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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 36(1): e22061, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34861075

RESUMEN

Corticosterone, the stress hormone, exacerbates alcohol-associated tissue injury, but the mechanism involved is unknown. We examined the role of the glucocorticoid receptor (GR) in corticosterone-mediated potentiation of alcohol-induced gut barrier dysfunction and systemic response. Hepatocyte-specific GR-deficient (GRΔHC ) and intestinal epithelial-specific GR-deficient (GRΔIEC ) mice were fed ethanol, combined with corticosterone treatment. Intestinal epithelial tight junction integrity, mucosal barrier function, microbiota dysbiosis, endotoxemia, systemic inflammation, liver damage, and neuroinflammation were assessed. Corticosterone potentiated ethanol-induced epithelial tight junction disruption, mucosal permeability, and inflammatory response in GRΔHC mouse colon; these effects of ethanol and corticosterone were absent in GRΔIEC mice. Gut microbiota compositions in ethanol-fed GRΔHC and GRΔIEC mice were similar to each other. However, corticosterone treatment in ethanol-fed mice shifted the microbiota composition to distinctly different directions in GRΔHC and GRΔIEC mice. Ethanol and corticosterone synergistically elevated the abundance of Enterobacteriaceae and Escherichia coli and reduced the abundance of Lactobacillus in GRΔHC mice but not in GRΔIEC mice. In GRΔHC mice, corticosterone potentiated ethanol-induced endotoxemia and systemic inflammation, but these effects were absent in GRΔIEC mice. Interestingly, ethanol-induced liver damage and its potentiation by corticosterone were observed in GRΔHC mice but not in GRΔIEC mice. GRΔIEC mice were also resistant to ethanol- and corticosterone-induced inflammatory response in the hypothalamus. These data indicate that the intestinal epithelial GR plays a central role in alcohol- and corticosterone-induced gut barrier dysfunction, microbiota dysbiosis, endotoxemia, systemic inflammation, liver damage, and neuroinflammation. This study identifies a novel target for potential therapeutic for alcohol-associated tissue injury.


Asunto(s)
Corticosterona/efectos adversos , Etanol/efectos adversos , Mucosa Intestinal/metabolismo , Receptores de Glucocorticoides/metabolismo , Uniones Estrechas/metabolismo , Animales , Corticosterona/farmacología , Escherichia coli/metabolismo , Etanol/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Lactobacillus/metabolismo , Ratones , Ratones Transgénicos , Permeabilidad/efectos de los fármacos , Receptores de Glucocorticoides/genética , Uniones Estrechas/genética
2.
Nutr Neurosci ; 25(4): 871-883, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32945721

RESUMEN

Neuroinflammation is implicated in the pathogenesis of alcohol use disorders. We investigated the role of Gut-Brain interactions in alcohol-induced neuroinflammation by probiotic-mediated manipulation of intestinal dysbiosis in mice. Chronic ethanol feeding induced dysbiosis, as evidenced by an increase in Firmicutes/Bacteroidetes ratio and depletion of Lactobacillus species in the colon. Ethanol increased the levels of IL-1ß, IL-6, and TNFα in plasma and the mRNA for IL-1ß, IL-6, TNFα, and MCP1 genes in the cerebral cortex and hippocampus. Ethanol feeding increased inulin flux from the circulation into different brain regions, accompanied by the increase in TLR4 mRNA levels in the cerebral cortex and hippocampus. The immunofluorescence confocal microscopy showed that ethanol elevates the expression of microglial activation marker TMEM119 in the cerebral cortex. Feeding L. plantarum suppressed the ethanol-induced dysbiosis to some extent, as evidenced by attenuation of ethanol effects on Firmicutes/Bacteroidetes ratio and abundance of Lactobacillus spp. L. plantarum blocked ethanol-induced elevation of plasma cytokines, inulin permeability to the brain, mRNA for TLR4, IL-1ß, IL-6, TNFα, and MCP1 in brain regions, and the expression of TMEM119 in the cerebral cortex. The L. plantarum effect was absent in mice that express a dominant-negative EGFR, suggesting that the EGFR receptor plays an essential role in the protective effect of L. plantarum against ethanol-induced neuroinflammation. L. plantarum, when administered after chronic ethanol-induced injury, rescued the ethanol-induced systemic inflammation and neuroinflammation. This study demonstrates that L. plantarum in the gut prevents and mitigates ethanol-induced neuroinflammation by an EGFR-dependent mechanism.


Asunto(s)
Alcoholismo , Lactobacillus plantarum , Animales , Receptores ErbB , Etanol/toxicidad , Ratones , Enfermedades Neuroinflamatorias
3.
Phys Biol ; 18(2): 026001, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33207323

