to 10-day-old mice by induction of hypoxia twice daily for 4 days. RESULTS: TLR4(-/-) mice and mice with enterocyte-specific deletion of TLR4 were protected from NEC; epithelial differentiation into goblet cells was increased via suppressed Notch signaling in the small intestinal epithelium. TLR4 also regulates differentiation of goblet cells in intestinal organoid and enterocyte cell cultures; differentiation was increased on deletion of TLR4 and restored when TLR4 was expressed ectopically. TLR4 signaling via Notch was increased in intestinal tissue samples from patients with NEC, and numbers of goblet cells were reduced. 16S pyrosequencing revealed that wild-type and TLR4-deficient mice had similar microbial profiles; increased numbers of goblet cells were observed in mice given antibiotics. TLR4 deficiency reduced levels of luminal bile acids in vivo, and addition of bile acids to TLR4-deficient cell cultures prevented differentiation of goblet cells. CONCLUSIONS: TLR4 signaling and Notch are increased in intestinal tissues of patients with NEC and required for induction of NEC in mice. TLR4 prevents goblet cell differentiation, independently of the microbiota. Bile acids might initiate goblet cell development.


Asunto(s)
Diferenciación Celular , Enterocolitis Necrotizante/metabolismo , Células Caliciformes/metabolismo , Intestino Delgado/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Animales Recién Nacidos , Ácidos y Sales Biliares/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/etiología , Enterocolitis Necrotizante/genética , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/prevención & control , Células Caliciformes/microbiología , Células Caliciformes/patología , Humanos , Hipoxia/complicaciones , Fórmulas Infantiles , Recién Nacido , Intestino Delgado/microbiología , Intestino Delgado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organoides , Interferencia de ARN , Ratas , Receptores Notch/metabolismo , Transducción de Señal , Técnicas de Cultivo de Tejidos , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Transfección
20.
J Biol Chem ; 285(7): 4995-5002, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-20007974

RESUMEN

Toll-like receptor-4 (TLR4) is the receptor for bacterial lipopolysaccharide, yet it may also respond to a variety of endogenous molecules. Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in newborn infants and is characterized by intestinal mucosal destruction and impaired enterocyte migration due to increased TLR4 signaling on enterocytes. The endogenous ligands for TLR4 that lead to impaired enterocyte migration remain unknown. High mobility group box-1 (HMGB1) is a DNA-binding protein that is released from injured cells during inflammation. We thus hypothesize that extracellular HMGB1 inhibits enterocyte migration via activation of TLR4 and sought to define the pathways involved. We now demonstrate that murine and human NEC are associated with increased intestinal HMGB1 expression, that serum HMGB1 is increased in murine NEC, and that HMGB1 inhibits enterocyte migration in vitro and in vivo in a TLR4-dependent manner. This finding was unique to enterocytes as HMGB1 enhanced migration of inflammatory cells in vitro and in vivo. In seeking to understand the mechanisms involved, TLR4-dependent HMGB1 signaling increased RhoA activation in enterocytes, increased phosphorylation of focal adhesion kinase, and increased phosphorylation of cofilin, resulting in increased stress fibers and focal adhesions. Using single cell force traction microscopy, the net effect of HMGB1 signaling was a TLR4-dependent increase in cell force adhesion, accounting for the impaired enterocyte migration. These findings demonstrate a novel pathway by which TLR4 activation by HMGB1 delays mucosal repair and suggest a novel potential therapeutic target in the amelioration of intestinal inflammatory diseases like NEC.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Enterocitos/citología , Proteína HMGB1/metabolismo , Proteína HMGB1/farmacología , Mucosa Intestinal/metabolismo , Receptor Toll-Like 4/metabolismo , Actinas/metabolismo , Animales , Línea Celular , Movimiento Celular/genética , Quimiotaxis/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Enterocolitis Necrotizante/metabolismo , Enterocitos/efectos de los fármacos , Citometría de Flujo , Humanos , Técnicas In Vitro , Recién Nacido , Mucosa Intestinal/citología , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratones , Receptor Toll-Like 4/genética , Proteína de Unión al GTP rhoA/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda