Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Vet Immunol Immunopathol ; 255: 110533, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36563567

RESUMEN

The aim of this study was to evaluate the potential of micronutrients and feed additives to modulate intestinal microbiota and systemic and mucosal immune responses in weaned pigs infected with Salmonella. At weaning, 32 litters of 12 piglets each were allocated to four dietary treatments: 1) control diet (CTRL), 2) CTRL supplemented with chlortetracycline (ATB), 3) CTRL supplemented with a cocktail of feed additives (CKTL); and 4) CKTL diet containing bovine colostrum in replacement of spray-dry animal plasma (CKTL+COL). The CKTL supplement included cranberry extract, encapsulated carvacrol and yeast-derived products and an enriched selenium and vitamin premix. Three weeks after weaning, four pigs per litter were orally inoculated with Salmonella Typhimurium DT104. Half of them were euthanized 3 days post-infection (dpi) and the other half, 7 dpi. The expression of IL6, TNF, IL8, monocyte chemoattractant protein 1 (MCP1), IFNG, cyclooxygenase 2 (COX2), glutathione peroxidase 2 (GPX2) and ß-defensin 2 (DEFB2) showed a peaked response at 3 dpi (P < 0.05). Results also revealed that DEFB2 expression was higher at 3 dpi in CTRL and CKTL groups than in ATB (P = 0.01 and 0.06, respectively) while GPX2 gene was markedly increased at 3 and 7 dpi in pigs fed CKTL or CKTL+COL diet compared to CTRL pigs (P < 0.05). In piglets fed CKTL or CKTL+COL diet, intestinal changes in microbial communities were less pronounced after exposure to Salmonella compared to CTRL and progressed faster toward the status before Salmonella challenge (AMOVA P < 0.01). Furthermore, the relative abundance of several families was either up- or down-regulated in pigs fed CKTL or CKTL+COL diet after Salmonella challenge. In conclusion, weaning diet enriched with bovine colostrum, vitamins and mixture of feed additives mitigated the influence of Salmonella infection on intestinal microbial populations and modulate systemic and intestinal immune defences.


Asunto(s)
Suplementos Dietéticos , Microbiota , Animales , Porcinos , Bovinos , Destete , Dieta/veterinaria , Salmonella typhimurium , Inmunidad , Alimentación Animal/análisis
2.
J Nutr ; 150(10): 2673-2686, 2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-32886125

RESUMEN

BACKGROUND: Recent meta-analyses suggest that the consumption of fermented dairy products reduces type 2 diabetes and cardiovascular disease (CVD) risk, although the underlying mechanisms remain unclear. OBJECTIVE: We evaluated whether dairy protein products modulated gut microbiota and cardiometabolic features in mouse models of diet-induced obesity and CVD. METHODS: Eight-week-old C57BL/6J wild-type (WT) and LDLr-/-ApoB100/100 (LRKO) male mice were fed for 12 and 24 wk, respectively, with a high-fat/high-sucrose diet [66% kcal lipids, 22% kcal carbohydrates (100% sucrose), 12% kcal proteins]. The protein sources of the 4 diets were 100% nondairy protein (NDP), or 50% of the NDP energy replaced by milk (MP), milk fermented by Lactobacillus helveticus (FMP), or Greek-style yogurt (YP) protein. Fecal 16S rRNA gene-based amplicon sequencing, intestinal gene expression, and glucose tolerance test were conducted. Hepatic inflammation and circulating adhesion molecules were measured by multiplex assays. RESULTS: Feeding WT mice for 12 wk led to a 74% increase in body weight, whereas after 24 wk the LRKO mice had a 101.5% increase compared with initial body weight. Compared with NDP and MP, the consumption of FMP and YP modulated the gut microbiota composition in a similar clustering pattern, upregulating the Streptococcus genus in both genotypes. In WT mice, feeding YP compared with NDP increased the expression of genes involved in jejunal (Reg3b, 7.3-fold, P = 0.049) and ileal (Ocln, 1.7-fold, P = 0.047; Il1-ß,1.7-fold, P = 0.038; Nos2, 3.8-fold, P = 0.018) immunity and integrity. In LRKO mice, feeding YP compared with MP improved insulin sensitivity by 65% (P = 0.039). In LRKO mice, feeding with FMP versus NDP attenuated hepatic inflammation (monocyte chemoattractant protein 1, 2.1-fold, P ˂ 0.0001; IL1-ß, 5.7-fold, P = 0.0003; INF-γ, 1.7-fold, P = 0.002) whereas both FMP [vascular adhesion molecule 1 (VCAM1), 1.3-fold, P = 0.0003] and YP (VCAM1, 1.04-fold, P = 0.013; intracellular adhesion molecule 1, 1.4-fold, P = 0.028) decreased circulating adhesion molecules. CONCLUSION: Both fermented dairy protein products reduce cardiometabolic risk factors in diet-induced obese mice, possibly by modulating the gut microbiota.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Productos Lácteos Cultivados/análisis , Microbioma Gastrointestinal/efectos de los fármacos , Enfermedades Metabólicas/prevención & control , Proteínas de la Leche/farmacología , Obesidad/inducido químicamente , Animales , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Bacterias/clasificación , Bacterias/efectos de los fármacos , Biomarcadores/sangre , Dieta , Dieta Alta en Grasa , Sacarosa en la Dieta/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Leche/química , Proteínas de la Leche/química , Receptores de LDL/genética , Receptores de LDL/metabolismo
3.
Atherosclerosis ; 304: 9-21, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32563005

RESUMEN

BACKGROUND AND AIMS: Poor dietary habits contribute to the obesity pandemic and related cardiovascular diseases but the respective impact of high saturated fat versus added sugar consumption remains debated. Herein, we aimed to disentangle the individual role of dietary fat versus sugar in cardiometabolic disease progression. METHODS: We fed pro-atherogenic LDLr-/-ApoB100/100 mice either a low-fat/high-sucrose (LFHS) or a high-fat/low-sucrose (HFLS) diet for 24 weeks. Weekly body weight gain was registered. 16S rRNA gene-based gut microbial analysis was performed to investigate gut microbial modulations. Intraperitoneal insulin (ipITT) and oral glucose tolerance test (oGTT) were conducted to assess glucose homeostasis and insulin sensitivity. Cytokines were assessed in fasted plasma, epididymal white adipose tissue and liver lysates. Heart function was evaluated by echocardiography. Aortic atheroma lesions were quantified according to the en face technique. RESULTS: HFLS feeding increased obesity, insulin resistance and dyslipidemia compared to LFHS feeding. Conversely, high sucrose consumption decreased gut microbial diversity while augmenting inflammation and the adaptative immune defense against metabolic endotoxemia and reduced macrophage cholesterol efflux capacity. This led to more severe cardiovascular complications as revealed by remarkably high level of atherosclerotic lesions and the early development of cardiac dysfunction in LFHS vs HFLS fed mice. CONCLUSIONS: We uncoupled obesity-associated insulin resistance from cardiovascular diseases and provided novel evidence that dietary sucrose, not fat, is the main driver of metabolic inflammation accelerating severe atherosclerosis in hyperlipidemic mice.


Asunto(s)
Aterosclerosis , Enfermedades Cardiovasculares , Sacarosa en la Dieta/efectos adversos , Inflamación , Resistencia a la Insulina , Animales , Apolipoproteína B-100 , Dieta Alta en Grasa , Grasas de la Dieta/efectos adversos , Microbioma Gastrointestinal , Hiperlipidemias , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Ribosómico 16S
4.
Vet Immunol Immunopathol ; 226: 110072, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32540688

RESUMEN

Immune system development of piglets is influenced by birth weight and colostrum and milk intake. Moreover, the dam transfer to piglets of vitamins A and D and copper, which play important role in immunity, is limited during lactation. In this study, we evaluated the potential of maternal and neonatal supplementations with vitamins A and D and copper, with or without neonatal supplementation of bovine colostrum (BC), to modulate the immune system development of low birth weight (LBW) and high birth weight (HBW) piglets during the peri-weaning period. Litters from 23 control sows (CONT) were assigned to one of the following treatments: 1) control (C); 2) oral administration at 2 and 8 days (d) of age of retinol-acetate, 25-hydroxyvitamin D and CuSO4 and exposure to UVB light for 15 min every second day from d 5 to d 21 (ADCu); 3) oral administration of dehydrated BC (4 g/d) from d 5 to d 10 (BC); 4) ADCu + BC. This experimental design was repeated with 24 sows fed extra daily supplements of 25-hydroxyvitamin D (4,000 IU), ß-carotene (30,000 IU) and Cu-yeast (equivalent 45 mg of Cu) from 90 d of gestation until weaning at d 21 (SUPPL). Within each litter, 2 LBW and 2 HBW piglets were euthanized at d 16 and d 23 in order to characterize leukocyte subsets in mesenteric lymph nodes (MLN) and blood by flow cytometry, and to measure gene expression in the MLN and jejunal mucosa by qPCR. At d 16, results revealed that the percentages of γδ and cytotoxic T lymphocytes were significantly reduced in LBW compared to HBW piglets. The jejunal expression of interleukin (IL) 22 was also up-regulated, along with MLN expression of C-C Motif Chemokine Ligand 23, bone morphogenetic protein 2 and secreted phosphoprotein 1 (SPP1), whereas jejunal expression of tumor necrosis factor α was decreased in LBW piglets. At d 23, LBW piglets showed lower amounts of γδ T lymphocytes, higher percentages of CD3- and CD3-CD8α+CD16+ leukocytes (which include Natural killer cells) and lower jejunal expression of IL18. Furthermore, supplementation with BC increased the blood percentage of CD3-CD16+ leukocytes and reduced jejunal IL5 and MLN IL15 expression whereas supplementation with ADCu + BC increased jejunal TNF superfamily 13B and MLN SPP1 expression. Our results suggest that immune system development after birth differed between LBW and HBW piglets and that early dietary supplementation with BC and ADCu has the potential to modulate development of immune functions.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales/inmunología , Animales Lactantes/inmunología , Peso al Nacer , Calostro/inmunología , Micronutrientes/administración & dosificación , Porcinos/inmunología , Alimentación Animal , Animales , Bovinos , Enfermedades de los Bovinos/prevención & control , Citocinas/genética , Citocinas/inmunología , Suplementos Dietéticos/análisis , Femenino , Inmunidad , Destete
5.
Vet Immunol Immunopathol ; 206: 25-34, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30502909

RESUMEN

The aim of this study was to investigate the effect of the piglet growth during the first week of life on ileal expression of genes and on development of the immune system. Eight litters adjusted to 12 piglets were used. Within each litter, the piglet that showed the lowest weight gain (LWG; n = 8) and the one that showed the highest weight gain (HWG; n = 8) in their first week of life were enrolled. Peripheral blood mononuclear cells (PBMC) were isolated on days 8 and 16 to characterize cellular population profiles and to assess ex-vivo secretion of interleukin-10 (IL-10), IL-6 and tumor necrosis factor-α (TNF-α). On day 16, piglets were euthanized and ileum samples were collected to extract RNA for microarray analysis and gene expression by qPCR. As expected, growth performance of LWG piglet was impaired compared to HWG piglets (P < 0.05). From day 8 to 16, the percentage of CD21+ B cells significantly increased in blood of heavier HWG piglets while the percentage remained constant in smaller LWG piglets (P weight x day = 0.01). For the CD4+CD8α- Th cells, a marked increase was observed in LWG piglets from 8 to 16 days of age (P = 0.002) whereas no significant change occurred in HWG piglets. Percentages of CD14+ monocytes and other MHC-II+ cells were respectively higher and lower on day 8 compared to day 16 for both groups of piglets (P < 0.01). On day 8, LPS-activated PBMC from LWG piglets produced less IL-6 compared to HWG piglets (P < 0.05). Microarray analysis of gene expression in piglets' ileum tissue indicated that several genes involed in defense response and response to oxidative stress were modulated differently in LWG compared to HWG. Gene analysis by Q-PCR confirmed microarray results and revealed that IL-10, SOD1, NOS2, NOD2, TLR4, TLR9, CD40 and CD74 expressions were significantly decreased (P < 0.05) in LWG in comparison to HWG piglets, while MYD88 and NFkBiA showed a tendency to decrease (0.05 ≤ P < 0.07). These results suggest that birth weight and milk intake affect the growth performances and the development of immunity by modulating the expression of genes associated with immunity and oxidative stress in piglets' intestinal tissue, and by affecting the leukocyte populations involved in innate and cell-mediated immunity in nursing piglets. Therefore, impaired development of immune system in LWG piglets might have an impact on their resistance to infections later in life.


Asunto(s)
Íleon/inmunología , Sistema Inmunológico/crecimiento & desarrollo , Lactancia , Porcinos/inmunología , Aumento de Peso/inmunología , Animales , Citocinas/sangre , Ensayo de Inmunoadsorción Enzimática/veterinaria , Femenino , Expresión Génica , Íleon/anatomía & histología , Íleon/crecimiento & desarrollo , Leucocitos Mononucleares/inmunología , Análisis por Micromatrices/veterinaria , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Porcinos/crecimiento & desarrollo
6.
Am J Physiol Gastrointest Liver Physiol ; 306(7): G594-605, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24525021

RESUMEN

Histone deacetylases (Hdac) remove acetyl groups from proteins, influencing global and specific gene expression. Hdacs control inflammation, as shown by Hdac inhibitor-dependent protection from dextran sulfate sodium (DSS)-induced murine colitis. Although tissue-specific Hdac knockouts show redundant and specific functions, little is known of their intestinal epithelial cell (IEC) role. We have shown previously that dual Hdac1/Hdac2 IEC-specific loss disrupts cell proliferation and determination, with decreased secretory cell numbers and altered barrier function. We thus investigated how compound Hdac1/Hdac2 or Hdac2 IEC-specific deficiency alters the inflammatory response. Floxed Hdac1 and Hdac2 and villin-Cre mice were interbred. Compound Hdac1/Hdac2 IEC-deficient mice showed chronic basal inflammation, with increased basal disease activity index (DAI) and deregulated Reg gene colonic expression. DSS-treated dual Hdac1/Hdac2 IEC-deficient mice displayed increased DAI, histological score, intestinal permeability, and inflammatory gene expression. In contrast to double knockouts, Hdac2 IEC-specific loss did not affect IEC determination and growth, nor result in chronic inflammation. However, Hdac2 disruption protected against DSS colitis, as shown by decreased DAI, intestinal permeability and caspase-3 cleavage. Hdac2 IEC-specific deficient mice displayed increased expression of IEC gene subsets, such as colonic antimicrobial Reg3b and Reg3g mRNAs, and decreased expression of immune cell function-related genes. Our data show that Hdac1 and Hdac2 are essential IEC homeostasis regulators. IEC-specific Hdac1 and Hdac2 may act as epigenetic sensors and transmitters of environmental cues and regulate IEC-mediated mucosal homeostatic and inflammatory responses. Different levels of IEC Hdac activity may lead to positive or negative outcomes on intestinal homeostasis during inflammation.


Asunto(s)
Colitis/enzimología , Colon/enzimología , Células Epiteliales/enzimología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Mucosa Intestinal/enzimología , Animales , Colitis/genética , Colitis/inmunología , Colitis/patología , Colon/inmunología , Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Epigénesis Genética , Células Epiteliales/inmunología , Células Epiteliales/patología , Regulación de la Expresión Génica , Genotipo , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/deficiencia , Histona Desacetilasa 2/genética , Homeostasis , Inmunidad Mucosa , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Permeabilidad , Fenotipo , Factores de Tiempo
7.
PLoS One ; 8(9): e73785, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24040068

RESUMEN

Acetylation and deacetylation of histones and other proteins depends on histone acetyltransferases and histone deacetylases (HDACs) activities, leading to either positive or negative gene expression. HDAC inhibitors have uncovered a role for HDACs in proliferation, apoptosis and inflammation. However, little is known of the roles of specific HDACs in intestinal epithelial cells (IEC). We investigated the consequences of ablating both HDAC1 and HDAC2 in murine IECs. Floxed Hdac1 and Hdac2 homozygous mice were crossed with villin-Cre mice. Mice deficient in both IEC HDAC1 and HDAC2 weighed less and survived more than a year. Colon and small intestinal sections were stained with hematoxylin and eosin, or with Alcian blue and Periodic Acid Schiff for goblet cell identification. Tissue sections from mice injected with BrdU for 2 h, 14 h and 48 h were stained with anti-BrdU. To determine intestinal permeability, 4-kDa FITC-labeled dextran was given by gavage for 3 h. Microarray analysis was performed on total colon RNAs. Inflammatory and IEC-specific gene expression was assessed by Western blot or semi-quantitative RT-PCR and qPCR with respectively total colon protein and total colon RNAs. HDAC1 and HDAC2-deficient mice displayed: 1) increased migration and proliferation, with elevated cyclin D1 expression and phosphorylated S6 ribosomal protein, a downstream mTOR target; 2) tissue architecture defects with cell differentiation alterations, correlating with reduction of secretory Paneth and goblet cells in jejunum and goblet cells in colon, increased expression of enterocytic markers such as sucrase-isomaltase in the colon, increased expression of cleaved Notch1 and augmented intestinal permeability; 3) loss of tissue homeostasis, as evidenced by modifications of claudin 3 expression, caspase-3 cleavage and Stat3 phosphorylation; 4) chronic inflammation, as determined by inflammatory molecular expression signatures and altered inflammatory gene expression. Thus, epithelial HDAC1 and HDAC2 restrain the intestinal inflammatory response, by regulating intestinal epithelial cell proliferation and differentiation.


Asunto(s)
Diferenciación Celular/genética , Células Epiteliales/metabolismo , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Mucosa Intestinal/metabolismo , Animales , Western Blotting , Peso Corporal/genética , Movimiento Celular/genética , Proliferación Celular , Colon/metabolismo , Colon/patología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Inflamación/genética , Intestinos/patología , Intestinos/fisiopatología , Ratones , Ratones Noqueados , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Tamaño de los Órganos/genética , Permeabilidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcriptoma
8.
J Cell Biochem ; 114(5): 1203-15, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23192652

RESUMEN

Polycomb-group proteins form multimeric protein complexes involved in transcriptional silencing. The Polycomb Repressive complex 2 (PRC2) contains the Suppressor of Zeste-12 protein (Suz12) and the histone methyltransferase Enhancer of Zeste protein-2 (Ezh2). This complex, catalyzing the di- and tri-methylation of histone H3 lysine 27, is essential for embryonic development and stem cell renewal. However, the role of Polycomb-group protein complexes in the control of the intestinal epithelial cell (IEC) phenotype is not known. We show that Suz12 and Ezh2 were differentially expressed along the intestinal crypt-villus axis. ShRNA-mediated Suz12 depletion in the IEC-6 rat crypt-derived cell line decreased Ezh2 expression and H3K27 di-trimethylation. Suz12-depleted cells achieved higher cell densities after confluence, with increased cyclin D2 and cyclin D3 protein levels, and increased STAT3 activation in post-confluent cells. Suz12 depletion specifically increased mostly developmental, cell adhesion and immune response gene expression, including neuronal and inflammatory genes. Suz12 depletion directly and indirectly de-regulated the IL-1ß-dependent inflammatory response, as demonstrated by decreased MAPK p38 activation as opposed to JNK activation, and altered basal and stimulated expression of inflammatory genes, including transcription factors such as C/EBPß. Of note, this positive effect on cell proliferation and inflammatory gene expression was revealed in the absence of the cyclin-dependent kinase inhibitor p16, a main target negatively regulated by PRC2. These results demonstrate that the PRC2 complex, in addition to keeping in check non-IEC differentiation pathways, insures the proper IEC response to cell density as well as to external growth and inflammatory signals, by controlling specific signaling pathways.


Asunto(s)
Células Epiteliales/metabolismo , Células Epiteliales/patología , Histonas/metabolismo , Inflamación/patología , Intestinos/patología , Lisina/metabolismo , Animales , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Proteína Potenciadora del Homólogo Zeste 2 , Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/genética , Interleucina-1beta/farmacología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Metilación/efectos de los fármacos , Ratones , Microvellosidades/efectos de los fármacos , Microvellosidades/metabolismo , Microvellosidades/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Complejo Represivo Polycomb 2/metabolismo , Ratas , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , beta Catenina/metabolismo
9.
J Cell Biochem ; 103(5): 1573-83, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17910034

RESUMEN

The C/EBPdelta transcription factor is involved in the positive regulation of the intestinal epithelial cell acute phase response. C/EBPdelta regulation by histone deacetylases (HDACs) during the course of inflammation remains to be determined. Our aim was to examine the effect of HDACs on C/EBPdelta-dependent regulation of haptoglobin, an acute phase protein induced in intestinal epithelial cells in response to pro-inflammatory cytokines. HDAC1, HDAC3, and HDAC4 were expressed in intestinal epithelial cells, as determined by Western blot. GST pull-down assays showed specific HDAC1 interactions with the transcriptional activation and the b-ZIP C/EBPdelta domains, while the co-repressor mSin3A interacts with the C-terminal domain. Immunoprecipitation assays confirmed the interaction between HDAC1 and the N-terminal C/EBPdelta amino acid 36-164 domain. HDAC1 overexpression decreased C/EBPdelta transcriptional activity of the haptoglobin promoter, as assessed by transient transfection and luciferase assays. Chromatin immunoprecipitation analysis showed a displacement of HDAC1 from the haptoglobin promoter in response to inflammatory stimuli and an increased acetylation of histone H3 and H4. HDAC1 silencing by shRNA expression increased both basal and IL-1beta-induced haptoglobin mRNA levels in epithelial intestinal cells. Our results suggest that interactions between C/EBPs and HDAC1 negatively regulate C/EBPdelta-dependent haptoglobin expression in intestinal epithelial cells.


Asunto(s)
Reacción de Fase Aguda/metabolismo , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Haptoglobinas/biosíntesis , Histona Desacetilasas/metabolismo , Mucosa Intestinal/metabolismo , Activación Transcripcional , Acetilación/efectos de los fármacos , Animales , Proteína delta de Unión al Potenciador CCAAT/antagonistas & inhibidores , Células CACO-2 , Inhibidores de Histona Desacetilasas , Histonas/metabolismo , Humanos , Interleucina-1beta/farmacología , Regiones Promotoras Genéticas , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , ARN Interferente Pequeño , Ratas , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos
10.
DNA Cell Biol ; 26(3): 133-47, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17417942

RESUMEN

Butyrate (NaBu), a product of intestinal microbial metabolism, has been proposed as an anti-inflammatory agent for treating inflammatory bowel diseases. However, the molecular mechanisms implicated in the modulation of intestinal epithelial cell inflammatory response to NaBu remain unknown. Here, microarray analysis performed on nontransformed human crypt intestinal epithelial cells (HIEC) shows that NaBu regulated specifically the short-term IL-1beta -dependent induction of different inflammatory genes. While NaBu significantly increased the IL-1beta -induction of genes like SAA2, C3, and IL-1alpha , other inflammatory genes like CXCL5, CXCL11, and IL-1beta were decreased. Induction of various genes such as CXCL8, CCL20, and IL-6 was unaffected by NaBu. We show that, compared to genes that are upregulated or downregulated by NaBu, genes that are unaffected by NaBu were induced more rapidly after IL-1beta treatment and contained a higher concentration of transcription factor binding sites in their promoter region. In addition, transient treatment with IL-1beta was sufficient for subsequent induction of NaBu-upregulated and NaBu-unaffected classes of genes, while a continuous presence of IL-1beta was required for NaBu-downregulated gene expression. In conclusion, our results suggest that fundamental differences predispose inflammatory genes to specific regulation by NaBu in intestinal epithelial cells, thereby allowing precise control of inflammation.


Asunto(s)
Butiratos/farmacología , Regulación de la Expresión Génica , Expresión Génica/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Interleucina-1beta/farmacología , Mucosa Intestinal/efectos de los fármacos , Sitios de Unión , Células Cultivadas , Humanos , Interleucina-8/genética , Mucosa Intestinal/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Proteína Amiloide A Sérica/genética , Factores de Transcripción/metabolismo
11.
Am J Physiol Gastrointest Liver Physiol ; 290(2): G310-8, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16239403

RESUMEN

CDX2, a member of the caudal family of transcription factors, is involved in enterocyte lineage specification. CDX2 activates many intestine-specific genes, such as sucrase-isomaltase and lactase-phlorizin hydrolase (LPH), and adhesion proteins, namely, LI-cadherin and claudin-2. In this study, we show that the proprotein convertase furin, involved in proteolytic maturation of proprotein substrates including LPH and cell surface proteins, is a CDX2 target. Indeed, expression of the rat furin homolog was induced 1.5-fold, as determined by microarray experiments that compared control with CDX2-expressing intestinal epithelial cells (IEC-6). As determined by transient transfection assays in Caco-2/15 cells, the furin P1 promoter 1.3-kb fragment between SacI and NheI was essential for CDX2 transcriptional activation. Electrophoretic mobility shift/supershift assays followed by site-specific mutagenesis and chromatin immunoprecipitation identified the CDX DNA-binding site (CBS)2 sequence from nt -1827 to -1821 as the major CBS involved in furin P1 promoter activation. Increased furin mRNA and protein expression correlated with both CDX2 expression and intestinal epithelial cell differentiation. In addition, furin mRNAs were detected predominantly in differentiated epithelial cells of the villus, as determined by in situ hybridization. Treatment of Caco-2/15 cells with a furin inhibitor led to inhibition of LPH activity. Morphological differentiation of enterocyte-like features in Caco-2/15 such as epithelial cell polarity and brush-border formation were strongly attenuated by furin inhibition. These results suggest that CDX2 regulates furin expression in intestinal epithelial cells. Furin may be important in modulating the maturation and/or activation of key factors involved in enterocyte differentiation.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Epiteliales/fisiología , Furina/biosíntesis , Furina/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Mucosa Intestinal/metabolismo , Transactivadores/genética , Transactivadores/fisiología , Fosfatasa Alcalina/metabolismo , Animales , Northern Blotting , Factor de Transcripción CDX2 , Línea Celular , Cromatina/genética , ADN/biosíntesis , ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Inhibidores Enzimáticos/farmacología , Células Epiteliales/ultraestructura , Humanos , Hibridación in Situ , Intestinos/ultraestructura , Luciferasas/genética , Microscopía Electrónica , Análisis de Secuencia por Matrices de Oligonucleótidos , Purinas/farmacología , Ratas , Retroviridae/genética , Roscovitina , Transfección
12.
DNA Cell Biol ; 24(4): 209-17, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15812237

RESUMEN

Butyrate (NaBu) regulates intestinal inflammatory gene expression in part through inhibition of deacetylase activity, but the exact mechanisms involved remain to be determined. In this study, we showed by Northern blot a synergistic induction of the acute phase protein gene SAA2 with a combination of deacetylase inhibitors (Trichostatin A or NaBu) and IL-1beta in the colon carcinoma cell line Caco-2. While the NF-kappa B DNA-binding site was essential for SAA2 regulation by IL-1beta and deacetylase inhibitors, the C/EBP DNA-binding site modulated SAA2 expression levels, as assessed by transient transfection assays and mutagenesis studies. NaBu was sufficient to induce SAA2 expression after transient treatment with IL-1beta and, conversely, IL-1beta induced SAA2 after transient treatment with NaBu. These data suggest that pretreatment with either NaBu or IL-1beta predisposes the SAA2 promoter to further stimulation. Indeed, both NaBu and IL-1beta led to increased recruitment of NF-kappa B p65, C/EBPbeta, and C/EBP delta, and decreased NF-kappa B p50 and C/EBP alpha DNA-binding to the proximal SAA2 promoter, as assessed by chromatin immunoprecipitation assays. Interestingly, while IL-1beta, in contrast to NaBu, induced histone H4 acetylation, addition of IL-1beta and NaBu increased histone H4 acetylation and both C/EBPbeta and NF-kappa B p65 DNA-binding. Therefore, these results suggest that NaBu and IL- 1beta mediate SAA2 synergistic induction by establishing and maintaining similar and complementary chromatin modifications and transcription factor recruitment as well. In addition to global effects, NaBu specifically regulate gene expression, as exemplified by SAA2.


Asunto(s)
Butiratos/metabolismo , Regulación de la Expresión Génica/fisiología , Ácidos Hidroxámicos/metabolismo , Interleucina-1/metabolismo , Regiones Promotoras Genéticas/genética , Proteína Amiloide A Sérica/genética , Sitios de Unión , Northern Blotting , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína delta de Unión al Potenciador CCAAT , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Colon/metabolismo , Cartilla de ADN , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Inhibidores de Histona Desacetilasas , Histonas/metabolismo , Humanos , Inmunoprecipitación , Luciferasas , Mutagénesis Sitio-Dirigida , FN-kappa B/metabolismo , Factor de Transcripción ReIA , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...