RESUMEN

Nanoscale structural alteration in the nuclei of cells with the progression of carcinogenesis is due to the rearrangements of the basic building blocks in the cell such as DNA, RNA, lipids, etc. Although epigenetic modifications underlie the development of cancer, exposure to carcinogenic chemicals such as alcohol also enhances the development of cancer. We report the effects of chronic alcoholism on early-carcinogenesis based on changes in the degree of nanoscale structural alterations (L d) in nuclei. For this, transmission electron microscopy (TEM) imaging of the nuclei of colonic cells is performed for the following four mouse models: control mice; chronic alcoholic mice treated with ethanol (i.e., EtOH mice); mice treated with colonic carcinogen azoxymethane (AOM) and dextran sulfate sodium (DSS) that induced colitis (i.e., AOM + DSS mice); and chronic alcoholic or EtOH treated mice, together with AOM and DSS treatment (i.e., AOM + DSS + EtOH mice). The disordered optical lattices are constructed from their respective TEM images of thin colonic cell nuclei and the L d values are calculated using the inverse participation ratio (IPR) technique from the spatially localized eigenfunctions of these lattices. Results show no significant difference in the average L d value of the colon cell nuclei of alcohol treated mice relative to its control [i.e., L d(C) ∼ L d(EtOH)]; however, an increase in the L d value of alcohol treated precancerous cells [i.e., L d(AOM + DSS + EtOH) > L d(AOM + DSS)], indicating that alcohol accelerates the early carcinogenic process.


Asunto(s)
Alcoholismo/complicaciones , Carcinogénesis/ultraestructura , Núcleo Celular/ultraestructura , Animales , Carcinogénesis/inducido químicamente , Enfermedad Crónica , Femenino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión
4.
FASEB J ; 34(9): 11641-11657, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32654268

RESUMEN

The tight junction (TJ) and barrier function of colonic epithelium is highly sensitive to ionizing radiation. We evaluated the effect of lysophosphatidic acid (LPA) and its analog, Radioprotein-1, on γ-radiation-induced colonic epithelial barrier dysfunction using Caco-2 and m-ICC12 cell monolayers in vitro and mice in vivo. Mice were subjected to either total body irradiation (TBI) or partial body irradiation (PBI-BM5). Intestinal barrier function was assessed by analyzing immunofluorescence localization of TJ proteins, mucosal inulin permeability, and plasma lipopolysaccharide (LPS) levels. Oxidative stress was analyzed by measuring protein thiol oxidation and antioxidant mRNA. In Caco-2 and m-ICC12 cell monolayers, LPA attenuated radiation-induced redistribution of TJ proteins, which was blocked by a Rho-kinase inhibitor. In mice, TBI and PBI-BM5 disrupted colonic epithelial tight junction and adherens junction, increased mucosal permeability, and elevated plasma LPS; TJ disruption by TBI was more severe in Lpar2-/- mice compared to wild-type mice. RP1, administered before or after irradiation, alleviated TBI and PBI-BM5-induced TJ disruption, barrier dysfunction, and endotoxemia accompanied by protein thiol oxidation and downregulation of antioxidant gene expression, cofilin activation, and remodeling of the actin cytoskeleton. These data demonstrate that LPAR2 receptor activation prevents and mitigates γ-irradiation-induced colonic mucosal barrier dysfunction and endotoxemia.


Asunto(s)
Colon/efectos de la radiación , Mucosa Intestinal/efectos de la radiación , Radiación Ionizante , Receptores del Ácido Lisofosfatídico/genética , Uniones Estrechas/efectos de la radiación , Uniones Adherentes/efectos de los fármacos , Uniones Adherentes/metabolismo , Uniones Adherentes/efectos de la radiación , Animales , Células CACO-2 , Línea Celular , Colon/efectos de los fármacos , Colon/metabolismo , Humanos , Uniones Intercelulares/efectos de los fármacos , Uniones Intercelulares/metabolismo , Uniones Intercelulares/efectos de la radiación , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Lisofosfolípidos/farmacología , Ratones Noqueados , Permeabilidad/efectos de los fármacos , Permeabilidad/efectos de la radiación , Receptores del Ácido Lisofosfatídico/metabolismo , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo
5.
J Cell Sci ; 131(7)2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29507118

RESUMEN

The apical junctional complex (AJC), which includes tight junctions (TJs) and adherens junctions (AJs), determines the epithelial polarity, cell-cell adhesion and permeability barrier. An intriguing characteristic of a TJ is the dynamic nature of its multiprotein complex. Occludin is the most mobile TJ protein, but its significance in TJ dynamics is poorly understood. On the basis of phosphorylation sites, we distinguished a sequence in the C-terminal domain of occludin as a regulatory motif (ORM). Deletion of ORM and expression of a deletion mutant of occludin in renal and intestinal epithelia reduced the mobility of occludin at the TJs. ORM deletion attenuated Ca2+ depletion, osmotic stress and hydrogen peroxide-induced disruption of TJs, AJs and the cytoskeleton. The double point mutations T403A/T404A, but not T403D/T404D, in occludin mimicked the effects of ORM deletion on occludin mobility and AJC disruption by Ca2+ depletion. Both Y398A/Y402A and Y398D/Y402D double point mutations partially blocked AJC disruption. Expression of a deletion mutant of occludin attenuated collective cell migration in the renal and intestinal epithelia. Overall, this study reveals the role of ORM and its phosphorylation in occludin mobility, AJC dynamics and epithelial cell migration.


Asunto(s)
Uniones Adherentes/química , Ocludina/química , Fosfoproteínas/química , Uniones Estrechas/química , Uniones Adherentes/genética , Animales , Calcio/metabolismo , Movimiento Celular/genética , Polaridad Celular/genética , Citoesqueleto/química , Citoesqueleto/genética , Perros , Células Epiteliales/química , Recuperación de Fluorescencia tras Fotoblanqueo , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Humanos , Uniones Intercelulares/química , Uniones Intercelulares/genética , Células de Riñón Canino Madin Darby , Ocludina/genética , Fosfoproteínas/genética , Fosforilación/genética , Mutación Puntual/genética , Dominios Proteicos/genética , Uniones Estrechas/genética
6.
J Cell Biochem ; 120(11): 18826-18844, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31286540

RESUMEN

Hepatic copper levels differ among patients with Wilson disease (WD) and normal individuals depending on the dietary intake, copper bioavailability, and genetic factors. Copper chloride (CuCl2 ) caused dose-dependent reduction in cell viability of human teratocarcinoma (HepG2) cell line, measured using the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay. Cells were exposed to different concentrations of CuCl2 in log doses and maximum cell viability reduction was recorded at 15 µg/mL. Toxic dose of CuCl2 is potent inducer of reactive oxygen species (ROS). Apoptosis as a pattern of cell death was confirmed through sub-G1 fraction and morphological changes such as mitochondrial depolarization, endoplasmic reticulum and lysosomal destabilization, phosphatidylserine translocation, and DNA damage. Our transcriptional and translational results strongly support apoptotic cell death. Using the available data present in dbSNP and bioinformatics tools, three nonsynonymous single nucleotide polymorphisms (nsSNPs) were identified as deleterious, reducing the stability of protein ATP7B. Structural analysis of native and mutant ATP7B proteins was investigated using molecular dynamics simulation (MDS) approach. Mutation in ATP7B gene might disturb the structural conformation and catalytic function of the ATP7B protein may be inducing WD. Hence, excess dietary intake of copper chloride must be avoided for safety of health to prevent from WD.


Asunto(s)
Carcinoma Hepatocelular , ATPasas Transportadoras de Cobre , Degeneración Hepatolenticular , Neoplasias Hepáticas , Modelos Biológicos , Proteínas de Neoplasias , Apoptosis , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Cobre/metabolismo , ATPasas Transportadoras de Cobre/genética , ATPasas Transportadoras de Cobre/metabolismo , Daño del ADN , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
FASEB J ; : fj201800351R, 2018 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-29912589

RESUMEN

Pathogenesis of alcohol-related diseases such as alcoholic hepatitis involves gut barrier dysfunction, endotoxemia, and toxin-mediated cellular injury. Here we show that Lactobacillus plantarum not only blocks but also mitigates ethanol (EtOH)-induced gut and liver damage in mice. L. plantarum blocks EtOH-induced protein thiol oxidation, and down-regulation of antioxidant gene expression in colon L. plantarum also blocks EtOH-induced expression of TNF-α, IL-1ß, IL-6, monocyte chemotactic protein 1 ( MCP1), C-X-C motif chemokine ligand ( CXCL)1, and CXCL2 genes in colon. Epidermal growth factor receptor (EGFR) signaling mediates the L. plantarum-mediated protection of tight junctions (TJs) and barrier function from acetaldehyde, the EtOH metabolite, in Caco-2 cell monolayers. In mice, doxycycline-mediated expression of dominant negative EGFR blocks L. plantarum-mediated prevention of EtOH-induced TJ disruption, mucosal barrier dysfunction, oxidative stress, and inflammatory response in colon. L. plantarum blocks EtOH-induced endotoxemia as well as EtOH-induced pathologic lesions, triglyceride deposition, oxidative stress, and inflammatory responses in the liver by an EGFR-dependent mechanism. L. plantarum treatment after injury accelerated recovery from EtOH-induced TJ, barrier dysfunction, oxidative stress, and inflammatory response in colon, endotoxemia, and liver damage. Results demonstrate that L. plantarum has both preventive and therapeutic values in treatment of alcohol-induced tissue injury, particularly in alcoholic hepatitis.-Shukla, P. K., Meena, A. S., Manda, B., Gomes-Solecki, M., Dietrich, P., Dragatsis, I., Rao, R. Lactobacillus plantarum prevents and mitigates alcohol-induced disruption of colonic epithelial tight junctions, endotoxemia, and liver damage by an EGF receptor-dependent mechanism.

8.
Biochem J ; 474(5): 731-749, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28057718

RESUMEN

The role of reactive oxygen species (ROS) in osmotic stress, dextran sulfate sodium (DSS) and cyclic stretch-induced tight junction (TJ) disruption was investigated in Caco-2 cell monolayers in vitro and restraint stress-induced barrier dysfunction in mouse colon in vivo Live cell imaging showed that osmotic stress, cyclic stretch and DSS triggered rapid production of ROS in Caco-2 cell monolayers, which was blocked by depletion of intracellular Ca2+ by 1,2-bis-(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid. Knockdown of CaV1.3 or TRPV6 channels blocked osmotic stress and DSS-induced ROS production and attenuated TJ disruption and barrier dysfunction. N-Acetyl l-cysteine (NAC) and l-NG-Nitroarginine methyl ester (l-NAME) blocked stress-induced TJ disruption and barrier dysfunction. NAC and l-NAME also blocked stress-induced activation of c-Jun N-terminal kinase (JNK) and c-Src. ROS was colocalized with the mitochondrial marker in stressed cells. Cyclosporin A blocked osmotic stress and DSS-induced ROS production, barrier dysfunction, TJ disruption and JNK activation. Mitochondria-targeted Mito-TEMPO blocked osmotic stress and DSS-induced barrier dysfunction and TJ disruption. Chronic restraint stress in mice resulted in the elevation of intracellular Ca2+, activation of JNK and c-Src, and disruption of TJ in the colonic epithelium. Furthermore, corticosterone administration induced JNK and c-Src activation, TJ disruption and protein thiol oxidation in colonic mucosa. The present study demonstrates that oxidative stress is a common signal in the mechanism of TJ disruption in the intestinal epithelium by different types of cellular stress in vitro and bio behavioral stress in vivo.


Asunto(s)
Calcio/metabolismo , Colon/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Estrés Psicológico/metabolismo , Uniones Estrechas/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Células CACO-2 , Canales de Calcio/genética , Canales de Calcio/metabolismo , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Quelantes/farmacología , Colon/citología , Colon/efectos de los fármacos , Corticosterona/farmacología , Ciclosporina/farmacología , Sulfato de Dextran/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Regulación de la Expresión Génica , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Mecanotransducción Celular , Ratones , Ratones Endogámicos C57BL , NG-Nitroarginina Metil Éster/farmacología , Presión Osmótica/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/agonistas , Estrés Mecánico , Estrés Psicológico/genética , Estrés Psicológico/fisiopatología , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/patología , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
9.
Biochim Biophys Acta ; 1860(4): 765-74, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26721332

RESUMEN

BACKGROUND: Disruption of epithelial tight junctions (TJ), gut barrier dysfunction and endotoxemia play crucial role in the pathogenesis of alcoholic tissue injury. Occludin, a transmembrane protein of TJ, is depleted in colon by alcohol. However, it is unknown whether occludin depletion influences alcoholic gut and liver injury. METHODS: Wild type (WT) and occludin deficient (Ocln(-/-)) mice were fed 1-6% ethanol in Lieber-DeCarli diet. Gut permeability was measured by vascular-to-luminal flux of FITC-inulin. Junctional integrity was analyzed by confocal microscopy. Liver injury was assessed by plasma transaminase, histopathology and triglyceride analyses. The effect of occludin depletion on acetaldehyde-induced TJ disruption was confirmed in Caco-2 cell monolayers. RESULTS: Ethanol feeding significantly reduced body weight gain in Ocln(-/-) mice. Ethanol increased inulin permeability in colon of both WT and Ocln(-/-) mice, but the effect was 4-fold higher in Ocln(-/-) mice. The gross morphology of colonic mucosa was unaltered, but ethanol disrupted the actin cytoskeleton, induced redistribution of occludin, ZO-1, E-cadherin and ß-catenin from the junctions and elevated TLR4, which was more severe in Ocln(-/-) mice. Occludin knockdown significantly enhanced acetaldehyde-induced TJ disruption and barrier dysfunction in Caco-2 cell monolayers. Ethanol significantly increased liver weight and plasma transaminase activity in Ocln(-/-) mice, but not in WT mice. Histological analysis indicated more severe lesions and fat deposition in the liver of ethanol-fed Ocln(-/-) mice. Ethanol-induced elevation of liver triglyceride was also higher in Ocln(-/-) mice. CONCLUSION: This study indicates that occludin deficiency increases susceptibility to ethanol-induced colonic mucosal barrier dysfunction and liver damage in mice.


Asunto(s)
Colon/metabolismo , Etanol/efectos adversos , Mucosa Intestinal/metabolismo , Hepatopatías/metabolismo , Ocludina/deficiencia , Uniones Estrechas/metabolismo , Animales , Células CACO-2 , Colon/patología , Etanol/farmacología , Humanos , Mucosa Intestinal/patología , Inulina/farmacocinética , Inulina/farmacología , Hígado/metabolismo , Hígado/patología , Hepatopatías/genética , Hepatopatías/patología , Ratones , Ratones Noqueados , Ocludina/metabolismo , Permeabilidad/efectos de los fármacos , Uniones Estrechas/genética , Triglicéridos/genética , Triglicéridos/metabolismo
10.
Phys Biol ; 14(2): 026001, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28248645

RESUMEN

Chronic alcoholism is known to alter the morphology of the hippocampus, an important region of cognitive function in the brain. Therefore, to understand the effect of chronic alcoholism on hippocampal neural cells, we employed a mouse model of chronic alcoholism and quantified intranuclear nanoscale structural alterations in these cells. Transmission electron microscopy (TEM) images of hippocampal neurons were obtained, and the degree of structural alteration in terms of mass density fluctuation was determined using the light-localization properties of optical media generated from TEM imaging. The results, which were obtained at length scales ranging from ~30 to 200 nm, show that 10-12 week-old mice fed a Lieber-DeCarli liquid (alcoholic) diet had a higher degree of structural alteration than control mice fed a normal diet without alcohol. The degree of structural alteration became significantly distinguishable at a sample length of ~100 nm, which is the typical length scale of the building blocks of cells, such as DNA, RNA, proteins and lipids. Interestingly, different degrees of structural alteration at such length scales suggest possible structural rearrangement of chromatin inside the nuclei in chronic alcoholism.


Asunto(s)
Alcoholismo/patología , Etanol/toxicidad , Hipocampo/patología , Microscopía Electrónica de Transmisión , Neuronas/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Femenino , Hipocampo/citología , Hipocampo/ultraestructura , Ratones , Ratones Endogámicos C57BL , Neuronas/ultraestructura
11.
Am J Physiol Gastrointest Liver Physiol ; 310(9): G705-15, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26822914

RESUMEN

The goals of this study were to evaluate the effects of ionizing radiation on apical junctions in colonic epithelium and mucosal barrier function in mice in vivo. Adult mice were subjected to total body irradiation (4 Gy) with or without N-acetyl-l-cysteine (NAC) feeding for 5 days before irradiation. At 2-24 h postirradiation, the integrity of colonic epithelial tight junctions (TJ), adherens junctions (AJ), and the actin cytoskeleton was assessed by immunofluorescence microscopy and immunoblot analysis of detergent-insoluble fractions for TJ and AJ proteins. The barrier function was evaluated by measuring vascular-to-luminal flux of fluorescein isothiocyanate (FITC)-inulin in vivo and luminal-to-mucosal flux in vitro. Oxidative stress was evaluated by measuring protein thiol oxidation. Confocal microscopy showed that radiation caused redistribution of occludin, zona occludens-1, claudin-3, E-cadherin, and ß-catenin, as well as the actin cytoskeleton as early as 2 h postirradiation, and this effect was sustained for at least 24 h. Feeding NAC before irradiation blocked radiation-induced disruption of TJ, AJ, and the actin cytoskeleton. Radiation increased mucosal permeability to inulin in colon, which was blocked by NAC feeding. The level of reduced-protein thiols in colon was depleted by radiation with a concomitant increase in the level of oxidized-protein thiol. NAC feeding blocked the radiation-induced protein thiol oxidation. These data demonstrate that radiation rapidly disrupts TJ, AJ, and the actin cytoskeleton by an oxidative stress-dependent mechanism that can be prevented by NAC feeding.


Asunto(s)
Colon/efectos de la radiación , Depuradores de Radicales Libres/uso terapéutico , Mucosa Intestinal/efectos de la radiación , Traumatismos por Radiación/prevención & control , Radiación Ionizante , Protectores contra Radiación/uso terapéutico , Uniones Estrechas/efectos de la radiación , Acetilcisteína/administración & dosificación , Acetilcisteína/farmacología , Acetilcisteína/uso terapéutico , Citoesqueleto de Actina/metabolismo , Animales , Colon/efectos de los fármacos , Colon/metabolismo , Suplementos Dietéticos , Femenino , Depuradores de Radicales Libres/administración & dosificación , Depuradores de Radicales Libres/farmacología , Absorción Intestinal , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Inulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Traumatismos por Radiación/tratamiento farmacológico , Protectores contra Radiación/administración & dosificación , Protectores contra Radiación/farmacología , Compuestos de Sulfhidrilo/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/metabolismo
12.
BMC Cancer ; 16: 189, 2016 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-26951793

RESUMEN

BACKGROUND: Alcohol consumption is one of the major risk factors for colorectal cancer. However, the mechanism involved in this effect of alcohol is unknown. METHODS: We evaluated the effect of chronic ethanol feeding on azoxymethane and dextran sulfate sodium (AOM/DSS)-induced carcinogenesis in mouse colon. Inflammation in colonic mucosa was assessed at a precancerous stage by evaluating mucosal infiltration of neutrophils and macrophages, and analysis of cytokine and chemokine gene expression. RESULTS: Chronic ethanol feeding significantly increased the number and size of polyps in colon of AOM/DSS treated mice. Confocal microscopic and immunoblot analyses showed a significant elevation of phospho-Smad, VEGF and HIF1α in the colonic mucosa. RT-PCR analysis at a precancerous stage indicated that ethanol significantly increases the expression of cytokines IL-1α, IL-6 and TNFα, and the chemokines CCL5/RANTES, CXCL9/MIG and CXCL10/IP-10 in the colonic mucosa of AOM/DSS treated mice. Confocal microscopy showed that ethanol feeding induces a dramatic elevation of myeloperoxidase, Gr1 and CD68-positive cells in the colonic mucosa of AOM/DSS-treated mice. Ethanol feeding enhanced AOM/DSS-induced suppression of tight junction protein expression and elevated cell proliferation marker, Ki-67 in the colonic epithelium. CONCLUSION: This study demonstrates that chronic ethanol feeding promotes colonic tumorigenesis potentially by enhancing inflammation and elevation of proinflammatory cytokines and chemokines.


Asunto(s)
Azoximetano/efectos adversos , Transformación Celular Neoplásica , Neoplasias del Colon/etiología , Sulfato de Dextran/efectos adversos , Etanol/administración & dosificación , Inflamación/complicaciones , Inflamación/etiología , Mucosa Intestinal/efectos de los fármacos , Animales , Biomarcadores de Tumor , Proliferación Celular , Quimiocinas/genética , Quimiocinas/metabolismo , Neoplasias del Colon/patología , Citocinas/genética , Citocinas/metabolismo , Femenino , Expresión Génica , Inflamación/metabolismo , Inflamación/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Transporte de Proteínas
13.
Funct Integr Genomics ; 15(6): 697-706, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26077032

RESUMEN

RNA interference (RNAi) has proved a powerful genetic tool for silencing genes in plants. Host-induced gene silencing of pathogen genes has provided a gene knockout strategy for a wide range of biotechnological applications. The RXLR effector Avr3a gene is largely responsible for virulence of oomycete plant pathogen Phytophthora infestans. In this study, we attempted to silence the Avr3a gene of P. infestans through RNAi technology. The P. infestans inoculation resulted in lower disease progression and a reduction in pathogen load, as demonstrated by disease scoring and quantification of pathogen biomass in terms of Pi08 repetitive elements, respectively. Transgenic plants induced moderate silencing of Avr3a, and the presence and/or expression of small interfering RNAs, as determined through Northern hybridization, indicated siRNA targeted against Avr3a conferred moderate resistance to P. infestans. The single effector gene did not provide complete resistance against P. infestans. Although the Avr3a effector gene could confer moderate resistance, for complete resistance, the cumulative effect of effector genes in addition to Avr3a needs to be considered. In this study, we demonstrated that host-induced RNAi is an effective strategy for functional genomics in oomycetes.


Asunto(s)
Resistencia a la Enfermedad/genética , Silenciador del Gen , Interacciones Huésped-Patógeno , Phytophthora infestans/genética , Solanum/inmunología , Factores de Virulencia/genética , Phytophthora infestans/patogenicidad , Solanum/genética , Solanum/microbiología
14.
Alcohol Clin Exp Res ; 39(8): 1465-75, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26173414

RESUMEN

BACKGROUND: Acetaldehyde, the toxic ethanol (EtOH) metabolite, disrupts intestinal epithelial barrier function. Aldehyde dehydrogenase (ALDH) detoxifies acetaldehyde into acetate. Subpopulations of Asians and Native Americans show polymorphism with loss-of-function mutations in ALDH2. We evaluated the effect of ALDH2 deficiency on EtOH-induced disruption of intestinal epithelial tight junctions and adherens junctions, gut barrier dysfunction, and liver injury. METHODS: Wild-type and ALDH2-deficient mice were fed EtOH (1 to 6%) in Lieber-DeCarli diet for 4 weeks. Gut permeability in vivo was measured by plasma-to-luminal flux of FITC-inulin, tight junction and adherens junction integrity was analyzed by confocal microscopy, and liver injury was assessed by the analysis of plasma transaminase activity, histopathology, and liver triglyceride. RESULTS: EtOH feeding elevated colonic mucosal acetaldehyde, which was significantly greater in ALDH2-deficient mice. ALDH2(-/-) mice showed a drastic reduction in the EtOH diet intake. Therefore, this study was continued only in wild-type and ALDH2(+/-) mice. EtOH feeding elevated mucosal inulin permeability in distal colon, but not in proximal colon, ileum, or jejunum of wild-type mice. In ALDH2(+/-) mice, EtOH-induced inulin permeability in distal colon was not only higher than that in wild-type mice, but inulin permeability was also elevated in the proximal colon, ileum, and jejunum. Greater inulin permeability in distal colon of ALDH2(+/-) mice was associated with a more severe redistribution of tight junction and adherens junction proteins from the intercellular junctions. In ALDH2(+/-) mice, but not in wild-type mice, EtOH feeding caused a loss of junctional distribution of tight junction and adherens junction proteins in the ileum. Histopathology, plasma transaminases, and liver triglyceride analyses showed that EtOH-induced liver damage was significantly greater in ALDH2(+/-) mice compared to wild-type mice. CONCLUSIONS: These data demonstrate that ALDH2 deficiency enhances EtOH-induced disruption of intestinal epithelial tight junctions, barrier dysfunction, and liver damage.


Asunto(s)
Aldehído Deshidrogenasa/deficiencia , Etanol/toxicidad , Hígado Graso/inducido químicamente , Hígado Graso/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Aldehído Deshidrogenasa Mitocondrial , Animales , Hígado Graso/patología , Femenino , Absorción Gastrointestinal/efectos de los fármacos , Absorción Gastrointestinal/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Uniones Estrechas/patología
15.
Alcohol Clin Exp Res ; 37(11): 1986-95, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23763370

RESUMEN

BACKGROUND: Fetal alcohol spectrum disorder (FASD) is characterized by neurodevelopmental anomalies manifesting in cognitive and behavioral deficits in the offspring with diverse severities. Social behavior is affected in FASD, and these deficits overlap with those of autism spectrum disorder (ASD). Identifying some of the molecular characteristics related to ASD in an animal model of FASD could ultimately provide details on the underlying molecular mechanisms of both disorders that could lead to novel treatments. METHODS: Pregnant Sprague-Dawley rats received the following diets: control (C; ad libitum standard laboratory chow), nutritional control pair-fed (PF), ethanol (EtOH), or an EtOH diet supplemented with 0.3, 1.5, or 7.5 mg thyroxine (T4)/l in the diet. Social behavior and memory were tested in the adult offspring. Plasma total T4, free T3 (fT3), and thyroid-stimulating hormone (TSH) levels were measured. Hippocampal expression of Gabrb3, Ube3a, Nr2b, Rasgrf1, and Dio3 were measured by RT-qPCR and protein levels of Mecp2 and Slc25a12 by Western blotting. RESULTS: Adult male offspring of EtOH dams showed elevated fT3 and low TSH levels. Adult male, but not female, offspring of EtOH dams exhibited social behavior and memory deficits. Expression of autism candidates, Gabrb3, Ube3a, Mecp2, and Slc25a12, was significantly increased in the hippocampus of male offspring of EtOH dams. Hippocampal Nr2b and Dio3 were also increased, while Rasgrf1 was decreased in the same population. Peripheral thyroid function, social behavioral deficits, and altered expression of the above genes were normalized by simultaneous administration of 0.3 mg/l T4 in the EtOH diet. CONCLUSIONS: Our data suggest that social interaction deficits of FASD share molecular mechanism with ASD by showing altered hippocampal expression of several ASD candidate genes. Social interaction deficits as well as the gene expression changes in the offspring of EtOH-consuming dams can be reversed by low dose of thyroid hormone supplementation to the mothers.


Asunto(s)
Trastorno Autístico/inducido químicamente , Etanol/efectos adversos , Trastornos del Espectro Alcohólico Fetal/prevención & control , Hipocampo/efectos de los fármacos , Tiroxina/administración & dosificación , Animales , Trastorno Autístico/prevención & control , Femenino , Hipocampo/metabolismo , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Sprague-Dawley , Conducta Social
16.
Cells ; 12(2)2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36672262

RESUMEN

Fetal alcohol spectrum disorders (FASDs) are associated with systemic inflammation and neurodevelopmental abnormalities. Several candidate genes were found to be associated with fetal alcohol exposure (FAE)-associated behaviors, but a sex-specific complete transcriptomic analysis was not performed at the adult stage. Recent studies have shown that they are regulated at the developmental stage. However, the sex-specific role of RNA in FAE offspring brain development and function has not been studied yet. Here, we carried out the first systematic RNA profiling by utilizing a high-throughput transcriptomic (RNA-seq) approach in response to FAE in the brain cortex of male and female offspring at adulthood (P60). Our RNA-seq data analysis suggests that the changes in RNA expression in response to FAE are marked sex-specific. We show that the genes Muc3a, Pttg1, Rec8, Clcnka, Capn11, and pnp2 exhibit significantly higher expression in the male offspring than in the female offspring at P60. FAE female mouse brain sequencing data also show an increased expression of Eno1, Tpm3, and Pcdhb2 compared to male offspring. We performed a pathway analysis using a commercial software package (Ingenuity Pathway Analysis). We found that the sex-specific top regulator genes (Rictor, Gaba, Fmri, Mlxipl) are highly associated with eIF2 (translation initiation), synaptogenesis (the formation of synapses between neurons in the nervous system), sirtuin (metabolic regulation), and estrogen receptor (involved in obesity, aging, and cancer) signaling. Taken together, our transcriptomic results demonstrate that FAE differentially alters RNA expression in the adult brain in a sex-specific manner.


Asunto(s)
Etanol , Trastornos del Espectro Alcohólico Fetal , Embarazo , Animales , Ratones , Humanos , Masculino , Femenino , Etanol/metabolismo , Perfilación de la Expresión Génica , Trastornos del Espectro Alcohólico Fetal/genética , Corteza Cerebral/metabolismo , Factores de Transcripción/metabolismo , ARN
17.
Front Immunol ; 14: 1093584, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36817471

RESUMEN

Introduction: Chronic stress is co-morbid with alcohol use disorder that feedback on one another, thus impeding recovery from both disorders. Stress and the stress hormone corticosterone aggravate alcohol-induced intestinal permeability and liver damage. However, the mechanisms involved in compounding tissue injury by stress/corticosterone and alcohol are poorly defined. Here we explored the involvement of the TRPV6 channel in stress (or corticosterone) 3and alcohol-induced intestinal epithelial permeability, microbiota dysbiosis, and systemic inflammation. Methods: Chronic alcohol feeding was performed on adult wild-type and Trpv6-/- mice with or without corticosterone treatment or chronic restraint stress (CRS). The barrier function was determined by evaluating inulin permeability in vivo and assessing tight junction (TJ) and adherens junction (AJ) integrity by immunofluorescence microscopy. The gut microbiota composition was evaluated by 16S rRNA sequencing and metagenomic analyses. Systemic responses were assessed by evaluating endotoxemia, systemic inflammation, and liver damage. Results: Corticosterone and CRS disrupted TJ and AJ, increased intestinal mucosal permeability, and caused endotoxemia, systemic inflammation, and liver damage in wild-type but not Trpv6-/- mice. Corticosterone and CRS synergistically potentiated the alcohol-induced breakdown of intestinal epithelial junctions, mucosal barrier impairment, endotoxemia, systemic inflammation, and liver damage in wild-type but not Trpv6-/- mice. TRPV6 deficiency also blocked the effects of CRS and CRS-mediated potentiation of alcohol-induced dysbiosis of gut microbiota. Conclusions: These findings indicate an essential role of TRPV6 in stress, corticosterone, and alcohol-induced intestinal permeability, microbiota dysbiosis, endotoxemia, systemic inflammation, and liver injury. This study identifies TRPV6 as a potential therapeutic target for developing treatment strategies for stress and alcohol-associated comorbidity.


Asunto(s)
Endotoxemia , Hepatopatías , Ratones , Animales , Corticosterona/metabolismo , Endotoxemia/metabolismo , Disbiosis/metabolismo , ARN Ribosómico 16S , Mucosa Intestinal/metabolismo , Etanol/farmacología , Hepatopatías/metabolismo , Inflamación/metabolismo , Canales de Calcio/metabolismo , Canales Catiónicos TRPV/metabolismo
18.
Front Immunol ; 14: 1174140, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37638013

RESUMEN

Introduction: The mechanism underlying radiation-induced gut microbiota dysbiosis is undefined. This study examined the effect of radiation on the intestinal Paneth cell α-defensin expression and its impact on microbiota composition and mucosal tissue injury and evaluated the radio-mitigative effect of human α-defensin 5 (HD5). Methods: Adult mice were subjected to total body irradiation, and Paneth cell α-defensin expression was evaluated by measuring α-defensin mRNA by RT-PCR and α-defensin peptide levels by mass spectrometry. Vascular-to-luminal flux of FITC-inulin was measured to evaluate intestinal mucosal permeability and endotoxemia by measuring plasma lipopolysaccharide. HD5 was administered in a liquid diet 24 hours before or after irradiation. Gut microbiota was analyzed by 16S rRNA sequencing. Intestinal epithelial junctions were analyzed by immunofluorescence confocal microscopy and mucosal inflammatory response by cytokine expression. Systemic inflammation was evaluated by measuring plasma cytokine levels. Results: Ionizing radiation reduced the Paneth cell α-defensin expression and depleted α-defensin peptides in the intestinal lumen. α-Defensin down-regulation was associated with the time-dependent alteration of gut microbiota composition, increased gut permeability, and endotoxemia. Administration of human α-defensin 5 (HD5) in the diet 24 hours before irradiation (prophylactic) significantly blocked radiation-induced gut microbiota dysbiosis, disruption of intestinal epithelial tight junction and adherens junction, mucosal barrier dysfunction, and mucosal inflammatory response. HD5, administered 24 hours after irradiation (treatment), reversed radiation-induced microbiota dysbiosis, tight junction and adherens junction disruption, and barrier dysfunction. Furthermore, HD5 treatment also prevents and reverses radiation-induced endotoxemia and systemic inflammation. Conclusion: These data demonstrate that radiation induces Paneth cell dysfunction in the intestine, and HD5 feeding prevents and mitigates radiation-induced intestinal mucosal injury, endotoxemia, and systemic inflammation.


Asunto(s)
Endotoxemia , Traumatismos por Radiación , alfa-Defensinas , Humanos , Adulto , Animales , Ratones , Células de Paneth , Disbiosis , Endotoxemia/etiología , ARN Ribosómico 16S , Traumatismos por Radiación/etiología , Citocinas , Inflamación
19.
FASEB J ; 25(7): 2313-24, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21429942

RESUMEN

Prenatal exposure to alcohol is thought to be the most prevalent nongenetic cause of a wide range of neurodevelopmental deficits. Insufficient thyroid hormone levels are one mechanism that hampers development of the alcohol-exposed brain, and we hypothesized that altered dosage of the imprinted thyroid hormone-inactivating gene deiodinase-III (Dio3) is responsible. To follow parent-of-origin allelic expression of Dio3 in the fetal and adult offspring of alcohol-consuming and control dams, we reciprocally crossed 2 polymorphic rat strains. In the frontal cortex, prenatal alcohol exposure altered imprinting patterns and total expression of Dio3 in the fetus and produced a permanent hypothyroid milieu in the adult. In the hippocampus, alcohol affected the paternal and total expression of Dio3 in the fetus and in the adult male, where thyroid hormone levels were concomitantly increased. Hippocampus-dependent behavioral deficits were identified exclusively in males, suggesting they are dependent on aberrant allelic Dio3 expression. None of these effects were observed in offspring of the reciprocal cross. Thus, genetic background and sex modify vulnerability to prenatal alcohol via brain region-specific expression of Dio3. This finding implies that phenotypic heterogeneity in human fetal alcohol spectrum disorder can be linked to genetic vulnerability in affected brain regions.


Asunto(s)
Etanol/farmacología , Trastornos del Espectro Alcohólico Fetal/genética , Hipocampo/metabolismo , Yoduro Peroxidasa/genética , Efectos Tardíos de la Exposición Prenatal/genética , Útero/metabolismo , Animales , Western Blotting , Depresores del Sistema Nervioso Central/farmacología , Cruzamientos Genéticos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Yoduro Peroxidasa/metabolismo , Masculino , Embarazo , Ratas , Ratas Endogámicas BN , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores Sexuales , Especificidad de la Especie , Triyodotironina/metabolismo
20.
J Biophotonics ; 15(3): e202100247, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34786860

RESUMEN

Molecular specific photonics localization method, the inverse participation ratio (IPR) technique, is a powerful procedure to probe the nano- to submicron scales structural alterations in cells/tissues in their abnormalities due to chronic alcoholism using confocal imaging. Chronic alcoholism introduces abnormalities in brain cells/tissue at the nanoscale level that results in behavioural and psychological disorders which are not well understood. On the other hand, probiotics such as Lactobacillus plantarum enhances brain functions in chronic alcoholism. Using the IPR technique, we probe the molecular specific spatial structural alterations in glial brain cells astrocytes and microglia, as well as in chromatins in the nuclei of cortex brain cells, with or without probiotic treatments in chronic alcoholism. The results show chronic alcoholism alone harms brain cells and the probiotic treatment in chronic alcoholism reverses alcoholic damage in the brain cells/tissues toward normalcy.


Asunto(s)
Alcoholismo , Lactobacillus plantarum , Probióticos , Alcoholismo/diagnóstico por imagen , Alcoholismo/terapia , Encéfalo/diagnóstico por imagen , Humanos , Óptica y Fotónica , Probióticos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA