Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pediatr Hematol Oncol ; 44(8): 486-488, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35426856

RESUMEN

Ewing sarcoma is an EWS-ETS family member-driven malignancy that most commonly arises from bone. Cutaneous Ewing sarcoma is a rare variant which harbors an EWS-ETS family fusion but demonstrates an immunohistochemical staining pattern distinct from classic Ewing tumors. EWSR1 fluorescence in situ hybridization testing interpretation can be challenging in the setting of cutaneous Ewing sarcoma, making an integrated histologic and sequencing approach key for an accurate diagnosis. Here, we report a pediatric patient with a history of neuroblastoma treated with surgery only that developed a cutaneous nodule and was diagnosed with cutaneous Ewing sarcoma as a second primary cancer.


Asunto(s)
Neoplasias Óseas , Neoplasias Primarias Secundarias , Sarcoma de Ewing , Humanos , Niño , Sarcoma de Ewing/diagnóstico , Sarcoma de Ewing/genética , Sarcoma de Ewing/patología , Neoplasias Primarias Secundarias/diagnóstico , Neoplasias Primarias Secundarias/genética , Hibridación Fluorescente in Situ , Proteínas de Fusión Oncogénica/genética , Proteína EWS de Unión a ARN/genética , Familia , Neoplasias Óseas/diagnóstico , Neoplasias Óseas/genética , Neoplasias Óseas/patología
2.
Pediatr Dev Pathol ; 24(3): 213-226, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33683985

RESUMEN

OBJECTIVES: Benign tumors with skeletal muscle differentiation are rare and their characterization in the literature is limited. We present a series of twelve pediatric benign tumors with rhabdomyomatous differentiation including seven rhabdomyomatous mesenchymal hamartomas, four fetal rhabdomyomas, and one benign triton tumor, analyzing myogenic markers as well as clinicopathologic and molecular features. A review of the literature was also performed with an emphasis on myogenic marker expression and correlation with molecular features. METHODS AND RESULTS: Cases obtained from three tertiary pediatric hospitals were retrospectively reviewed. Eleven of twelve cases expressed myogenin in rare to greater than 15% of cells. Five of nine cases had rare to 70-80% of cells positive for MyoD1. One fetal rhabdomyoma demonstrated homozygous deletions in ZEB2. The benign triton tumor harbored a CTNNB1 mutation. Review of the literature identified 160 pediatric benign tumors with skeletal muscle differentiation of which 9 reported myogenin positivity. CONCLUSIONS: Myogenin and MyoD1 may be variably expressed in benign lesions with skeletal muscle differentiation. Recognition of key morphologic features remains critical to diagnose these lesions and, in rhabdomyoma, to exclude malignancy. Our series expands the knowledge of the relationship between rhabdomyoma and rhabdomyosarcoma (RMS) by identifying a shared molecular alteration in ZEB2.


Asunto(s)
Miogenina/metabolismo , Neoplasias de Tejido Muscular/patología , Biomarcadores de Tumor/metabolismo , Niño , Preescolar , Diagnóstico Diferencial , Femenino , Humanos , Lactante , Masculino
3.
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
4.
Pediatr Blood Cancer ; 65(10): e27283, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29893485

RESUMEN

Post-transplant lymphoproliferative disorder (PTLD) related plasma cell neoplasms are rare in pediatric patients. We report a pediatric liver transplant recipient with plasma cell myeloma type PTLD. Cytogenetics included 1q duplication, associated with poor prognosis in adult multiple myeloma, and t(8;14). High-risk cytogenetics has not been reported in pediatric plasma cell myeloma type PTLD. The patient was treated with bortezomib, dexamethasone, and lenalidomide with subsequent autologous stem cell transplant. He achieved a 6-year remission, demonstrating tolerance to and efficacy of this modern myeloma regimen in a pediatric patient. Unfortunately, he subsequently died from complications of repeat liver transplant.


Asunto(s)
Huésped Inmunocomprometido , Trasplante de Hígado/efectos adversos , Mieloma Múltiple/inmunología , Mieloma Múltiple/terapia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niño , Preescolar , Infecciones por Virus de Epstein-Barr/etiología , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Masculino
5.
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
6.
J Immunol ; 194(10): 4931-9, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25862813

RESUMEN

The mechanisms that lead to the development of remote lung injury after trauma remain unknown, although a central role for the gut in the induction of lung injury has been postulated. We hypothesized that the development of remote lung injury after trauma/hemorrhagic shock requires activation of TLR4 in the intestinal epithelium, and we sought to determine the mechanisms involved. We show that trauma/hemorrhagic shock caused lung injury in wild-type mice, but not in mice that lack TLR4 in the intestinal epithelium, confirming the importance of intestinal TLR4 activation in the process. Activation of intestinal TLR4 after trauma led to increased endoplasmic reticulum (ER) stress, enterocyte apoptosis, and the release of circulating HMGB1, whereas inhibition of ER stress attenuated apoptosis, reduced circulating HMGB1, and decreased lung injury severity. Neutralization of circulating HMGB1 led to reduced severity of lung injury after trauma, and mice that lack HMGB1 in the intestinal epithelium were protected from the development of lung injury, confirming the importance of the intestine as the source of HMGB1, whose release of HMGB1 induced a rapid protein kinase C ζ-mediated internalization of surface tight junctions in the pulmonary epithelium. Strikingly, the use of a novel small-molecule TLR4 inhibitor reduced intestinal ER stress, decreased circulating HMGB1, and preserved lung architecture after trauma. Thus, intestinal epithelial TLR4 activation leads to HMGB1 release from the gut and the development of lung injury, whereas strategies that block upstream TLR4 signaling may offer pulmonary protective strategies after trauma.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Mucosa Intestinal/inmunología , Choque Hemorrágico/complicaciones , Receptor Toll-Like 4/inmunología , Animales , Línea Celular , Modelos Animales de Enfermedad , Proteína HMGB1/biosíntesis , Proteína HMGB1/inmunología , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Toll-Like 4/metabolismo
7.
Am J Pathol ; 185(12): 3304-15, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26476347

RESUMEN

The mechanisms by which drugs induce pancreatitis are unknown. A definite cause of pancreatitis is due to the antiepileptic drug valproic acid (VPA). On the basis of three crucial observations-that VPA inhibits histone deacetylases (HDACs), HDACs mediate pancreas development, and aspects of pancreas development are recapitulated during recovery of the pancreas after injury-we hypothesized that VPA does not cause injury on its own, but it predisposes patients to pancreatitis by inhibiting HDACs and provoking an imbalance in pancreatic recovery. In an experimental model of pancreatic injury, we found that VPA delayed recovery of the pancreas and reduced acinar cell proliferation. In addition, pancreatic expression of class I HDACs (which are the primary VPA targets) increased in the midphase of pancreatic recovery. VPA administration inhibited pancreatic HDAC activity and led to the persistence of acinar-to-ductal metaplastic complexes, with prolonged Sox9 expression and sustained ß-catenin nuclear activation, findings that characterize a delay in regenerative reprogramming. These effects were not observed with valpromide, an analog of VPA that lacks HDAC inhibition. This is the first report, to our knowledge, that VPA shifts the balance toward pancreatic injury and pancreatitis through HDAC inhibition. The work also identifies a new paradigm for therapies that could exploit epigenetic reprogramming to enhance pancreatic recovery and disorders of pancreatic injury.


Asunto(s)
Células Acinares/efectos de los fármacos , Anticonvulsivantes/toxicidad , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/fisiología , Pancreatitis/inducido químicamente , Ácido Valproico/toxicidad , Células Acinares/patología , Animales , Anticonvulsivantes/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ceruletida , Masculino , Ratones , Páncreas/fisiología , Pancreatitis/enzimología , Pancreatitis/patología , Regeneración/efectos de los fármacos , Regulación hacia Arriba , Ácido Valproico/farmacología
8.
J Inherit Metab Dis ; 39(1): 3-16, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26541327

RESUMEN

BACKGROUND: Acute liver failure (ALF) in infancy and childhood is a life-threatening emergency and in about 50% the etiology remains unknown. Recently biallelic mutations in NBAS were identified as a new molecular cause of ALF with onset in infancy, leading to recurrent acute liver failure (RALF). METHODS: The phenotype and medical history of 14 individuals with NBAS deficiency was studied in detail and functional studies were performed on patients' fibroblasts. RESULTS: The phenotypic spectrum of NBAS deficiency ranges from isolated RALF to a multisystemic disease with short stature, skeletal dysplasia, immunological abnormalities, optic atrophy, and normal motor and cognitive development resembling SOPH syndrome. Liver crises are triggered by febrile infections; they become less frequent with age but are not restricted to childhood. Complete recovery is typical, but ALF crises can be fatal. Antipyretic therapy and induction of anabolism including glucose and parenteral lipids effectively ameliorates the course of liver crises. Patients' fibroblasts showed an increased sensitivity to high temperature at protein and functional level and a disturbed tethering of vesicles, pointing at a defect of intracellular transport between the endoplasmic reticulum and Golgi. CONCLUSIONS: Mutations in NBAS cause a complex disease with a wide clinical spectrum ranging from isolated RALF to a multisystemic phenotype. Thermal susceptibility of the syntaxin 18 complex is the basis of fever dependency of ALF episodes. NBAS deficiency is the first disease related to a primary defect of retrograde transport. Identification of NBAS deficiency allows optimized therapy of liver crises and even prevention of further episodes.


Asunto(s)
Fallo Hepático Agudo/genética , Fallo Hepático Agudo/patología , Proteínas de Neoplasias/deficiencia , Adolescente , Adulto , Niño , Preescolar , Femenino , Fibroblastos/patología , Humanos , Lactante , Hígado/patología , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Masculino , Mutación/genética , Fenotipo , Recurrencia , Adulto Joven
9.
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
10.
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
11.
Pattern Recognit ; 51: 453-462, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26858466

RESUMEN

We present a new approach to facilitate the application of the optimal transport metric to pattern recognition on image databases. The method is based on a linearized version of the optimal transport metric, which provides a linear embedding for the images. Hence, it enables shape and appearance modeling using linear geometric analysis techniques in the embedded space. In contrast to previous work, we use Monge's formulation of the optimal transport problem, which allows for reasonably fast computation of the linearized optimal transport embedding for large images. We demonstrate the application of the method to recover and visualize meaningful variations in a supervised-learning setting on several image datasets, including chromatin distribution in the nuclei of cells, galaxy morphologies, facial expressions, and bird species identification. We show that the new approach allows for high-resolution construction of modes of variations and discrimination and can enhance classification accuracy in a variety of image discrimination problems.

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.
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
15.
Am J Physiol Gastrointest Liver Physiol ; 306(11): G1021-32, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24742987

RESUMEN

Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in premature infants and develops partly from an exaggerated intestinal epithelial immune response to indigenous microbes. There has been interest in administering probiotic bacteria to reduce NEC severity, yet concerns exist regarding infection risk. Mechanisms of probiotic activity in NEC are unknown although activation of the microbial DNA receptor Toll-like receptor-9 (TLR9) has been postulated. We now hypothesize that the Gram-positive bacterium Lactobacillus rhamnosus HN001 can attenuate NEC in small and large animal models, that its microbial DNA is sufficient for its protective effects, and that protection requires activation of the Toll-like receptor 9 (TLR9). We now show that oral administration of live or UV-inactivated Lactobacillus rhamnosus HN001 attenuates NEC severity in newborn mice and premature piglets, as manifest by reduced histology score, attenuation of mucosal cytokine response, and improved gross morphology. TLR9 was required for Lactobacillus rhamnosus-mediated protection against NEC in mice, as the selective decrease of TLR9 from the intestinal epithelium reversed its protective effects. Strikingly, DNA of Lactobacillus rhamnosus HN001 reduced the extent of proinflammatory signaling in cultured enterocytes and in samples of resected human ileum ex vivo, suggesting the therapeutic potential of this probiotic in clinical NEC. Taken together, these findings illustrate that Lactobacillus rhamnosus HN001 is an effective probiotic for NEC via activation of the innate immune receptor TLR9 and that Lactobacillus rhamnosus DNA is sufficient for its protective effects, potentially reducing concerns regarding the infectious risk of this novel therapeutic approach.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Lacticaseibacillus rhamnosus/fisiología , Probióticos/farmacología , Receptor Toll-Like 9/metabolismo , Animales , Animales Recién Nacidos , ADN Bacteriano/farmacología , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Ratones , Nacimiento Prematuro , Porcinos , Receptor Toll-Like 9/genética
16.
AJR Am J Roentgenol ; 203(6): W697-705, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25415736

RESUMEN

OBJECTIVE: Although neurofibromas are rare, their initial clinical and imaging presentation can mimic those of vascular anomalies, particularly if the characteristic clinical features of neurofibromatosis are not present. The diagnostic challenges encountered in five cases of histologically proven neurofibromas, initially diagnosed as vascular anomalies, are reviewed and discussed. CONCLUSION: The clinical and imaging differences between neurofibromas and vascular anomalies are detailed with the histopathologic features to better understand why some neurofibromas are diagnosed as vascular anomalies.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Neurofibroma/patología , Neoplasias del Sistema Nervioso Periférico/patología , Neoplasias de los Tejidos Blandos/patología , Malformaciones Vasculares/patología , Adolescente , Adulto , Niño , Preescolar , Diagnóstico Diferencial , Femenino , Humanos , Lactante
17.
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
18.
Pattern Recognit Lett ; 42: 115-121, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24910485

RESUMEN

Methods for extracting quantitative information regarding nuclear morphology from histopathology images have been long used to aid pathologists in determining the degree of differentiation in numerous malignancies. Most methods currently in use, however, employ the naïve Bayes approach to classify a set of nuclear measurements extracted from one patient. Hence, the statistical dependency between the samples (nuclear measurements) is often not directly taken into account. Here we describe a method that makes use of statistical dependency between samples in thyroid tissue to improve patient classification accuracies with respect to standard naïve Bayes approaches. We report results in two sample diagnostic challenges.

19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA