Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Pathol ; 194(9): 1664-1683, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38885924

RESUMEN

Bifidobacterium bifidum strain BB1 causes a strain-specific enhancement in intestinal epithelial tight junction (TJ) barrier. Tumor necrosis factor (TNF)-α induces an increase in intestinal epithelial TJ permeability and promotes intestinal inflammation. The major purpose of this study was to delineate the protective effect of BB1 against the TNF-α-induced increase in intestinal TJ permeability and to unravel the intracellular mechanisms involved. TNF-α produces an increase in intestinal epithelial TJ permeability in Caco-2 monolayers and in mice. Herein, the addition of BB1 inhibited the TNF-α increase in Caco-2 intestinal TJ permeability and mouse intestinal permeability in a strain-specific manner. BB1 inhibited the TNF-α-induced increase in intestinal TJ permeability by interfering with TNF-α-induced enterocyte NF-κB p50/p65 and myosin light chain kinase (MLCK) gene activation. The BB1 protective effect against the TNF-α-induced increase in intestinal permeability was mediated by toll-like receptor-2/toll-like receptor-6 heterodimer complex activation of peroxisome proliferator-activated receptor γ (PPAR-γ) and PPAR-γ pathway inhibition of TNF-α-induced inhibitory kappa B kinase α (IKK-α) activation, which, in turn, resulted in a step-wise inhibition of NF-κB p50/p65, MLCK gene, MLCK kinase activity, and MLCK-induced opening of the TJ barrier. In conclusion, these studies unraveled novel intracellular mechanisms of BB1 protection against the TNF-α-induced increase in intestinal TJ permeability. The current data show that BB1 protects against the TNF-α-induced increase in intestinal epithelial TJ permeability via a PPAR-γ-dependent inhibition of NF-κB p50/p65 and MLCK gene activation.


Asunto(s)
Bifidobacterium bifidum , Mucosa Intestinal , Quinasa de Cadena Ligera de Miosina , PPAR gamma , Permeabilidad , Uniones Estrechas , Receptor Toll-Like 2 , Factor de Transcripción ReIA , Factor de Necrosis Tumoral alfa , Animales , Humanos , Ratones , Bifidobacterium bifidum/metabolismo , Bifidobacterium bifidum/fisiología , Células CACO-2 , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ratones Endogámicos C57BL , Quinasa de Cadena Ligera de Miosina/metabolismo , Permeabilidad/efectos de los fármacos , PPAR gamma/metabolismo , Probióticos/farmacología , Uniones Estrechas/metabolismo , Receptor Toll-Like 2/metabolismo , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Receptor Toll-Like 6
2.
Am J Pathol ; 191(5): 872-884, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33607043

RESUMEN

Defective intestinal tight junction (TJ) barrier is an important pathogenic factor of inflammatory bowel disease. To date, no effective therapies that specifically target the intestinal TJ barrier are available. The purpose of this study was to identify probiotic bacterial species or strains that induce a rapid and sustained enhancement of intestinal TJ barrier and protect against the development of intestinal inflammation by targeting the TJ barrier. After high-throughput screening of >20 Lactobacillus and other probiotic bacterial species or strains, a specific strain of Lactobacillus acidophilus, referred to as LA1, uniquely produced a marked enhancement of the intestinal TJ barrier. LA1 attached to the apical membrane surface of intestinal epithelial cells in a Toll-like receptor (TLR)-2-dependent manner and caused a rapid increase in enterocyte TLR-2 membrane expression and TLR-2/TLR-1 and TLR-2/TLR-6 hetero-complex-dependent enhancement in intestinal TJ barrier function. Oral administration of LA1 caused a rapid enhancement in mouse intestinal TJ barrier, protected against a dextran sodium sulfate (DSS) increase in intestinal permeability, and prevented the DSS-induced colitis in a TLR-2- and intestinal TJ barrier-dependent manner. In conclusion, we report for the first time that a specific strain of LA causes a strain-specific enhancement of intestinal TJ barrier through a novel mechanism that involves the TLR-2 receptor complex and protects against the DSS-induced colitis by targeting the intestinal TJ barrier.


Asunto(s)
Colitis/prevención & control , Inflamación/prevención & control , Lactobacillus acidophilus/fisiología , Probióticos , Receptor Toll-Like 2/metabolismo , Animales , Colitis/inducido químicamente , Colitis/microbiología , Colitis/patología , Sulfato de Dextran/efectos adversos , Células Epiteliales/patología , Intestinos/efectos de los fármacos , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Permeabilidad/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/patología , Receptor Toll-Like 2/genética
3.
Gastroenterology ; 159(4): 1375-1389, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32569770

RESUMEN

BACKGROUND & AIMS: Defects in the epithelial tight junction (TJ) barrier contribute to development of intestinal inflammation associated with diseases. Interleukin 1 beta (IL1B) increases intestinal permeability in mice. We investigated microRNAs that are regulated by IL1B and their effects on expression of TJ proteins and intestinal permeability. METHODS: We used Targetscan to identify microRNAs that would bind the 3' untranslated region (3'UTR) of occludin mRNA; regions that interacted with microRNAs were predicted using the V-fold server and Assemble2, and 3-dimensional models were created using UCSF Chimera linked with Assemble2. Caco-2 cells were transfected with vectors that express microRNAs, analyzed by immunoblots and real-time polymerase chain reaction (PCR), and grown as monolayers; permeability in response to IL1B was assessed with the marker inulin. Male C57BL/6 mice were given intraperitoneal injections of IL1B and intestinal recycling perfusion was measured; some mice were given dextran sodium sulfate to induce colitis and/or gavage with an antagonist to MIR200C-3p (antagomiR-200C) or the nonspecific antagomiR (control). Intestinal tissues were collected from mice and analyzed by histology and real-time PCR; enterocytes were isolated by laser capture microdissection. We also analyzed colon tissues and organoids from patients with and without ulcerative colitis. RESULTS: Incubation of Caco-2 monolayers with IL1B increased TJ permeability and reduced levels of occludin protein and mRNA without affecting the expression of other transmembrane TJ proteins. Targetscan identified MIR122, MIR200B-3p, and MIR200C-3p, as miRNAs that might bind to the occludin 3'UTR. MIR200C-3p was rapidly increased in Caco-2 cells incubated with IL1B; the antagomiR-200c prevented the IL1B-induced decrease in occludin mRNA and protein and reduced TJ permeability. Administration of IL1B to mice increased small intestinal TJ permeability, compared with mice given vehicle; enterocytes isolated from mice given IL1B had increased expression of MIR200C-3p and decreased levels of occludin messenger RNA (mRNA) and protein. Intestinal tissues from mice with colitis had increased levels of IL1B mRNA and MIR200C-3p and decreased levels of occludin mRNA; gavage of mice with antagomiR-200C reduced levels of MIR200C-3p and prevented the decrease in occludin mRNA and the increase in colonic permeability. Colon tissues and organoids from patients with ulcerative colitis had increased levels of IL1B mRNA and MIR200C-3p compared with healthy controls. Using 3-dimensional molecular modeling and mutational analyses, we identified the nucleotide bases in the occluding mRNA 3'UTR that interact with MIR200C-3p. CONCLUSIONS: Intestine tissues from patients with ulcerative colitis and mice with colitis have increased levels of IL1B mRNA and MIR200C-3p, which reduces expression of occludin by enterocytes and thereby increases TJ permeability. Three-dimensional modeling of the interaction between MIR200C-3p and the occludin mRNA 3'UTR identified sites of interaction. The antagomiR-200C prevents the decrease in occludin in enterocytes and intestine tissues of mice with colitis, maintaining the TJ barrier.


Asunto(s)
Colitis Ulcerosa/patología , Interleucina-1beta/metabolismo , MicroARNs/metabolismo , Ocludina/metabolismo , Uniones Estrechas/metabolismo , Animales , Células CACO-2 , Técnicas de Cultivo de Célula , Colitis Ulcerosa/etiología , Colitis Ulcerosa/metabolismo , Enterocitos , Humanos , Absorción Intestinal/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ocludina/genética , Permeabilidad , ARN Mensajero/metabolismo , Regulación hacia Arriba
4.
Int J Mol Sci ; 22(15)2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34360835

RESUMEN

Defective intestinal tight junction (TJ) barrier is a hallmark in the pathogenesis of inflammatory bowel disease (IBD). To date, there are no effective therapies that specifically target the intestinal TJ barrier. Among the various probiotic bacteria, Bifidobacterium, is one of the most widely studied to have beneficial effects on the intestinal TJ barrier. The main purpose of this study was to identify Bifidobacterium species that cause a sustained enhancement in the intestinal epithelial TJ barrier and can be used therapeutically to target the intestinal TJ barrier and to protect against or treat intestinal inflammation. Our results showed that Bifidobacterium bifidum caused a marked, sustained enhancement in the intestinal TJ barrier in Caco-2 monolayers. The Bifidobacterium bifidum effect on TJ barrier was strain-specific, and only the strain designated as BB1 caused a maximal enhancement in TJ barrier function. The mechanism of BB1 enhancement of intestinal TJ barrier required live bacterial cell/enterocyte interaction and was mediated by the BB1 attachment to Toll-like receptor-2 (TLR-2) at the apical membrane surface. The BB1 enhancement of the intestinal epithelial TJ barrier function was mediated by the activation of the p38 kinase pathway, but not the NF-κB signaling pathway. Moreover, the BB1 caused a marked enhancement in mouse intestinal TJ barrier in a TLR-2-dependent manner and protected against dextran sodium sulfate (DSS)-induced increase in mouse colonic permeability, and treated the DSS-induced colitis in a TJ barrier-dependent manner. These studies show that probiotic bacteria BB1 causes a strain-specific enhancement of the intestinal TJ barrier through a novel mechanism involving BB1 attachment to the enterocyte TLR-2 receptor complex and activation of p38 kinase pathway.


Asunto(s)
Bifidobacterium bifidum/fisiología , Colitis/microbiología , Mucosa Intestinal/microbiología , Transducción de Señal , Uniones Estrechas , Receptor Toll-Like 2/metabolismo , Animales , Células CACO-2 , Colitis/prevención & control , Humanos , Mucosa Intestinal/metabolismo , Ratones , FN-kappa B , Permeabilidad , Probióticos
5.
Am J Pathol ; 189(4): 797-812, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30711488

RESUMEN

Lipopolysaccharides (LPSs) are a major component of Gram-negative bacterial cell wall and play an important role in promoting intestinal inflammatory responses. Recent studies have shown that physiologically relevant concentrations of LPS (0 to 2000 pg/mL) cause an increase in intestinal epithelial tight junction (TJ) permeability without causing cell death. However, the intracellular pathways and the mechanisms that mediate LPS-induced increase in intestinal TJ permeability remain unclear. The aim was to delineate the intracellular pathways that mediate the LPS-induced increase in intestinal permeability using in vitro and in vivo intestinal epithelial models. LPS-induced increase in intestinal epithelial TJ permeability was preceded by an activation of transforming growth factor-ß-activating kinase-1 (TAK-1) and canonical NF-κB (p50/p65) pathways. The siRNA silencing of TAK-1 inhibited the activation of NF-κB p50/p65. The siRNA silencing of TAK-1 and p65/p50 subunit inhibited the LPS-induced increase in intestinal TJ permeability and the increase in myosin light chain kinase (MLCK) expression, confirming the regulatory role of TAK-1 and NF-κB p65/p50 in up-regulating MLCK expression and the subsequent increase in TJ permeability. The data also showed that toll-like receptor (TLR)-4/myeloid differentiation primary response (MyD)88 pathway was crucial upstream regulator of TAK-1 and NF-κB p50/p65 activation. In conclusion, activation of TAK-1 by the TLR-4/MyD88 signal transduction pathway and MLCK by NF-κB p65/p50 regulates the LPS-induced increase in intestinal epithelial TJ permeability.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Quinasa I-kappa B/metabolismo , Mucosa Intestinal/fisiología , Lipopolisacáridos/farmacología , Quinasas Quinasa Quinasa PAM/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Animales , Células CACO-2 , Proteínas de Unión al Calcio/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Quinasa I-kappa B/genética , Mucosa Intestinal/efectos de los fármacos , Quinasas Quinasa Quinasa PAM/genética , Masculino , Ratones Endogámicos C57BL , Quinasa de Cadena Ligera de Miosina/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Transducción de Señal , Uniones Estrechas/efectos de los fármacos , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
6.
Am J Physiol Gastrointest Liver Physiol ; 316(2): G278-G290, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30543452

RESUMEN

Matrix metalloproteinase-9 (MMP-9) has been implicated as being an important pathogenic factor in inflammatory bowel disease (IBD). MMP-9 is markedly elevated in intestinal tissue of patients with IBD, and IBD patients have a defective intestinal tight-junction (TJ) barrier manifested by an increase in intestinal permeability. The loss of intestinal epithelial barrier function is an important contributing factor in the development and prolongation of intestinal inflammation; however, the role of MMP-9 in intestinal barrier function remains unclear. The purpose of this study was to investigate the effect of MMP-9 on the intestinal epithelial TJ barrier and to delineate the intracellular mechanisms involved by using in vitro (filter-grown Caco-2 monolayers) and in vivo (mouse small intestine recycling perfusion) systems. MMP-9 caused a time- and dose-dependent increase in Caco-2 TJ permeability. MMP-9 also caused an increase in myosin light-chain kinase (MLCK) gene activity, protein expression, and enzymatic activity. The pharmacological MLCK inhibition and siRNA-induced knockdown of MLCK inhibited the MMP-9-induced increase in Caco-2 TJ permeability. MMP-9 caused a rapid activation of the p38 kinase signaling pathway and inhibition of p38 kinase activity prevented the MMP-9-induced increase in MLCK gene activity and the increase in Caco-2 TJ permeability. MMP-9 also caused an increase in mouse intestinal permeability in vivo, which was accompanied by an increase in MLCK expression. The MMP-9-induced increase in mouse intestinal permeability was inhibited in MLCK-deficient mice. These data show for the first time that the MMP-9-induced increase in intestinal TJ permeability in vitro and in vivo was mediated by the p38 kinase signal transduction pathway upregulation of MLCK gene activity and that therapeutic targeting of these pathways can prevent the MMP-9-induced increase in intestinal TJ permeability. NEW & NOTEWORTHY MMP-9 is highly elevated in patients with IBD. IBD patients have compromised intestinal TJ barrier function manifested by an increase in intestinal permeability and intestinal inflammation. This study shows that MMP-9, at clinically achievable concentrations, causes an increase in intestinal TJ permeability in vitro and in vivo. In addition, a MMP-9-induced increase in intestinal TJ permeability was mediated by an increase in MLCK gene and protein expression via the p38 kinase pathway.


Asunto(s)
Permeabilidad de la Membrana Celular/genética , Sistema de Señalización de MAP Quinasas/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Células CACO-2 , Células Epiteliales , Humanos , Intestinos/fisiología , Metaloproteinasa 9 de la Matriz/genética , Permeabilidad , Uniones Estrechas/genética , Uniones Estrechas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Am J Pathol ; 187(12): 2698-2710, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29157665

RESUMEN

Lipopolysaccharides (LPSs) are a major component of the Gram-negative bacterial cell wall and play an important role in mediating intestinal inflammatory responses in inflammatory bowel disease. Although recent studies suggested that physiologically relevant concentrations of LPS (0 to 1 ng/mL) cause an increase in intestinal epithelial tight junction (TJ) permeability, the mechanisms that mediate an LPS-induced increase in intestinal TJ permeability remain unclear. Herein, we show that myosin light chain kinase (MLCK) plays a central role in the LPS-induced increase in TJ permeability. Filter-grown Caco-2 intestinal epithelial monolayers and C57BL/6 mice were used as an in vitro and in vivo intestinal epithelial model system, respectively. LPS caused a dose- and time-dependent increase in MLCK expression and kinase activity in Caco-2 monolayers. The pharmacologic MLCK inhibition and siRNA-induced knock-down of MLCK inhibited the LPS-induced increase in Caco-2 TJ permeability. The LPS increase in TJ permeability was mediated by toll-like receptor 4 (TLR-4)/MyD88 signal-transduction pathway up-regulation of MLCK expression. The LPS-induced increase in mouse intestinal permeability also required an increase in MLCK expression. The LPS-induced increase in intestinal permeability was inhibited in MLCK-/- and TLR-4-/- mice. These data show, for the first time, that the LPS-induced increase in intestinal permeability was mediated by TLR-4/MyD88 signal-transduction pathway up-regulation of MLCK. Therapeutic targeting of these pathways can prevent an LPS-induced increase in intestinal permeability.


Asunto(s)
Mucosa Intestinal/metabolismo , Lipopolisacáridos/toxicidad , Factor 88 de Diferenciación Mieloide/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Uniones Estrechas/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Células CACO-2 , Humanos , Inflamación/metabolismo , Mucosa Intestinal/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Permeabilidad/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Uniones Estrechas/efectos de los fármacos
8.
Exp Cell Res ; 352(1): 113-122, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28161538

RESUMEN

Previous studies have demonstrated that the chloride channel ClC-2 plays a critical role in intestinal epithelial tight junction (TJ) barrier function via intracellular trafficking of TJ protein occludin. To study the mechanism of ClC-2-mediated TJ barrier function and intracellular trafficking of occludin, we established ClC-2 over-expressing Caco-2 cell line (Caco-2CLCN2) by full length ClC-2 ORF transfection. ClC-2 over-expression (Caco-2CLCN2) significantly enhanced TJ barrier (increased TER by ≥2 times and reduced inulin flux by 50%) compared to control Caco-2pEZ cells. ClC-2 over-expression (Caco-2CLCN2) increased occludin protein level compared to control Caco-2pEZ cells. Surface biotinylation assay revealed reduced steady state endocytosis of occludin in Caco-2CLCN2 cells. Furthermore, ClC-2 over-expression led to reduction in caveolin-1 protein level and diminishment of caveolae assembly. Caveolae disruption increased TJ permeability in control but not ClC-2 over-expressing Caco-2CLCN2 cells. Selective ClC-2 channel blocker GaTx2 caused an increase in caveolin-1 protein level and reduced occludin level. Delivery of cell permeable caveolin-1 scaffolding domain reduced the occludin protein level. Over all, these results suggest that ClC- 2 enhances TJ barrier function in intestinal epithelial cells via regulation of caveolin-1 and caveolae-mediated trafficking of occludin.


Asunto(s)
Caveolas/metabolismo , Caveolina 1/metabolismo , Canales de Cloruro/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Ocludina/metabolismo , Uniones Estrechas/metabolismo , Western Blotting , Canales de Cloruro CLC-2 , Permeabilidad de la Membrana Celular , Proliferación Celular , Células Cultivadas , Endocitosis/fisiología , Células Epiteliales/citología , Técnica del Anticuerpo Fluorescente , Humanos , Intestinos/citología , Transporte de Proteínas
9.
Am J Pathol ; 186(5): 1151-65, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26948423

RESUMEN

Tumor necrosis factor (TNF)-α, a key mediator of intestinal inflammation, causes an increase in intestinal epithelial tight junction (TJ) permeability by activating myosin light chain kinase (MLCK; official name MYLK3) gene. However, the precise signaling cascades that mediate the TNF-α-induced activation of MLCK gene and increase in TJ permeability remain unclear. Our aims were to delineate the upstream signaling mechanisms that regulate the TNF-α modulation of intestinal TJ barrier function with the use of in vitro and in vivo intestinal epithelial model systems. TNF-α caused a rapid activation of both canonical and noncanonical NF-κB pathway. NF-κB-inducing kinase (NIK) and mitogen-activated protein kinase kinase-1 (MEKK-1) were activated in response to TNF-α. NIK mediated the TNF-α activation of inhibitory κB kinase (IKK)-α, and MEKK1 mediated the activation of IKK complex, including IKK-ß. NIK/IKK-α axis regulated the activation of both NF-κB p50/p65 and RelB/p52 pathways. Surprisingly, the siRNA induced knockdown of NIK, but not MEKK-1, prevented the TNF-α activation of both NF-κB p50/p65 and RelB/p52 and the increase in intestinal TJ permeability. Moreover, NIK/IKK-α/NF-κB p50/p65 axis mediated the TNF-α-induced MLCK gene activation and the subsequent MLCK increase in intestinal TJ permeability. In conclusion, our data show that NIK/IKK-α/regulates the activation of NF-κB p50/p65 and plays an integral role in the TNF-α-induced activation of MLCK gene and increase in intestinal TJ permeability.


Asunto(s)
Quinasa I-kappa B/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , FN-kappa B/metabolismo , Uniones Estrechas/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Células CACO-2 , Células Cultivadas , Humanos , Intestino Delgado/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/fisiología , FN-kappa B/antagonistas & inhibidores , Subunidad p50 de NF-kappa B/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , Permeabilidad , Regiones Promotoras Genéticas/fisiología , ARN Interferente Pequeño/metabolismo , Factor de Transcripción ReIA/metabolismo , Transfección , Factor de Necrosis Tumoral alfa/farmacología
10.
J Immunol ; 195(10): 4999-5010, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26466961

RESUMEN

Gut-derived bacterial LPS plays an essential role in inducing intestinal and systemic inflammatory responses and have been implicated as a pathogenic factor in necrotizing enterocolitis and inflammatory bowel disease. The defective intestinal tight junction barrier was shown to be an important factor contributing to the development of intestinal inflammation. LPS, at physiological concentrations, causes an increase in intestinal tight junction permeability (TJP) via a TLR4-dependent process; however, the intracellular mechanisms that mediate LPS regulation of intestinal TJP remain unclear. The aim of this study was to investigate the adaptor proteins and the signaling interactions that mediate LPS modulation of intestinal tight junction barrier using in vitro and in vivo model systems. LPS caused a TLR4-dependent activation of membrane-associated adaptor protein focal adhesion kinase (FAK) in Caco-2 monolayers. LPS caused an activation of both MyD88-dependent and -independent pathways. Small interfering RNA silencing of MyD88 prevented an LPS-induced increase in TJP. LPS caused MyD88-dependent activation of IL-1R-associated kinase 4. TLR4, FAK, and MyD88 were colocalized. Small interfering silencing of TLR4 inhibited TLR4-associated FAK activation, and FAK knockdown prevented MyD88 activation. In vivo studies also confirmed that the LPS-induced increase in mouse intestinal permeability was associated with FAK and MyD88 activation; knockdown of intestinal epithelial FAK prevented an LPS-induced increase in intestinal permeability. Additionally, high-dose LPS-induced intestinal inflammation was dependent on the TLR4/FAK/MyD88 signal transduction axis. To our knowledge, our data show for the first time that the LPS-induced increases in intestinal TJP and intestinal inflammation were regulated by TLR4-dependent activation of the FAK/MyD88/IL-1R-associated kinase 4 signaling pathway.


Asunto(s)
Quinasa 1 de Adhesión Focal/inmunología , Lipopolisacáridos/farmacología , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal/efectos de los fármacos , Uniones Estrechas/inmunología , Receptor Toll-Like 4/inmunología , Animales , Células CACO-2 , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Activación Enzimática/inmunología , Quinasa 1 de Adhesión Focal/genética , Humanos , Intestinos/inmunología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Permeabilidad/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Uniones Estrechas/genética , Receptor Toll-Like 4/genética
11.
J Biol Chem ; 290(11): 7234-46, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25616664

RESUMEN

Autophagy is an intracellular degradation pathway and is considered to be an essential cell survival mechanism. Defects in autophagy are implicated in many pathological processes, including inflammatory bowel disease. Among the innate defense mechanisms of intestinal mucosa, a defective tight junction (TJ) barrier has been postulated as a key pathogenic factor in the causation and progression of inflammatory bowel disease by allowing increased antigenic permeation. The cross-talk between autophagy and the TJ barrier has not yet been described. In this study, we present the novel finding that autophagy enhances TJ barrier function in Caco-2 intestinal epithelial cells. Nutrient starvation-induced autophagy significantly increased transepithelial electrical resistance and reduced the ratio of sodium/chloride paracellular permeability. Nutrient starvation reduced the paracellular permeability of small-sized urea but not larger molecules. The role of autophagy in the modulation of paracellular permeability was confirmed by pharmacological induction as well as pharmacological and genetic inhibition of autophagy. Consistent with the autophagy-induced reduction in paracellular permeability, a marked decrease in the level of the cation-selective, pore-forming TJ protein claudin-2 was observed after cell starvation. Starvation reduced the membrane presence of claudin-2 and increased its cytoplasmic, lysosomal localization. Therefore, our data show that autophagy selectively reduces epithelial TJ permeability of ions and small molecules by lysosomal degradation of the TJ protein claudin-2.


Asunto(s)
Autofagia , Claudina-2/metabolismo , Células Epiteliales/citología , Mucosa Intestinal/citología , Proteolisis , Uniones Estrechas/metabolismo , Células CACO-2 , Células Epiteliales/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Permeabilidad
12.
J Immunol ; 190(12): 6596-606, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23656735

RESUMEN

The defective intestinal epithelial tight junction (TJ) barrier has been postulated to be an important pathogenic factor contributing to intestinal inflammation. It has been shown that the proinflammatory cytokine IL-1ß causes an increase in intestinal permeability; however, the signaling pathways and the molecular mechanisms involved remain unclear. The major purpose of this study was to investigate the role of the p38 kinase pathway and the molecular processes involved. In these studies, the in vitro intestinal epithelial model system (Caco-2 monolayers) was used to delineate the cellular and molecular mechanisms, and a complementary in vivo mouse model system (intestinal perfusion) was used to assess the in vivo relevance of the in vitro findings. Our data indicated that the IL-1ß increase in Caco-2 TJ permeability correlated with an activation of p38 kinase. The activation of p38 kinase caused phosphorylation and activation of p38 kinase substrate, activating transcription factor (ATF)-2. The activated ATF-2 translocated to the nucleus where it attached to its binding motif on the myosin L chain kinase (MLCK) promoter region, leading to the activation of MLCK promoter activity and gene transcription. Small interfering RNA induced silencing of ATF-2, or mutation of the ATF-2 binding motif prevented the activation of MLCK promoter and MLCK mRNA transcription. Additionally, in vivo intestinal perfusion studies also indicated that the IL-1ß increase in mouse intestinal permeability required p38 kinase-dependent activation of ATF-2. In conclusion, these studies show that the IL-1ß-induced increase in intestinal TJ permeability in vitro and in vivo was regulated by p38 kinase activation of ATF-2 and by ATF-2 regulation of MLCK gene activity.


Asunto(s)
Factor de Transcripción Activador 2/metabolismo , Interleucina-1beta/metabolismo , Mucosa Intestinal/metabolismo , Quinasa de Cadena Ligera de Miosina/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Factor de Transcripción Activador 2/genética , Animales , Western Blotting , Células CACO-2 , Permeabilidad de la Membrana Celular/fisiología , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/fisiología , Humanos , Inmunidad Mucosa/fisiología , Ratones , Regiones Promotoras Genéticas , Transporte de Proteínas/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Uniones Estrechas/genética , Uniones Estrechas/metabolismo , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/genética
13.
Am J Pathol ; 182(2): 375-87, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23201091

RESUMEN

Bacterial-derived lipopolysaccharides (LPS) play an essential role in the inflammatory process of inflammatory bowel disease. A defective intestinal tight junction (TJ) barrier is an important pathogenic factor of inflammatory bowel disease and other inflammatory conditions of the gut. Despite its importance in mediating intestinal inflammation, the physiological effects of LPS on the intestinal epithelial barrier remain unclear. The major aims of this study were to determine the effects of physiologically relevant concentrations of LPS (0 to 1 ng/mL) on intestinal barrier function using an in vitro (filter-grown Caco-2 monolayers) and an in vivo (mouse intestinal perfusion) intestinal epithelial model system. LPS, at physiologically relevant concentrations (0 to 1 ng/mL), in the basolateral compartment produced a time-dependent increase in Caco-2 TJ permeability without inducing cell death. Intraperitoneal injection of LPS (0.1 mg/kg), leading to clinically relevant plasma concentrations, also caused a time-dependent increase in intestinal permeability in vivo. The LPS-induced increase in intestinal TJ permeability was mediated by an increase in enterocyte membrane TLR-4 expression and a TLR-4-dependent increase in membrane colocalization of membrane-associated protein CD14. In conclusion, these studies show for the first time that LPS causes an increase in intestinal permeability via an intracellular mechanism involving TLR-4-dependent up-regulation of CD14 membrane expression.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Enterocitos/citología , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Uniones Estrechas/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células CACO-2 , Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Uniones Estrechas/efectos de los fármacos , Factores de Tiempo , Receptor Toll-Like 4/sangre , Regulación hacia Arriba/efectos de los fármacos
14.
Am J Pathol ; 183(6): 1871-1884, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24121020

RESUMEN

Tumor necrosis factor (TNF-α) is a proinflammatory cytokine that plays a critical role in the pathogenesis of inflammatory bowel disease. TNF-α causes an increase in intestinal permeability; however, the signaling pathways and the molecular mechanisms involved remain unclear. The major purpose of this study was to investigate the role of MAP kinase pathways (ERK1/2 and p38 kinase) and the molecular processes involved. An in vitro intestinal epithelial model system consisting of Caco-2 monolayers and an in vivo mouse model system were used to delineate the cellular and molecular mechanisms involved in TNF-α effects on tight junction barrier. The TNF-α-induced increase in Caco-2 tight junction permeability was mediated by activation of the ERK1/2 signaling pathway, but not the p38 kinase pathway. Activation of the ERK1/2 pathway led to phosphorylation and activation of the ETS domain-containing transcription factor Elk-1. The activated Elk-1 translocated to the nucleus, where it bound to its binding motif on the myosin light chain kinase (MLCK) promoter region, leading to the activation of MLCK promoter activity and gene transcription. In addition, in vivo intestinal perfusion studies also indicated that the TNF-α-induced increase in mouse intestinal permeability requires ERK1/2-dependent activation of Elk-1. These studies provide novel insight into the cellular and molecular processes that regulate the TNF-α-induced increase in intestinal epithelial tight junction permeability.


Asunto(s)
Núcleo Celular/enzimología , Mucosa Intestinal/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Uniones Estrechas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína Elk-1 con Dominio ets/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Células CACO-2 , Núcleo Celular/genética , Activación Enzimática , Humanos , Mucosa Intestinal/citología , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Permeabilidad , Regiones Promotoras Genéticas/fisiología , Uniones Estrechas/genética , Transcripción Genética/fisiología , Factor de Necrosis Tumoral alfa/genética , Proteína Elk-1 con Dominio ets/genética
15.
Front Immunol ; 15: 1348010, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39081324

RESUMEN

Background: Defective intestinal epithelial tight junction (TJ), characterized by an increase in intestinal TJ permeability, has been shown to play a critical role in the pathogenesis of inflammatory bowel disease (IBD). Tumor necrosis factor-α (TNF-α) is a key pro-inflammatory cytokine involved in the immunopathology of IBD and has been shown to cause an increase in intestinal epithelial TJ permeability. Although TNF-α antibodies and other biologics have been advanced for use in IBD treatment, these therapies are associated with severe side effects and have limited efficacy, and there is an urgent need for therapies with benign profiles and high therapeutic efficacy. Probiotic bacteria have beneficial effects and are generally safe and represent an important class of potential therapeutic agents in IBD. Lactobacillus acidophilus (LA) is one of the most used probiotics for wide-ranging health benefits, including in gastrointestinal, metabolic, and inflammatory disorders. A specific strain of LA, LA1, was recently demonstrated to have protective and therapeutic effects on the intestinal epithelial TJ barrier. However, the mechanisms of actions of LA1 remain largely unknown. Methods: The primary aim of this study was to investigate microbial-epithelial interactions and novel signaling pathways that regulate the effect of LA1 on TNF-α-induced increase in intestinal epithelial TJ permeability, using cell culture and animal model systems. Results and Conclusion: Pre-treatment of filter-grown Caco-2 monolayers with LA1 prevented the TNF-α-induced increase in intestinal epithelial TJ permeability by inhibiting TNF-α-induced activation of NF-κB p50/p65 and myosin light chain kinase (MLCK) gene and kinase activity in a TLR-2-dependent manner. LA1 produced a TLR-2- and MyD88-dependent activation of NF-κB p50/p65 in immune cells; however, LA1, in intestinal cells, inhibited the NF-κB p50/p65 activation in a TLR-2-dependent but MyD88-independent manner. In addition, LA1 inhibition of NF-κB p50/p65 and MLCK gene was mediated by TLR-2 pathway activation of phosphatidylinositol 3-kinase (PI3K) and IKK-α phosphorylation. Our results demonstrated novel intracellular signaling pathways by which LA1/TLR-2 suppresses the TNF-α pathway activation of NF-κB p50/p65 in intestinal epithelial cells and protects against the TNF-α-induced increase in intestinal epithelial TJ permeability.


Asunto(s)
Mucosa Intestinal , Lactobacillus acidophilus , FN-kappa B , Fosfatidilinositol 3-Quinasas , Probióticos , Uniones Estrechas , Receptor Toll-Like 2 , Factor de Necrosis Tumoral alfa , Lactobacillus acidophilus/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Uniones Estrechas/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Animales , Probióticos/farmacología , Receptor Toll-Like 2/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , FN-kappa B/metabolismo , Ratones , Permeabilidad , Transducción de Señal/efectos de los fármacos , Células CACO-2 , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo
16.
Microbiol Resour Announc ; 13(4): e0067723, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38488370

RESUMEN

We present the complete genome sequence of the probiotic strain Lactobacillus acidophilus ATCC 9224. The genome sequence provides a valuable resource for investigating the phylogenetic evolution of this lineage and conducting comparative genomics with other Lactobacillus strains and species.

17.
Biochim Biophys Acta ; 1808(12): 3016-21, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21854757

RESUMEN

Transport of riboflavin (RF) across both the brush border membrane (BBM) and basolateral membrane (BLM) of the polarized enterocyte occurs via specific carrier-mediated mechanisms. Although, three human riboflavin transporters (hRFTs), i.e., hRFT-1, hRFT-2 and hRFT-3 are expressed in the intestine, little is known about the cell surface domain(s) at which these specific hRFTs are expressed. Here, we used live cell confocal imaging of intestinal epithelial Caco-2 and renal MDCK cells to show that the hRFT-1 is mainly expressed at the BLM, hRFT-2 is exclusively expressed at the apical membrane, while hRFT-3 is mostly localized inside intracellular vesicular structures (with some expression at the BLM). Further the level of hRFT-2 mRNA expression in Caco-2 cells and in native human intestine is significantly higher than that of hRFT-1 and -3; hRFT-2 was also more efficient in transporting 3H-RF than hRFT-1 and -3. These findings implied an important role for hRFT-2 in intestinal RF uptake, a conclusion that was further supported by findings of hRFT-2 gene-specific siRNA knockdown investigation. These results show that members of the hRFT family are differentially expressed in polarized epithelia, and that the apically expressed hRFT-2 plays a key role in intestinal RF accumulation.


Asunto(s)
Proteínas Portadoras/metabolismo , Mucosa Intestinal/metabolismo , Riboflavina/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Mucosa Intestinal/citología , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Gastroenterology ; 141(4): 1323-33, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21763238

RESUMEN

BACKGROUND & AIMS: Defects in the intestinal epithelial tight junction (TJ) barrier contribute to intestinal inflammation. A tumor necrosis factor (TNF)-α-induced increase in intestinal TJ permeability contributes to the intestinal TJ barrier defect in inflammatory disorders. We investigated the mechanisms by which TNF-α induces occludin depletion and an increase in intestinal TJ permeability. METHODS: We assessed intestinal TJ barrier function using intestinal epithelial model systems: filter-grown Caco-2 monolayers and recycling perfusion studies of mouse small intestine. RESULTS: TNF-α caused a rapid increase in expression of microRNA (miR)-122a in enterocytes, cultured cells, and intestinal tissue. The overexpressed miR-122a bound to a binding motif at the 3'-untranslated region of occludin messenger RNA (mRNA) to induce its degradation; mRNA degradation depleted occludin from enterocytes, resulting in increased intestinal TJ permeability. Transfection of enterocytes with an antisense oligoribonucleotide against miR-122a blocked the TNF-α-induced increase in enterocyte expression of miR-122a, degradation of occludin mRNA, and increase in intestinal permeability. Overexpression of miR-122a in enterocytes using pre-miR-122a was sufficient to induce degradation of occludin mRNA and an increase in intestinal permeability. CONCLUSIONS: TNF-α regulates intestinal permeability by inducing miR-122a-mediated degradation of occludin mRNA. These studies show the feasibility of therapeutically targeting miR-122a in vivo to preserve the intestinal barrier.


Asunto(s)
Células Epiteliales/metabolismo , Absorción Intestinal , Mucosa Intestinal/metabolismo , MicroARNs/metabolismo , Uniones Estrechas/metabolismo , Regiones no Traducidas 3' , Animales , Células CACO-2 , Humanos , Inulina/metabolismo , Cinética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ocludina , Oligorribonucleótidos Antisentido/metabolismo , Permeabilidad , Estabilidad del ARN , ARN Mensajero/metabolismo , Transfección , Factor de Necrosis Tumoral alfa/metabolismo
19.
J Cell Mol Med ; 15(4): 970-82, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20406328

RESUMEN

Interleukin-1ß (IL-1ß) is a prototypical multifunctional cytokine that plays an important role in intestinal inflammation of Crohn's disease and other inflammatory conditions of the gut. Previous studies have shown that IL-1ß causes an increase in intestinal epithelial tight junction (TJ) permeability both in in vivo animal and in vitro cell culture model systems. The IL-1ß-induced increase in intestinal epithelial TJ permeability has been postulated to be an important pathogenic mechanism contributing to intestinal inflammation. However, the signalling pathways and the molecular processes that mediate the IL-1ß modulation of intestinal epithelial TJ barrier remain unclear. Here, we show that the IL-1ß-induced increase in Caco-2 monolayer TJ permeability was mediated by activation of extracellular signal-regulated kinases 1/2 (ERK1/2) signalling pathway and that inhibition of ERK1/2 activity inhibits the IL-1ß-induced increase in Caco-2 TJ permeability. The activation of ERK1/2 pathway caused a downstream activation of nuclear transcription factor Elk-1. The activated Elk-1 translocated to the nucleus and binds to the cis-binding motif on myosin light chain kinase (MLCK) promoter region, triggering MLCK gene activation, MLCK mRNA transcription and MLCK protein synthesis and MLCK catalysed opening of the intestinal epithelial TJ barrier. These studies provide novel insight into the cellular and molecular processes that mediate the IL-1ß-induced increase in intestinal epithelial TJ permeability.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Interleucina-1beta/farmacología , Intestinos/citología , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Secuencia de Bases , Células CACO-2 , ADN/metabolismo , Activación Enzimática/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/enzimología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Flavonoides/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Datos de Secuencia Molecular , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Permeabilidad/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Factores de Tiempo , Factor de Transcripción AP-1/metabolismo , Proteína Elk-1 con Dominio ets/metabolismo
20.
Am J Pathol ; 177(5): 2310-22, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21048223

RESUMEN

IL-1ß is a proinflammatory cytokine that plays a central role in the inflammatory process of the gut. IL-1ß causes an increase in intestinal epithelial tight junction (TJ) permeability, but the intracellular pathways that mediate intestinal TJ permeability remain unclear. The major aims of this study were to delineate the protein kinases that regulate the IL-1ß modulation of intestinal TJ barrier function and to determine the intracellular mechanisms involved, using filter-grown Caco-2 monolayers as the in vitro model system. Our results showed that IL-1ß caused a rapid activation of MEKK-1 and NIK. The knockdown of MEKK-1, but not NIK, inhibited the IL-1ß increase in Caco-2 TJ permeability. IL-1ß caused an activation of both canonical and noncanonical NF-κB pathways; MEKK-1 regulated the activation of the canonical pathway, while NIK regulated the activation of the noncanonical pathway. Inhibition of MEKK-1 activation of the canonical pathway prevented the IL-1ß increase in TJ permeability. Our data also indicated that inhibitory κB kinase was the catalytic subunit primarily involved in canonical pathway activation and TJ barrier opening. MEKK-1 also played an essential role in myosin light chain kinase gene activation. In conclusion, our data show for the first time that MEKK-1 plays an integral role in IL-1ß modulation of Caco-2 TJ barrier function by regulating the activation of the canonical NF-κB pathway and the MLCK gene.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Interleucina-1beta/metabolismo , Mucosa Intestinal/citología , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , Uniones Estrechas/metabolismo , Factor de Transcripción ReIA/metabolismo , Células CACO-2 , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Mucosa Intestinal/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Subunidad p52 de NF-kappa B/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factor de Transcripción ReIA/genética , Quinasa de Factor Nuclear kappa B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA