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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Gastrointest Liver Physiol ; 306(6): G515-25, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24458024

RESUMEN

Obesity is a growing epidemic with limited effective treatments. The neurotrophic factor glial cell line-derived neurotrophic factor (GDNF) was recently shown to enhance ß-cell mass and improve glucose control in rodents. Its role in obesity is, however, not well characterized. In this study, we investigated the ability of GDNF to protect against high-fat diet (HFD)-induced obesity. GDNF transgenic (Tg) mice that overexpress GDNF under the control of the glial fibrillary acidic protein promoter and wild-type (WT) littermates were maintained on a HFD or regular rodent diet for 11 wk, and weight gain, energy expenditure, and insulin sensitivity were monitored. Differentiated mouse brown adipocytes and 3T3-L1 white adipocytes were used to study the effects of GDNF in vitro. Tg mice resisted the HFD-induced weight gain, insulin resistance, dyslipidemia, hyperleptinemia, and hepatic steatosis seen in WT mice despite similar food intake and activity levels. They exhibited significantly (P<0.001) higher energy expenditure than WT mice and increased expression in skeletal muscle and brown adipose tissue of peroxisome proliferator activated receptor-α and ß1- and ß3-adrenergic receptor genes, which are associated with increased lipolysis and enhanced lipid ß-oxidation. In vitro, GDNF enhanced ß-adrenergic-mediated cAMP release in brown adipocytes and suppressed lipid accumulation in differentiated 3T3L-1 cells through a p38MAPK signaling pathway. Our studies demonstrate a novel role for GDNF in the regulation of high-fat diet-induced obesity through increased energy expenditure. They show that GDNF and its receptor agonists may be potential targets for the treatment or prevention of obesity.


Asunto(s)
Dieta Alta en Grasa , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Obesidad/prevención & control , Células 3T3-L1 , Animales , Metabolismo Energético , Hígado Graso/prevención & control , Resistencia a la Insulina , Masculino , Ratones , Ratones Transgénicos , Triglicéridos/metabolismo
2.
Am J Physiol Gastrointest Liver Physiol ; 304(9): G793-803, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23471340

RESUMEN

Inflammatory bowel disease (IBD), which includes ulcerative colitis and Crohn's disease, is a chronic inflammatory disease associated with an increased risk for colon cancer. Matrix metalloproteinases (MMPs) are the predominant proteinases expressed in the gut mucosa during active IBD. Our laboratory has previously demonstrated that epithelial-derived MMP9 is absent in normal colonic tissue but is upregulated during IBD. In this study MMP9 transgenic mice (Tg-villin-MMP9) are generated specifically to overexpress MMP9 in intestinal epithelium to examine the role and underlying mechanism by which it modulates the pathogenesis of acute colitis. Dextran sodium sulfate (3% DSS)- and Salmonella typhimurium (S.T.)-induced colitis models were used to study gut inflammation in Tg-villin-MMP9 and wild-type littermates (WT). Colonic tissue was analyzed via Western blot, histology, myeloperoxidase (MPO) assay, and quantitative PCR. Tg-villin-MMP9 mice expressed significantly increased MMP9 mRNA and protein expression at basal level. There was a significant decrease in the goblet cells, but a significant increase in proliferation and apoptosis were observed among Tg-villin-MMP9 mice compared with WT mice. There was also a significant increase in the proinflammatory chemokine Kc among Tg-villin-MMP9 compared with WT mice. Tg-villin-MMP9 exhibited a severe inflammatory response than WT mice in both DSS- and S.T.-induced colitis models as evident by greater weight loss and higher clinical score, histological score, and MPO activity, which correlated with relative levels of Kc mRNA. MMP9 expressed by intestinal epithelial cells mediates inflammation in colitis with simultaneous increase in proinflammatory cytokine Kc.


Asunto(s)
Quimiocina CXCL1/metabolismo , Colitis/metabolismo , Mucosa Intestinal/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colitis/inducido químicamente , Colitis/microbiología , Colitis/patología , Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Células HCT116 , Humanos , Enfermedades Inflamatorias del Intestino/fisiopatología , Ratones , Ratones Transgénicos , Salmonelosis Animal/patología , Salmonelosis Animal/fisiopatología , Salmonella typhimurium
3.
Gastroenterology ; 143(4): 1006-16.e4, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22732731

RESUMEN

BACKGROUND & AIMS: Altered gastrointestinal motility is associated with significant morbidity and health care costs. Toll-like receptors (TLR) regulate intestinal homeostasis. We examined the roles of TLR4 signaling in survival of enteric neurons and gastrointestinal motility. METHODS: We assessed changes in intestinal motility by assessing stool frequency, bead expulsion, and isometric muscle recordings of colonic longitudinal muscle strips from mice that do not express TLR4 (Tlr4(Lps-d) or TLR4(-/-)) or Myd88 (Myd88(-/-)), in wild-type germ-free mice or wild-type mice depleted of the microbiota, and in mice with neural crest-specific deletion of Myd88 (Wnt1Cre(+/-)/Myd88(fl/fl)). We studied the effects of the TLR4 agonist lipopolysaccharide (LPS) on survival of cultured, immortalized fetal enteric neurons and enteric neuronal cells isolated from wild-type and Tlr4(Lps-d) mice at embryonic day 13.5. RESULTS: There was a significant delay in gastrointestinal motility and reduced numbers of nitrergic neurons in TLR4(Lps-d), TLR4(-/-), and Myd88(-/-) mice compared with wild-type mice. A similar phenotype was observed in germ-free mice, mice depleted of intestinal microbiota, and Wnt1Cre(+/-)/Myd88(fl/fl) mice. Incubation of enteric neuronal cells with LPS led to activation of the transcription factor nuclear factor (NF)-κB and increased cell survival. CONCLUSIONS: Interactions between enteric neurons and microbes increases neuron survival and gastrointestinal motility in mice. LPS activation of TLR4 and NF-κB appears to promote survival of enteric neurons. Factors that regulate TLR4 signaling in neurons might be developed to alter gastrointestinal motility.


Asunto(s)
Sistema Nervioso Entérico/metabolismo , Motilidad Gastrointestinal , Metagenoma , Factor 88 de Diferenciación Mieloide/metabolismo , Neuronas Nitrérgicas/metabolismo , Receptor Toll-Like 4/metabolismo , Análisis de Varianza , Animales , Antibacterianos/farmacología , Apoptosis , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Neuronas Colinérgicas/fisiología , Colon/fisiología , Defecación , Ingestión de Alimentos , Endotoxinas/sangre , Sistema Nervioso Entérico/microbiología , Heces/microbiología , Femenino , Motilidad Gastrointestinal/efectos de los fármacos , Lipopolisacáridos/sangre , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Contracción Muscular , Músculo Liso/fisiología , Factor 88 de Diferenciación Mieloide/genética , Neuronas Nitrérgicas/microbiología , Neuronas Nitrérgicas/fisiología , Fenotipo , Transducción de Señal , Receptor Toll-Like 4/genética
4.
J Immunol ; 187(3): 1496-505, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21705622

RESUMEN

Inflammatory bowel disease, mainly Crohn's disease and ulcerative colitis, are characterized by epithelial barrier disruption and altered immune regulation. Colonic Ste20-like proline/alanine-rich kinase (SPAK) plays a role in intestinal inflammation, but its underlying mechanisms need to be defined. Both SPAK-transfected Caco2-BBE cells and villin-SPAK transgenic (TG) FVB/6 mice exhibited loss of intestinal barrier function. Further studies demonstrated that SPAK significantly increased paracellular intestinal permeability to FITC-dextran. In vivo studies using the mouse models of colitis induced by dextran sulfate sodium (DSS) and trinitrobenzene sulfonic acid showed that TG FVB/6 mice were more susceptible to DSS and trinitrobenzene sulfonic acid treatment than wild-type FVB/6 mice, as demonstrated by clinical and histological characteristics and enzymatic activities. Consistent with this notion, we found that SPAK increased intestinal epithelial permeability, which likely facilitated the production of inflammatory cytokines in vitro and in vivo, aggravated bacterial translocation in TG mice under DSS treatment, and consequently established a context favorable for the triggering of intestinal inflammation cascades. In conclusion, overexpression of SPAK inhibits maintenance of intestinal mucosal innate immune homeostasis, which makes regulation of SPAK important to attenuate pathological responses in inflammatory bowel disease.


Asunto(s)
Predisposición Genética a la Enfermedad , Enfermedades Inflamatorias del Intestino/enzimología , Enfermedades Inflamatorias del Intestino/genética , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Inmunidad Adaptativa/genética , Animales , Células CACO-2 , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata/genética , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/enzimología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Ratones , Ratones Transgénicos , Permeabilidad , Proteínas Serina-Treonina Quinasas/fisiología
5.
J Biol Chem ; 286(35): 31003-31013, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21757725

RESUMEN

The oligopeptide transporter PepT1 expressed in inflamed colonic epithelial cells transports small bacterial peptides, such as muramyl dipeptide (MDP) and l-Ala-γ-D-Glu-meso-diaminopimelic acid (Tri-DAP) into cells. The innate immune system uses various proteins to sense pathogen-associated molecular patterns. Nucleotide-binding oligomerization domain (NOD)-like receptors of which there are more than 20 related family members are present in the cytosol and recognize intracellular ligands. NOD proteins mediate NF-κB activation via receptor-interacting serine/threonine-protein kinase 2 (RICK or RIPK). The specific ligands for some NOD-like receptors have been identified. NOD type 1 (NOD1) is activated by peptides that contain a diaminophilic acid, such as the PepT1 substrate Tri-DAP. In other words, PepT1 transport activity plays an important role in controlling intracellular loading of ligands for NOD1 in turn determining the activation level of downstream inflammatory pathways. However, no direct interaction between Tri-DAP and NOD1 has been identified. In the present work, surface plasmon resonance and atomic force microscopy experiments showed direct binding between NOD1 and Tri-DAP with a K(d) value of 34.5 µM. In contrast, no significant binding was evident between muramyl dipeptide and NOD1. Furthermore, leucine-rich region (LRR)-truncated NOD1 did not interact with Tri-DAP, indicating that Tri-DAP interacts with the LRR domain of NOD1. Next, we examined binding between RICK and NOD1 proteins and found that such binding was significant with a K(d) value of 4.13 µM. However, NOD1/RICK binding was of higher affinity (K(d) of 3.26 µM) when NOD1 was prebound to Tri-DAP. Furthermore, RICK phosphorylation activity was increased when NOD was prebound to Tri-DAP. In conclusion, we have shown that Tri-DAP interacts directly with the LRR domain of NOD1 and consequently increases RICK/NOD1 association and RICK phosphorylation activity.


Asunto(s)
Ácido Diaminopimélico/análogos & derivados , Leucina/química , Proteína Adaptadora de Señalización NOD1/química , Oligopéptidos/química , Biofisica/métodos , Células CACO-2 , Ácido Diaminopimélico/química , Ácido Diaminopimélico/metabolismo , Humanos , Inmunidad Innata , Inflamación , Microscopía de Fuerza Atómica/métodos , Nucleótidos/química , Oligopéptidos/metabolismo , Péptidos/química , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Biochim Biophys Acta ; 1813(6): 1137-43, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21296110

RESUMEN

Prohibitin 1 (PHB1), a pleiotropic protein in the cell, has been implicated in the regulation of proliferation, apoptosis, transcription, mitochondrial protein folding, and as a cell-surface receptor. This diverse array of functions of PHB1 is attributed to the cell type studied and its subcellular localization. This review discusses recent data that indicate a diverse role of PHB1 in disease pathogenesis and suggest that targeting PHB1 may be a potential therapeutic option for treatment of diseases including cancer, inflammatory bowel disease, insulin resistance/type 2 diabetes, and obesity. These diseases are associated with increased oxidative stress and mitochondrial dysfunction and therefore, the role of PHB1 in both responses will also be discussed.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Enfermedades Inflamatorias del Intestino/fisiopatología , Neoplasias/fisiopatología , Obesidad/fisiopatología , Proteínas Represoras/fisiología , Diabetes Mellitus Tipo 2/genética , Regulación de la Expresión Génica , Humanos , Enfermedades Inflamatorias del Intestino/genética , Mitocondrias/metabolismo , Modelos Biológicos , Neoplasias/genética , Obesidad/genética , Estrés Oxidativo/fisiología , Prohibitinas , Proteínas Represoras/metabolismo
7.
Am J Physiol Gastrointest Liver Physiol ; 302(11): G1282-91, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22499850

RESUMEN

The transmembrane glycoprotein CD98 is known to be involved in intestinal inflammation. In the present study, we found that CD98 overexpression in intestinal epithelial cells does not normally affect the expression of colonic (epithelial and immune cell) microRNAs (miRNAs), small noncoding RNAs that posttranscriptionally regulate a wide variety of biological processes. However, upon dextran sulfate sodium (DSS) treatment, the expression of several colonic miRNAs, but not miRNAs from other tissues such as liver and spleen, were differentially regulated in mice overexpressing CD98 in epithelial cells compared with wild-type (WT) animals. For example, the level of colonic miRNA 132 was not affected by DSS treatment in WT animals but was upregulated in mice overexpressing CD98 in intestinal epithelial cells. Other colonic miRNAs, including colonic miRNA 23a and 23b, were downregulated in WT animals after DSS treatment but not in colonic epithelial cell CD98-overexpressing mice. Interestingly, the expression of potential miRNA target genes affected intestinal epithelial cells that overexpress CD98 and cell types that did not overexpress CD98 but were in close proximity to CD98-overexpressing intestinal epithelial cells. Taken together, these observations show that the combination of an inflammatory context and intestinal epithelial cell expression of CD98 affects the regulation of miRNA expression in colonic epithelial and immune cells. This is new evidence that protein expression modulates miRNA expression and suggests the existence of regulatory crosstalk between proteins and miRNAs in diseases such as colitis.


Asunto(s)
Colitis/metabolismo , Colon/metabolismo , Proteína-1 Reguladora de Fusión/biosíntesis , Mucosa Intestinal/metabolismo , MicroARNs/metabolismo , Animales , Colitis/genética , Células Epiteliales/metabolismo , Inflamación , Ratones , MicroARNs/genética
8.
Gastroenterology ; 140(2): 540-549.e2, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21078320

RESUMEN

BACKGROUND & AIMS: Krüppel-like factor 5 (KLF5) is a transcription factor that promotes proliferation, is highly expressed in dividing crypt cells of the gastrointestinal epithelium, and is induced by various stress stimuli. We sought to determine the role of KLF5 in colonic inflammation and recovery by studying mice with dextran sulfate sodium (DSS)-induced colitis. METHODS: Wild-type (WT) and Klf5(+/-) mice were given DSS in the drinking water to induce colitis. For recovery experiments, mice were given normal drinking water for 5 days after DSS administration. The extent of colitis was determined using established clinical and histological scoring systems. Immunohistochemical and immunoblotting analyses were used to examine proliferation, migration, and expression of the epidermal growth factor receptor. RESULTS: Klf5 expression was increased in colonic tissues of WT mice given DSS; induction of Klf5 was downstream of mitogen-activated protein kinase signaling. In DSS-induced colitis, Klf5(+/-) mice exhibited greater sensitivity to DSS than WT mice, with significantly higher clinical and histological colitis scores. In recovery experiments, Klf5(+/-) mice showed poor recovery, with continued weight loss and higher mortality than WT mice. Klf5(+/-) mice from the recovery period had reduced epithelial proliferation and cell migration at sites of ulceration compared to WT mice; these reductions correlated with reduced expression of epidermal growth factor receptor. CONCLUSIONS: Epithelial repair is an important aspect of recovery from DSS-induced colitis. The transcription factor KLF5 regulates mucosal healing through its effects on epithelial proliferation and migration.


Asunto(s)
Colitis/fisiopatología , Factores de Transcripción de Tipo Kruppel/fisiología , Regeneración , Animales , Movimiento Celular , Proliferación Celular , Colitis/inducido químicamente , Colitis/patología , Colon/efectos de los fármacos , Sulfato de Dextran/farmacología , Modelos Animales de Enfermedad , Receptores ErbB/análisis , Femenino , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/análisis
9.
Gastroenterology ; 141(4): 1334-45, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21762661

RESUMEN

BACKGROUND & AIMS: The human di/tripeptide transporter human intestinal H-coupled oligonucleotide transporter (hPepT1) is abnormally expressed in colons of patients with inflammatory bowel disease, although its exact role in pathogenesis is unclear. We investigated the contribution of PepT1 to intestinal inflammation in mouse models of colitis and the involvement of the nucleotide-binding oligomerization domain 2 (NOD2) signaling pathway in the pathogenic activity of colonic epithelial hPepT1. METHODS: Transgenic mice were generated in which hPepT1 expression was regulated by the ß-actin or villin promoters; colitis was induced using 2,4,6-trinitrobenzene sulfonic acid (TNBS) or dextran sodium sulfate (DSS) and the inflammatory responses were assessed. The effects of NOD2 deletion in the hPepT1 transgenic mice also was studied to determine the involvement of the PepT1-NOD2 signaling pathway. RESULTS: TNBS and DSS induced more severe levels of inflammation in ß-actin-hPepT1 transgenic mice than wild-type littermates. Intestinal epithelial cell-specific hPepT1 overexpression in villin-hPepT1 transgenic mice increased the severity of inflammation induced by DSS, but not TNBS. Bone marrow transplantation studies showed that hPepT1 expression in intestinal epithelial cells and immune cells has an important role in the proinflammatory response. Antibiotics abolished the effect of hPepT1 overexpression on the inflammatory response in DSS-induced colitis in ß-actin-hPepT1 and villin-hPepT1 transgenic mice, indicating that commensal bacteria are required to aggravate intestinal inflammation. Nod2-/-, ß-actin-hPepT1 transgenic/Nod2-/-, and villin-hPepT1 transgenic/Nod2-/- littermates had similar levels of susceptibility to DSS-induced colitis, indicating that hPepT1 overexpression increased intestinal inflammation in a NOD2-dependent manner. CONCLUSIONS: The PepT1-NOD2 signaling pathway is involved in aggravation of DSS-induced colitis in mice.


Asunto(s)
Colitis/metabolismo , Colon/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Transducción de Señal , Simportadores/metabolismo , Actinas/genética , Animales , Antibacterianos/farmacología , Trasplante de Médula Ósea , Colitis/inducido químicamente , Colitis/genética , Colitis/inmunología , Colitis/microbiología , Colitis/patología , Colitis/prevención & control , Colon/efectos de los fármacos , Colon/inmunología , Colon/microbiología , Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteína Adaptadora de Señalización NOD2/deficiencia , Proteína Adaptadora de Señalización NOD2/genética , Transportador de Péptidos 1 , Regiones Promotoras Genéticas , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Simportadores/genética , Factores de Tiempo , Ácido Trinitrobencenosulfónico
10.
Gastroenterology ; 141(4): 1381-92, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21723221

RESUMEN

BACKGROUND & AIMS: Inflammatory bowel disease increases the risks of colon cancer and colitis-associated cancer (CAC). Epithelial cell-derived matrix metalloproteinase (MMP)-9 mediates inflammation during acute colitis and the cleavage and activation of the transcription factor Notch1, which prevents differentiation of progenitor cells into goblet cells. However, MMP-9 also protects against the development of CAC and acts as a tumor suppressor. We investigated the mechanisms by which MMP-9 protects against CAC in mice. METHODS: C57/B6 wild-type mice were given a single dose of azoxymethane and 2 cycles of dextran sulfate sodium (DSS). Mice were also given the γ-secretase inhibitor difluorophenacetyl-l-alanyl-S-phenylglycine t-butyl ester (DAPT) or dimethyl sulfoxide (control) during each DSS cycle; they were killed on day 56. We analyzed embryonic fibroblasts isolated from wild-type and MMP-9-/- mice and HCT116 cells that were stably transfected with MMP-9. RESULTS: Wild-type mice were more susceptible to CAC following inhibition of Notch1 by DAPT, shown by increased numbers of tumors and level of dysplasia compared with controls. Inhibition of Notch1 signaling significantly reduced protein levels of active Notch1, p53, p21WAF1/Cip1, Bax-1, active caspase-3, as well as apoptosis, compared with controls. Similar results were observed in transgenic HCT116 cells and embryonic fibroblasts from MMP-9-/- mice on γ-radiation-induced damage of DNA. CONCLUSIONS: MMP-9 mediates Notch1 signaling via p53 to regulate apoptosis, cell cycle arrest, and inflammation. By these mechanisms, it might prevent CAC.


Asunto(s)
Colitis/enzimología , Colon/enzimología , Neoplasias del Colon/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Receptor Notch1/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Apoptosis , Azoximetano , Caspasa 3/metabolismo , Colitis/inducido químicamente , Colitis/patología , Colon/efectos de los fármacos , Colon/inmunología , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citocinas/genética , Citocinas/metabolismo , Daño del ADN , Sulfato de Dextran , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Fibroblastos/enzimología , Fibroblastos/efectos de la radiación , Rayos gamma , Células HCT116 , Humanos , Metaloproteinasa 9 de la Matriz/deficiencia , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Receptor Notch1/antagonistas & inhibidores , Transducción de Señal , Factores de Tiempo , Transfección , Proteína p53 Supresora de Tumor/metabolismo
11.
Dig Dis Sci ; 57(6): 1647-51, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22113428

RESUMEN

BACKGROUND: While ulcerative colitis (UC) is a risk factor for colorectal cancer, the association of UC with survival after colorectal cancer has not been studied in an older population. AIMS: The objective of our study was to compare the survival of colorectal cancer between persons with and without UC. METHODS: All cases of colorectal cancer (CRC) in persons 67 and older residing in a SEER catchment area and enrolled in the Medicare between 1993 and 1999 were assessed. We identified diagnosis of UC using ICD-9 codes on Medicare outpatient, office, and inpatient claims in the 2 years prior to the date of diagnosis. We used Cox proportional hazards model and Kaplan-Meier curves to compare survival between individuals with UC and CRC (UC-CRC) and sporadic CRC RESULTS: We identified 47,543 cases of colorectal cancer. Cases with UC-CRC tend to be diagnosed at earlier stages compared to sporadic CRC (42 vs. 37% local (TNM stage 1 and 2) and 11 vs. 17% distant spread (TNM stage 4), respectively; P value = 0.04). Controlling for age, gender, race and stage, diagnosis of UC did not affect the 3-year survival for CRC. CONCLUSIONS: Colorectal cancers tend to be diagnosed at earlier stages among persons with UC, but there is no difference in 3-year survival rates for colorectal cancer among individuals with and without UC.


Asunto(s)
Causas de Muerte/tendencias , Colitis Ulcerosa/diagnóstico , Colitis Ulcerosa/mortalidad , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/mortalidad , Factores de Edad , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Colitis Ulcerosa/complicaciones , Neoplasias Colorrectales/complicaciones , Supervivencia sin Enfermedad , Femenino , Humanos , Clasificación Internacional de Enfermedades , Estimación de Kaplan-Meier , Masculino , Medicare , Estadificación de Neoplasias , Pronóstico , Modelos de Riesgos Proporcionales , Valores de Referencia , Medición de Riesgo , Programa de VERF , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Estados Unidos
12.
J Biol Chem ; 285(24): 18184-90, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20388705

RESUMEN

We have reported that epithelial adenosine 2B receptor (A(2B)AR) mRNA and protein are up-regulated in colitis, which we demonstrated to be regulated by tumor necrosis factor alpha (TNF-alpha). Here, we examined the mechanism that governs A(2B)AR expression during colitis. A 1.4-kb sequence of the A(2B)AR promoter was cloned into the pFRL7 luciferase vector. Anti-microRNA (miRNA) was custom-synthesized based on specific miRNA binding sites. The binding of miRNA to the 3'-untranslated region (UTR) of A(2B)AR mRNA was examined by cloning this 3'-UTR downstream of the luciferase gene in pMIR-REPORT. In T84 cells, TNF-alpha induced a 35-fold increase in A(2B)AR mRNA but did not increase promoter activity in luciferase assays. By nuclear run-on assay, no increase in A(2B)AR mRNA following TNF-alpha treatment was observed. Four putative miRNA target sites (miR27a, miR27b, miR128a, miR128b) in the 3'-UTR of the A(2B)AR mRNA were identified in T84 cells and mouse colon. Pretreatment of cells with TNF-alpha reduced the levels of miR27b and miR128a by 60%. Over expression of pre-miR27b and pre-miR128a reduced A(2B)AR levels by >60%. Blockade of miR27b increased A(2B)AR mRNA levels by 6-fold in vitro. miR27b levels declined significantly in colitis-affected tissue in mice in the presence of increased A(2B)AR mRNA. Collectively, these data demonstrate that TNF-alpha-induced A(2B)AR expression in colonic epithelial cells is post-transcriptionally regulated by miR27b and miR128a and show that miR27b influences A(2B)AR expression in murine colitis.


Asunto(s)
MicroARNs/metabolismo , Receptor de Adenosina A2B/biosíntesis , Transcripción Genética , Regiones no Traducidas 3' , Animales , Núcleo Celular/metabolismo , Colitis/metabolismo , AMP Cíclico/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Procesamiento Postranscripcional del ARN , Factor de Necrosis Tumoral alfa/metabolismo
13.
J Biol Chem ; 285(2): 1479-89, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19892711

RESUMEN

The transmembrane glycoprotein CD98 regulates multiple cellular functions, including extracellular signaling, epithelial cell adhesion/polarity, amino acid transport, and cell-cell interactions. MicroRNAs post-transcriptionally regulate gene expression, thereby functioning as modulators of numerous cellular processes, such as cell differentiation, proliferation, and apoptosis. Here, we investigated if microRNAs regulate CD98 expression during intestinal epithelial cell differentiation and inflammation. We found that microRNA-7 repressed CD98 expression in Caco2-BBE cells by directly targeting the 3'-untranslated region of human CD98 mRNA. Expression of CD98 was decreased, whereas that of microRNA-7 was increased in well-differentiated Caco2-BBE cells compared with undifferentiated cells. Undifferentiated crypt cells isolated from mouse jejunum showed higher CD98 levels and lower levels of mmu-microRNA-706, a murine original microRNA candidate for CD98, than well-differentiated villus cells. Importantly, microRNA-7 decreased Caco2-BBE cell attachment on laminin-1, and CD98 overexpression recovered this inhibition, suggesting that microRNA-7 modulates epithelial cell adhesion to extracellular matrix, which in turn could affect proliferation and differentiation during the migration of enterocytes across the crypt-villus axis, by regulating CD98 expression. In a pathological context, the pro-inflammatory cytokine interleukin 1-beta increased CD98 expression in Caco2-BBE cells by decreasing microRNA-7 levels. Consistent with the in vitro findings, microRNA-7 levels were decreased in actively inflamed Crohn disease colonic tissues, where CD98 expression was up-regulated, compared with normal tissues. Together, these results reveal a novel mechanism underlying regulation of CD98 expression during patho-physiological states. This study raises microRNAs as a promising target for therapeutic modulations of CD98 expression in intestinal inflammatory disorders.


Asunto(s)
Diferenciación Celular , Proteína-1 Reguladora de Fusión/biosíntesis , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , MicroARNs/metabolismo , Regiones no Traducidas 3'/genética , Animales , Células CACO-2 , Adhesión Celular/genética , Comunicación Celular/genética , Polaridad Celular/genética , Enfermedad de Crohn/genética , Enfermedad de Crohn/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Proteína-1 Reguladora de Fusión/genética , Humanos , Inflamación/genética , Inflamación/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Yeyuno/metabolismo , Laminina/genética , Laminina/metabolismo , Masculino , Ratones , MicroARNs/genética
14.
Am J Physiol Gastrointest Liver Physiol ; 300(3): G371-83, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21148398

RESUMEN

Recent advances in nanotechnology offer new hope for disease detection, prevention, and treatment. Nanomedicine is a rapidly evolving field wherein targeted therapeutic approaches using nanotechnology based on the pathophysiology of gastrointestinal diseases are being developed. Nanoparticle vectors capable of delivering drugs specifically and exclusively to regions of the gastrointestinal tract affected by disease for a prolonged period of time are likely to significantly reduce the side effects of existing otherwise effective treatments. This review aims at integrating various applications of the most recently developed nanomaterials that have tremendous potential for the detection and treatment of gastrointestinal diseases.


Asunto(s)
Gastroenterología/métodos , Enfermedades Gastrointestinales , Nanomedicina , Nanoestructuras , Diagnóstico por Imagen/métodos , Portadores de Fármacos , Fármacos Gastrointestinales/administración & dosificación , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/terapia , Técnicas de Transferencia de Gen , Terapia Genética , Humanos , Nanoestructuras/administración & dosificación
15.
Artículo en Inglés | MEDLINE | ID: mdl-21030610

RESUMEN

MicroRNAs (miRNAs), which are noncoding RNAs that posttranscriptionally inhibit expression of target genes, have recently emerged as important regulators of many cellular functions such as cell differentiation. The epithelial di/tripeptide membrane transporter PepT1 is expressed in highly differentiated cells (the villous tip) but not in undifferentiated cells (the crypt) of the small intestine. Here, we investigated the regulation of PepT1 expression by miRNAs and its functional consequences. We observed a reverse correlation between the expression levels of PepT1 and mature miRNA-92b (miR-92b) during the differentiation of intestinal epithelial Caco2-BBE cells, suggesting a miR-92b-mediated regulation of PepT1 expression. We demonstrate that miR-92b suppressed PepT1 expression at both mRNA and protein levels, with subsequent reduced PepT1 transport activity, in Caco2-BBE cells by directly targeting the PepT1 3'-untranslated region. In addition, miR-92b suppresses bacterial peptide-induced proinflammatory responses in intestinal epithelial cells by inhibiting PepT1 expression. Altogether, our study provides for the first time evidence for the regulation of PepT1 expression at a posttranscriptional level by miRNAs in intestinal epithelial cells during pathophysiological states.


Asunto(s)
Mucosa Intestinal/metabolismo , MicroARNs/fisiología , Simportadores/biosíntesis , Células CACO-2 , Diferenciación Celular , Ácido Diaminopimélico/análogos & derivados , Ácido Diaminopimélico/farmacología , Regulación hacia Abajo , Humanos , Inflamación/inducido químicamente , Inflamación/prevención & control , Mucosa Intestinal/citología , Oligopéptidos/farmacología , Transportador de Péptidos 1
16.
Gastroenterology ; 138(3): 843-53.e1-2, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19909746

RESUMEN

BACKGROUND & AIMS: One of the challenges to treating inflammatory bowel disease (IBD) is to target the site of inflammation. We engineered nanoparticles (NPs) to deliver an anti-inflammatory tripeptide Lys-Pro-Val (KPV) to the colon and assessed its therapeutic efficacy in a mouse model of colitis. METHODS: NPs were synthesized by double-emulsion/solvent evaporation. KPV was loaded into the NPs during the first emulsion of the synthesis process. To target KPV to the colon, loaded NPs (NP-KPV) were encapsulated into a polysaccharide gel containing 2 polymers: alginate and chitosan. The effect of KPV-loaded NPs on inflammatory parameters was determined in vitro as well as in the dextran sodium sulfate-induced colitis mouse model. RESULTS: NPs (400 nm) did not affect cell viability or barrier functions. A swelling degree study showed that alginate-chitosan hydrogel containing dextran-fluorescein isothiocyanate-labeled NPs collapsed in the colon. Once delivered, NPs quickly released KPV on or within the closed area of colonocytes. The inflammatory responses to lipopolysaccharide were reduced in Caco2-BBE (brush border enterocyte) cells exposed to NP-KPV compared with those exposed to NPs alone, in a dose-dependent fashion. Mice given dextran sodium sulfate (DSS) followed by NP-KPV were protected against inflammatory and histologic parameters, compared with mice given only DSS. CONCLUSIONS: Nanoparticles are a versatile drug delivery system that can overcome physiologic barriers and target anti-inflammatory agents such as the peptide KPV to inflamed areas. By using NPs, KPV can be delivered at a concentration that is 12,000-fold lower than that of KPV in free solution, but with similar therapeutic efficacy. Administration of encapsulated drug-loaded NPs is a novel therapeutic approach for IBD.


Asunto(s)
Antiinflamatorios/administración & dosificación , Colitis/prevención & control , Portadores de Fármacos , Fármacos Gastrointestinales/administración & dosificación , Hidrogeles , Hormonas Estimuladoras de los Melanocitos/administración & dosificación , Nanopartículas , Fragmentos de Péptidos/administración & dosificación , Polisacáridos/química , Administración Oral , Alginatos/química , Animales , Antiinflamatorios/química , Antiinflamatorios/metabolismo , Células CACO-2 , Química Farmacéutica , Quitosano/química , Colitis/inducido químicamente , Colitis/patología , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Composición de Medicamentos , Femenino , Fármacos Gastrointestinales/química , Fármacos Gastrointestinales/metabolismo , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Hormonas Estimuladoras de los Melanocitos/química , Hormonas Estimuladoras de los Melanocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Solubilidad , Factores de Tiempo
17.
Nat Mater ; 9(11): 923-8, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20935658

RESUMEN

Small interfering RNAs (siRNAs) directed against proinflammatory cytokines have the potential to treat numerous diseases associated with intestinal inflammation; however, the side-effects caused by the systemic depletion of cytokines demands that the delivery of cytokine-targeted siRNAs be localized to diseased intestinal tissues. Although various delivery vehicles have been developed to orally deliver therapeutics to intestinal tissue, none of these strategies has demonstrated the ability to protect siRNA from the harsh environment of the gastrointestinal tract and target its delivery to inflamed intestinal tissue. Here, we present a delivery vehicle for siRNA, termed thioketal nanoparticles (TKNs), that can localize orally delivered siRNA to sites of intestinal inflammation, and thus inhibit gene expression in inflamed intestinal tissue. TKNs are formulated from a polymer, poly-(1,4-phenyleneacetone dimethylene thioketal), that degrades selectively in response to reactive oxygen species (ROS). Therefore, when delivered orally, TKNs release siRNA in response to the abnormally high levels of ROS specific to sites of intestinal inflammation. Using a murine model of ulcerative colitis, we demonstrate that orally administered TKNs loaded with siRNA against the proinflammatory cytokine tumour necrosis factor-alpha (TNF-α) diminish TNF-α messenger RNA levels in the colon and protect mice from ulcerative colitis.


Asunto(s)
Colitis Ulcerosa/prevención & control , Colon/metabolismo , Nanopartículas/administración & dosificación , Nanopartículas/química , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética , Administración Oral , Animales , Colitis Ulcerosa/genética , Colitis Ulcerosa/terapia , Sistemas de Liberación de Medicamentos , Femenino , Ratones , Ratones Endogámicos C57BL , Nanotecnología , Polímeros/química , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo
18.
Dig Dis Sci ; 56(8): 2378-83, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21409378

RESUMEN

BACKGROUND: While ulcerative colitis (UC) and Crohn's disease (CD) are thought to predispose to colorectal cancer (CRC), the association has not been well studied in an older population. AIMS: The objective of our study was to evaluate the association of ulcerative colitis and Crohn's disease and colorectal cancer in a population-based, case-control study. We also wished to estimate the incidence rates of colorectal cancer among older individuals with UC/CD. METHODS: All cases of colorectal cancer in persons 67 and older in the SEER catchment area and in the Medicare claims database were compared with cancer-free controls residing in the same geographic area. We used multivariable logistic regression models adjusted for demographic and other factors. RESULTS: We identified 47,543 cases of CRC and 142,273 controls. We found a modest association between UC and CRC (OR 1.93; 95% CI 1.54-2.49; P-value<0.001) and a significant, albeit modest, association between CD and CRC (OR 1.45; 95% CI 1.08-1.91; P-value 0.01). We found the incidence of CRC to be 8.2 per 10,000 person-years (95% CI 6.5-10.1/10,000 person-years) among those with UC/CD, and 6.1 per 10,000 person-years (95% CI 4.6-7.8/10,000 person-years) among those without UC/CD, resulting in an incidence rate ratio of 1.34. CONCLUSIONS: Among older persons ulcerative colitis and Crohn's disease are modest risk factors for CRC, and the incidence rate ratio for CRC is modest, suggesting that risk of CRC in patients with IBD may be lower than previously thought.


Asunto(s)
Neoplasias Colorrectales/epidemiología , Enfermedades Inflamatorias del Intestino/epidemiología , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Neoplasias Colorrectales/etiología , Femenino , Humanos , Incidencia , Enfermedades Inflamatorias del Intestino/complicaciones , Masculino , Prevalencia , Factores de Riesgo , Estados Unidos/epidemiología
19.
Differentiation ; 80(2-3): 147-54, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20638171

RESUMEN

MicroRNAs (miRNAs) are small, non-coding RNA molecules that post-transcriptionally regulate gene expression. Evidence has shown that miRNAs play important roles in various cellular processes, including proliferation, differentiation and survival. The intestinal epithelium is regenerated throughout life, and enterocytes undergo differentiation during migration along the crypt/villus axis. Our study aimed at establishing the expression profiles of miRNAs during intestinal epithelial cell (IEC) differentiation and determining a miRNA "signature" that distinguishes between small and large IECs. MiRNA arrays were employed to profile miRNA expression in two IEC models: the enterocyte-like Caco2-BBE and the colonocyte-like HT29-Cl.19A cell lines. Microarray data showed that in both cell lineages, the differentiated stage exhibited a different miRNA expression profile from undifferentiated stage. Interestingly, Caco2-BBE cells were distinguished from HT29-Cl.19A cells by their unique miRNA expression profile. Notably, HT29-Cl.19A cells exhibited down-regulation of miR-1269 and up-regulation of miR-99b and miR-125a-5p compared with Caco2-BBE cells. Most importantly, transfection of Caco2-BBE cells with mature miR-99b, mature miR-125a-5p and antisense of mature miR-1269 decreased growth rate and trans-epithelial resistance of the cells, indicating their shift toward HT29-Cl.19A cell phenotype. In conclusion, our study shows that miRNAs might play a role in determining the unique physiological characteristics of IECs.


Asunto(s)
Linaje de la Célula/fisiología , Intestinos/citología , MicroARNs/fisiología , Diferenciación Celular , Línea Celular , Humanos , Mucosa Intestinal/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
J Clin Invest ; 117(12): 3909-21, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18008007

RESUMEN

Activation of TLRs by bacterial products results in rapid activation of genes encoding products designed to protect the host from perturbing microbes. In the intestine, which is colonized by a large and diverse population of commensal bacteria, TLR signaling may not function in a simple on/off mode. Here, we show that the flagellin receptor TLR5 has an essential and nonredundant role in protecting the gut from enteric microbes. Mice lacking TLR5 (TLR5KO mice) developed spontaneous colitis, as assessed by well-defined clinical, serologic, and histopathologic indicators of this disorder. Compared with WT littermates, TLR5KO mice that had not yet developed robust colitis exhibited decreased intestinal expression of TLR5-regulated host defense genes despite having an increased bacterial burden in the colon. In contrast, such TLR5KO mice displayed markedly increased colonic expression of hematopoietic-derived proinflammatory cytokines, suggesting that elevated levels of bacterial products may result in activation of other TLRs that drive colitis in TLR5KO mice. In accordance, deletion of TLR4 rescued the colitis of TLR5KO mice in that mice lacking both TLR4 and TLR5 also had elevated bacterial loads in the colon but lacked immunological, histopathological, and clinical evidence of colitis. That an engineered innate immune deficiency ultimately results in spontaneous intestinal inflammation supports the notion that an innate immune deficiency might underlie some instances of inflammatory bowel disease.


Asunto(s)
Colitis/genética , Eliminación de Gen , Inmunidad Innata/genética , Enfermedades Inflamatorias del Intestino/genética , Receptor Toll-Like 5/genética , Animales , Bacterias/inmunología , Colitis/inmunología , Colitis/microbiología , Colitis/patología , Colon/inmunología , Colon/patología , Citocinas/genética , Citocinas/inmunología , Flagelina/inmunología , Mediadores de Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Ratones , Ratones Noqueados , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 5/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA