Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Gastrointest Liver Physiol ; 315(2): G159-G170, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29597354

RESUMEN

The amount of dietary protein is associated with intestinal disease in different vertebrate species. In humans, this is exemplified by the association between high-protein intake and fermentation metabolite concentrations in patients with inflammatory bowel disease. In production animals, dietary protein intake is associated with postweaning diarrhea in piglets and with the occurrence of wet litter in poultry. The underlying mechanisms by which dietary protein contributes to intestinal problems remain largely unknown. Fermentation of undigested protein in the hindgut results in formation of fermentation products including short-chain fatty acids, branched-chain fatty acids, ammonia, phenolic and indolic compounds, biogenic amines, hydrogen sulfide, and nitric oxide. Here, we review the mechanisms by which these metabolites may cause intestinal disease. Studies addressing how different metabolites induce epithelial damage rely mainly on cell culture studies and occasionally on mice or rat models. Often, contrasting results were reported. The direct relevance of such studies for human, pig, and poultry gut health is therefore questionable and does not suffice for the development of interventions to improve gut health. We discuss a roadmap to improve our understanding of gut metabolites and microbial species associated with intestinal health in humans and production animals and to determine whether these metabolite/bacterial networks cause epithelial damage. The outcomes of these studies will dictate proof-of-principle studies to eliminate specific metabolites and or bacterial strains and will provide the basis for interventions aiming to improve gut health.


Asunto(s)
Proteínas en la Dieta/metabolismo , Tracto Gastrointestinal/metabolismo , Enfermedades Intestinales , Animales , Aves , Carbohidratos de la Dieta/metabolismo , Fermentación , Tracto Gastrointestinal/fisiopatología , Humanos , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/fisiopatología , Porcinos
2.
Gastroenterology ; 152(6): 1462-1476.e10, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28130067

RESUMEN

BACKGROUND & AIMS: The nuclear receptor subfamily 1 group H member 4 (NR1H4 or farnesoid X receptor [FXR]) regulates bile acid synthesis, transport, and catabolism. FXR also regulates postprandial lipid and glucose metabolism. We performed quantitative proteomic analyses of liver tissues from mice to evaluate these functions and investigate whether FXR regulates amino acid metabolism. METHODS: To study the role of FXR in mouse liver, we used mice with a disruption of Nr1h4 (FXR-knockout mice) and compared them with floxed control mice. Mice were gavaged with the FXR agonist obeticholic acid or vehicle for 11 days. Proteome analyses, as well as targeted metabolomics and chromatin immunoprecipitation, were performed on the livers of these mice. Primary rat hepatocytes were used to validate the role of FXR in amino acid catabolism by gene expression and metabolomics studies. Finally, control mice and mice with liver-specific disruption of Nr1h4 (liver FXR-knockout mice) were re-fed with a high-protein diet after 6 hours fasting and gavaged a 15NH4Cl tracer. Gene expression and the metabolome were studied in the livers and plasma from these mice. RESULTS: In livers of control mice and primary rat hepatocytes, activation of FXR with obeticholic acid increased expression of proteins that regulate amino acid degradation, ureagenesis, and glutamine synthesis. We found FXR to bind to regulatory sites of genes encoding these proteins in control livers. Liver tissues from FXR-knockout mice had reduced expression of urea cycle proteins, and accumulated precursors of ureagenesis, compared with control mice. In liver FXR-knockout mice on a high-protein diet, the plasma concentration of newly formed urea was significantly decreased compared with controls. In addition, liver FXR-knockout mice had reduced hepatic expression of enzymes that regulate ammonium detoxification compared with controls. In contrast, obeticholic acid increased expression of genes encoding enzymes involved in ureagenesis compared with vehicle in C57Bl/6 mice. CONCLUSIONS: In livers of mice, FXR regulates amino acid catabolism and detoxification of ammonium via ureagenesis and glutamine synthesis. Failure of the urea cycle and hyperammonemia are common in patients with acute and chronic liver diseases; compounds that activate FXR might promote ammonium clearance in these patients.


Asunto(s)
Amoníaco/metabolismo , Glutamina/biosíntesis , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Urea/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/farmacología , Proteínas en la Dieta/administración & dosificación , Expresión Génica , Hepatocitos , Hígado/enzimología , Masculino , Metaboloma , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteoma , Ratas , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores
3.
Proc Natl Acad Sci U S A ; 112(32): 10038-43, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26216954

RESUMEN

Colorectal cancer risk is associated with diets high in red meat. Heme, the pigment of red meat, induces cytotoxicity of colonic contents and elicits epithelial damage and compensatory hyperproliferation, leading to hyperplasia. Here we explore the possible causal role of the gut microbiota in heme-induced hyperproliferation. To this end, mice were fed a purified control or heme diet (0.5 µmol/g heme) with or without broad-spectrum antibiotics for 14 d. Heme-induced hyperproliferation was shown to depend on the presence of the gut microbiota, because hyperproliferation was completely eliminated by antibiotics, although heme-induced luminal cytotoxicity was sustained in these mice. Colon mucosa transcriptomics revealed that antibiotics block heme-induced differential expression of oncogenes, tumor suppressors, and cell turnover genes, implying that antibiotic treatment prevented the heme-dependent cytotoxic micelles to reach the epithelium. Our results indicate that this occurs because antibiotics reinforce the mucus barrier by eliminating sulfide-producing bacteria and mucin-degrading bacteria (e.g., Akkermansia). Sulfide potently reduces disulfide bonds and can drive mucin denaturation and microbial access to the mucus layer. This reduction results in formation of trisulfides that can be detected in vitro and in vivo. Therefore, trisulfides can serve as a novel marker of colonic mucolysis and thus as a proxy for mucus barrier reduction. In feces, antibiotics drastically decreased trisulfides but increased mucin polymers that can be lysed by sulfide. We conclude that the gut microbiota is required for heme-induced epithelial hyperproliferation and hyperplasia because of the capacity to reduce mucus barrier function.


Asunto(s)
Colon/microbiología , Colon/patología , Dieta , Células Epiteliales/patología , Hemo/farmacología , Microbiota/efectos de los fármacos , Moco/metabolismo , Animales , Antibacterianos/farmacología , Antioxidantes/farmacología , Biomarcadores/metabolismo , Peso Corporal/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Recuento de Colonia Microbiana , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Heces/microbiología , Inmunohistoquímica , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Antígeno Ki-67/metabolismo , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Moco/efectos de los fármacos , Sulfuros/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
4.
Biochim Biophys Acta ; 1862(2): 166-73, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26554605

RESUMEN

Inflammatory Bowel Disease (IBD) is a multifactorial disorder involving dysregulation of the immune response and bacterial translocation through the intestinal mucosal barrier. Previously, we have shown that activation of the bile acid sensor Farnesoid X Receptor (FXR), which belongs to the family of nuclear receptors, improves experimental intestinal inflammation, decreasing expression of pro-inflammatory cytokines and protecting the intestinal barrier. Here, we aimed to investigate the immunological mechanisms that ameliorate colitis when FXR is activated. We analyzed by FACS immune cell populations in mesenteric lymph nodes (MLN) and in the spleen to understand whether FXR activation alters the systemic immune response. We show that FXR activation by obeticholic acid (OCA) has systemic anti-inflammatory effects that include increased levels of plasma IL-10, inhibition of both DSS-colitis associated decrease in splenic dendritic cells (DCs) and increase in Tregs. Impact of OCA on DC relative abundance was seen in spleen but not MLN, possibly related to the increased FXR expression in splenic DCs compared to MLN DCs. Moreover, FXR activation modulates the chemotactic environment in the colonic site of inflammation, as Madcam1 expression is decreased, while Ccl25 is upregulated. Together, our data suggest that OCA treatment elicits an anti-inflammatory immune status including retention of DCs in the spleen, which is associated with decreased colonic inflammation. Pharmacological FXR activation is therefore an attractive new drug target for treatment of IBD.


Asunto(s)
Colitis/inducido químicamente , Colitis/inmunología , Células Dendríticas/inmunología , Sulfato de Dextran , Receptores Citoplasmáticos y Nucleares/inmunología , Bazo/inmunología , Animales , Quimiotaxis , Colitis/patología , Colon/citología , Colon/inmunología , Colon/patología , Células Dendríticas/patología , Interleucina-10/inmunología , Masculino , Ratones Endogámicos C57BL , Bazo/citología , Bazo/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
5.
Biochim Biophys Acta Mol Basis Dis ; 1863(3): 687-700, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27956139

RESUMEN

Nuclear receptors (NRs) are ligand-activated transcription factors regulating a large variety of processes involved in reproduction, development, and metabolism. NRs are ideal drug targets because they are activated by lipophilic ligands that easily pass cell membranes. Immortalized cell lines recapitulate NR biology poorly and generating primary cultures is laborious and requires a constant need for donor material. There is a clear need for development of novel preclinical model systems that better resemble human physiology. Uncertainty due to technical limitations early in drug development is often the cause of preclinical drugs not reaching the clinic. Here, we studied whether organoids, mini-organs derived from the respective mouse tissue's stem cells, can serve as a novel model system to study NR biology and targetability. We characterized mRNA expression profiles of the NR superfamily in mouse liver, ileum, and colon organoids. Tissue-specific expression patterns were largely maintained in the organoids, indicating their suitability for NR research. Metabolic NRs Fxrα, Lxrα, Lxrß, Pparα, and Pparγ induced expression of and binding to endogenous target genes. Transcriptome analyses of wildtype colon organoids stimulated with Rosiglitazone showed that lipid metabolism was the highest significant changed function, greatly mimicking the PPARs and Rosiglitazone function in vivo. Finally, using organoids we identify Trpm6, Slc26a3, Ang1, and Rnase4, as novel Fxr target genes. Our results demonstrate that organoids represent a framework to study NR biology that can be further expanded to human organoids to improve preclinical testing of novel drugs that target this pharmacologically important class of ligand activated transcription factors.


Asunto(s)
Colon/citología , Íleon/citología , Hígado/citología , Organoides/citología , Receptores Citoplasmáticos y Nucleares/genética , Células Madre/citología , Transcriptoma , Animales , Colon/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Íleon/metabolismo , Hígado/metabolismo , Ratones , Técnicas de Cultivo de Órganos/métodos , Organoides/metabolismo , ARN Mensajero/genética , Células Madre/metabolismo
6.
J Hepatol ; 64(5): 1158-1166, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26812075

RESUMEN

BACKGROUND & AIMS: The bile acid-activated farnesoid X receptor (FXR) is a nuclear receptor regulating bile acid, glucose and cholesterol homeostasis. Obeticholic acid (OCA), a promising drug for the treatment of non-alcoholic steatohepatitis (NASH) and type 2 diabetes, activates FXR. Mouse studies demonstrated that FXR activation by OCA alters hepatic expression of many genes. However, no data are available on the effects of OCA in the human liver. Here we generated gene expression profiles in human precision cut liver slices (hPCLS) after treatment with OCA. METHODS: hPCLS were incubated with OCA for 24 h. Wild-type or FXR(-/-) mice received OCA or vehicle by oral gavage for 7 days. RESULTS: Transcriptomic analysis showed that well-known FXR target genes, including NR0B2 (SHP), ABCB11 (BSEP), SLC51A (OSTα) and SLC51B (OSTß), and ABCB4 (MDR3) are regulated by OCA in hPCLS. Ingenuity pathway analysis confirmed that 'FXR/RXR activation' is the most significantly changed pathway upon OCA treatment. Comparison of gene expression profiles in hPCLS and mouse livers identified 18 common potential FXR targets. ChIP-sequencing in mouse liver confirmed FXR binding to IR1 sequences of Akap13, Cgnl1, Dyrk3, Pdia5, Ppp1r3b and Tbx6. CONCLUSIONS: Our study shows that hPCLS respond to OCA treatment by upregulating well-known FXR target genes, demonstrating its suitability to study FXR-mediated gene regulation. We identified six novel bona-fide FXR target genes in both mouse and human liver. Finally, we discuss a possible explanation for changes in high or low density lipoprotein observed in NASH and primary biliary cholangitis patients treated with OCA based on the genomic expression profile in hPCLS.


Asunto(s)
Ácido Quenodesoxicólico/análogos & derivados , ADN/genética , Regulación de la Expresión Génica , Hígado/patología , Enfermedad del Hígado Graso no Alcohólico/genética , Receptores Citoplasmáticos y Nucleares/genética , Animales , Ácido Quenodesoxicólico/farmacología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/biosíntesis , Activación Transcripcional
7.
PLoS One ; 19(1): e0290493, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38181033

RESUMEN

Currently, over 88 million people are estimated to have adopted a vegan or vegetarian diet. Cysteine is a semi-essential amino acid, which availability is largely dependent on dietary intake of meat, eggs and whole grains. Vegan/vegetarian diets are therefore inherently low in cysteine. Sufficient uptake of cysteine is crucial, as it serves as substrate for protein synthesis and can be converted to taurine and glutathione. We found earlier that intermolecular cystine bridges are essential for the barrier function of the intestinal mucus layer. Therefore, we now investigate the effect of low dietary cystine on the intestine. Mice (8/group) received a high fat diet with a normal or low cystine concentration for 2 weeks. We observed no changes in plasma methionine, cysteine, taurine or glutathione levels or bile acid conjugation after 2 weeks of low cystine feeding. In the colon, dietary cystine restriction results in an increase in goblet cell numbers, and a borderline significant increase mucus layer thickness. Gut microbiome composition and expression of stem cell markers did not change on the low cystine diet. Remarkably, stem cell markers, as well as the proliferation marker Ki67, were increased upon cystine restriction in the small intestine. In line with this, gene set enrichment analysis indicated enrichment of Wnt signaling in the small intestine of mice on the low cystine diet, indicative of increased epithelial proliferation. In conclusion, 2 weeks of cystine restriction did not result in apparent systemic effects, but the low cystine diet increased the proliferative capacity specifically of the small intestine and induced the number of goblet cells in the colon.


Asunto(s)
Cisteína , Cistina , Humanos , Animales , Ratones , Intestino Delgado , Glutatión , Taurina
8.
Carcinogenesis ; 34(7): 1628-35, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23455377

RESUMEN

Red meat consumption is associated with an increased colon cancer risk. Heme, present in red meat, injures the colon surface epithelium by generating cytotoxic and oxidative stress. Recently, we found that this surface injury is compensated by hyperproliferation and hyperplasia of crypt cells, which was induced by a changed surface to crypt signaling. It is unknown whether this changed signaling is caused by cytotoxic stress and/or oxidative stress, as these processes were never studied separately. The aim of this study was to determine the possible differential effects of dietary heme on these luminal stressors and their impact on the colonic mucosa after 2, 4, 7 and 14 days of heme feeding. Mice received a purified, humanized, control diet or the diet supplemented with 0.2 µmol heme/g. Oxidative and cytotoxic stress were measured in fecal water. Proliferation was determined by Ki67-immunohistochemistry and mucosal responses by whole-genome transcriptomics. After heme ingestion, there was an acute increase in reactive oxygen species (ROS) leading to increased levels of lipid peroxidation products. Mucosal gene expression showed an acute antioxidant response, but no change in cell turnover. After day 4, cytotoxicity of the colonic contents was increased and this coincided with differential signaling and hyperproliferation, indicating that cytotoxicity was the causal factor. Simultaneously, several oncogenes were activated, whereas the tumor suppressor p53 was inhibited. In conclusion, luminal cytotoxicity, but not ROS, caused differential surface to crypt signaling resulting in mucosal hyperproliferation and the differential expression of oncogenes and tumor suppressor genes.


Asunto(s)
Proliferación Celular , Colon/efectos de los fármacos , Suplementos Dietéticos , Regulación Neoplásica de la Expresión Génica , Hemo/administración & dosificación , Estrés Oxidativo , Animales , Colon/química , Colon/patología , Heces/química , Hemo/farmacología , Inmunohistoquímica , Mucosa Intestinal/química , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Peroxidación de Lípido , Masculino , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/química , Factores de Tiempo , Transcriptoma
9.
Br J Nutr ; 110(2): 363-74, 2013 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-23228223

RESUMEN

Different lifestyle patterns across Europe may influence plasma concentrations of B-vitamins and one-carbon metabolites and their relation to chronic disease. Comparison of published data on one-carbon metabolites in Western European regions is difficult due to differences in sampling procedures and analytical methods between studies. The present study aimed, to compare plasma concentrations of one-carbon metabolites in Western European regions with one laboratory performing all biochemical analyses. We performed the present study in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort among 5446 presumptively healthy individuals. Quantile regression was used to compare sex-specific median concentrations between Northern (Denmark and Sweden), Central (France, Germany, The Netherlands and United Kingdom) and Southern (Greece, Spain and Italy) European regions. The lowest folate concentrations were observed in Northern Europe (men, 10·4 nmol/l; women, 10·7 nmol/l) and highest concentrations in Central Europe. Cobalamin concentrations were slightly higher in Northern Europe (men, 330 pmol/l; women, 352 pmol/l) compared with Central and Southern Europe, but did not show a clear north-south gradient. Vitamin B2 concentrations were highest in Northern Europe (men, 22·2 nmol/l; women, 26·0 nmol/l) and decreased towards Southern Europe (P trend< 0·001). Vitamin B(6) concentrations were highest in Central Europe in men (77·3 nmol/l) and highest in the North among women (70·4 nmol/l), with decreasing concentrations towards Southern Europe in women (P trend< 0·001). In men, concentrations of serine, glycine and sarcosine increased from the north to south. In women, sarcosine increased from Northern to Southern Europe. These findings may provide relevant information for the study of regional differences of chronic disease incidence in association with lifestyle.


Asunto(s)
Ácido Fólico/sangre , Glicina/sangre , Conductas Relacionadas con la Salud/etnología , Estilo de Vida/etnología , Sarcosina/sangre , Serina/sangre , Vitamina B 12/sangre , Adulto , Anciano , Consumo de Bebidas Alcohólicas/etnología , Enfermedad Crónica/etnología , Etnicidad , Europa (Continente) , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Factores Sexuales , Fumar/etnología
10.
Gut ; 61(7): 1041-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21948946

RESUMEN

OBJECTIVE: Colon cancer is a leading cause of cancer deaths in Western countries and is associated with diets high in red meat. Haem, the iron-porphyrin pigment of red meat, induces cytotoxicity of gut contents and damages the colon surface epithelium. Compensatory hyperproliferation leads to epithelial hyperplasia which increases the risk of colon cancer. The aim of this study was to identify molecules signalling from the surface epithelium to the crypt to initiate hyperproliferation upon stress induced by haem. METHODS: C57Bl6/J mice (n=9/group) received a 'westernised' control diet (40 en% fat) with or without 0.5 µmol/g haem for 14 days. Colon mucosa was used to quantify cell proliferation and for microarray transcriptome analysis. Gene expression profiles of surface and crypt cells were compared using laser capture microdissection. Protein levels of potential signalling molecules were quantified. RESULTS: Haem-fed mice showed epithelial hyperproliferation and decreased apoptosis, resulting in hyperplasia. Microarray analysis of the colon mucosa showed 3710 differentially expressed genes (false discovery rate (q) <0.01), with many involved in the cell cycle. Expression levels of haem- and stress-related genes showed that haem affected surface cells but did not directly affect crypt cells. Injured surface cells should therefore signal to crypt cells to induce compensatory hyperproliferation. Haem downregulated the inhibitors of proliferation, Wnt inhibitory factor 1, Indian Hedgehog and bone morphogenetic protein 2. Interleukin-15 was also downregulated. Haem upregulated amphiregulin, epiregulin and cyclo-oxygenase-2 mRNA in surface cells. Their protein/metabolite levels were, however, not increased as haem induced surface-specific inhibition of translation by increasing 4E-BP1. CONCLUSIONS: Haem induces colonic hyperproliferation and hyperplasia by inhibiting the surface to crypt signalling of feedback inhibitors of proliferation.


Asunto(s)
Colon/citología , Células Epiteliales/efectos de los fármacos , Hemo/farmacología , Mucosa Intestinal/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Colon/metabolismo , Neoplasias del Colon/etiología , Suplementos Dietéticos , Regulación hacia Abajo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Retroalimentación Fisiológica , Expresión Génica , Perfilación de la Expresión Génica , Captura por Microdisección con Láser , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal , Transcriptoma
11.
Heliyon ; 8(2): e08883, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35169646

RESUMEN

Dysbiosis-related perturbations in bile acid (BA) metabolism were observed in inflammatory bowel disease (IBD) patients, which was characterized by increased levels of sulfated BAs at the expense of secondary BAs. However, the exact effects of sulfated BAs on the etiology of IBD are not investigated yet. Therefore, we aimed to investigate the effects of sulfated deoxycholic acid (DCA), sulfated lithocholic acid (LCA) and their unsulfated forms on intestinal barrier function and immune response. To this end, we first established a novel in vitro human intestinal model to mimic chronic intestinal inflammation as seen during IBD. This model consisted of a co-culture of Caco-2 and HT29-MTX-E12 cells grown on a semi-wet interface with mechanical stimulation to represent the mucus layer. A pro-inflammatory environment was created by combining the co-culture with LPS-activated dendritic cells (DCs) in the basolateral compartment. The presence of activated DCs caused a decrease in transepithelial electrical resistance (TEER), which was slightly restored by LCA and sulfated DCA. The expression of genes related to intestinal epithelial integrity and the mucus layer were slightly, but not significantly increased. These results imply that sulfated BAs have a minor effect on intestinal barrier function in Caco-2 and HT29-MTX-E12 cells. When exposed directly to DCs, our results point towards anti-inflammatory effects of secondary BAs, but to a minor extent for sulfated secondary BAs. Future research should focus on the importance of proper transformation of BAs by bacterial enzymes and the potential involvement of BA dysmetabolism in IBD progression.

12.
Biochim Biophys Acta Mol Basis Dis ; 1867(11): 166229, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34329708

RESUMEN

Necrotizing enterocolitis (NEC) is an often lethal, inflammatory disease of the preterm intestine. The underdeveloped immune system plays an important role; however, the initial trigger for NEC development is likely a damaged intestinal epithelial layer. We hypothesize that due to incomplete maturation of different epithelial cell lineages, nutrients and bacteria are able to damage the epithelial cells and cause the (immature) inflammatory response, food intolerance and malabsorption seen in NEC. Intestinal organoid research has shown that maturation of intestinal epithelial cell lineages is orchestrated by two key signaling pathways: Wnt and Notch. In NEC, these pathways are dysregulated by hyperactivation of Toll-like-receptor-4. Breastfeeding decreases the risk of developing NEC compared to formula milk. Here, we review the intricate link between breast milk components, Wnt and Notch signaling and intestinal epithelial maturation. We argue that (nutritional) interventions regulating these pathways may decrease the risk of NEC development in preterm infants.


Asunto(s)
Enterocolitis Necrotizante/patología , Mucosa Intestinal/patología , Leche Humana/metabolismo , Nutrientes/metabolismo , Lactancia Materna , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/prevención & control , Humanos , Recién Nacido , Recien Nacido Prematuro , Mucosa Intestinal/metabolismo , Leche Humana/química , Receptores Notch/antagonistas & inhibidores , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/fisiología
13.
JHEP Rep ; 3(5): 100344, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34604725

RESUMEN

BACKGROUND & AIMS: The interorgan crosstalk between the liver and the intestine has been the focus of intense research. Key in this crosstalk are bile acids, which are secreted from the liver into the intestine, interact with the microbiome, and upon absorption reach back to the liver. The bile acid-activated farnesoid X receptor (Fxr) is involved in the gut-to-liver axis. However, liver-to-gut communication and the roles of bile acids and Fxr remain elusive. Herein, we aim to get a better understanding of Fxr-mediated liver-to-gut communication, particularly in colon functioning. METHODS: Fxr floxed/floxed mice were crossed with cre-expressing mice to yield Fxr ablation in the intestine (Fxr-intKO), liver (Fxr-livKO), or total body (Fxr-totKO). The effects on colonic gene expression (RNA sequencing), the microbiome (16S sequencing), and mucus barrier function by ex vivo imaging were analysed. RESULTS: Despite relatively small changes in biliary bile acid concentration and composition, more genes were differentially expressed in the colons of Fxr-livKO mice than in those of Fxr-intKO and Fxr-totKO mice (3272, 731, and 1824, respectively). The colons of Fxr-livKO showed increased expression of antimicrobial genes, Toll-like receptors, inflammasome-related genes and genes belonging to the 'Mucin-type O-glycan biosynthesis' pathway. Fxr-livKO mice have a microbiome profile favourable for the protective capacity of the mucus barrier. The thickness of the inner sterile mucus layer was increased and colitis symptoms reduced in Fxr-livKO mice. CONCLUSIONS: Targeting of FXR is at the forefront in the battle against metabolic diseases. We show that ablation of Fxr in the liver greatly impacts colonic gene expression and increased the colonic mucus barrier. Increasing the mucus barrier is of utmost importance to battle intestinal diseases such as inflammatory bowel disease, and we show that this might be done by antagonising FXR in the liver. LAY SUMMARY: This study shows that the communication of the liver to the intestine is crucial for intestinal health. Bile acids are key players in this liver-to-gut communication, and when Fxr, the master regulator of bile acid homoeostasis, is ablated in the liver, colonic gene expression is largely affected, and the protective capacity of the mucus barrier is increased.

14.
Trends Mol Med ; 22(3): 190-199, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26852376

RESUMEN

The gut epithelium is covered by mucus consisting of mucin polymers connected via disulfide bonds. The mucus layer limits exposure of epithelial cells to toxins and bacteria. A recent study has shown that sulfide, produced by certain bacteria, reduces disulfide bonds in the mucus network. The resulting breaks in the mucus barrier allow exposure of the epithelium to bacteria and toxins, causing inflammation. In this opinion article we argue that this mechanism may be involved in the etiology and/or severity of inflammatory bowel disease (IBD) because IBD is associated with decreased mucus barrier function, altered microbial species, and increased sulfide concentrations. Increasing the mucus integrity by reducing sulfide concentrations in the intestine may be a novel therapeutic option for IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Sulfuros/metabolismo , Animales , Microbioma Gastrointestinal , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Sulfatos/metabolismo
15.
PLoS One ; 7(8): e43260, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22905243

RESUMEN

Red meat consumption is associated with an increased colon cancer risk. Heme, present in red meat, injures the colon surface epithelium by luminal cytotoxicity and reactive oxygen species. This surface injury is overcompensated by hyperproliferation and hyperplasia of crypt cells. Transcriptome analysis of mucosa of heme-fed mice showed, besides stress- and proliferation-related genes, many upregulated lipid metabolism-related PPARα target genes. The aim of this study was to investigate the role of PPARα in heme-induced hyperproliferation and hyperplasia. Male PPARα KO and WT mice received a purified diet with or without heme. As PPARα is proposed to protect against oxidative stress and lipid peroxidation, we hypothesized that the absence of PPARα leads to more surface injury and crypt hyperproliferation in the colon upon heme-feeding. Heme induced luminal cytotoxicity and lipid peroxidation and colonic hyperproliferation and hyperplasia to the same extent in WT and KO mice. Transcriptome analysis of colonic mucosa confirmed similar heme-induced hyperproliferation in WT and KO mice. Stainings for alkaline phosphatase activity and expression levels of Vanin-1 and Nrf2-targets indicated a compromised antioxidant defense in heme-fed KO mice. Our results suggest that the protective role of PPARα in antioxidant defense involves the Nrf2-inhibitor Fosl1, which is upregulated by heme in PPARα KO mice. We conclude that PPARα plays a protective role in colon against oxidative stress, but PPARα does not mediate heme-induced hyperproliferation. This implies that oxidative stress of surface cells is not the main determinant of heme-induced hyperproliferation and hyperplasia.


Asunto(s)
Neoplasias del Colon/prevención & control , Hemo/química , PPAR alfa/metabolismo , Alimentación Animal , Animales , Antioxidantes/metabolismo , Proliferación Celular , Colon/metabolismo , Neoplasias del Colon/etiología , Inmunohistoquímica/métodos , Metabolismo de los Lípidos , Masculino , Carne , Ratones , Ratones Noqueados , Membrana Mucosa/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo , Especies Reactivas de Oxígeno , Riesgo
16.
BMC Med Genomics ; 5: 38, 2012 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-22929163

RESUMEN

BACKGROUND: By regulating digestion and absorption of nutrients and providing a barrier against the external environment the intestine provides a crucial contribution to the maintenance of health. To what extent aging-related changes in the intestinal system contribute to the functional decline associated with aging is still under debate. METHODS: Young (4 M) and old (21 M) male C57BL/6J mice were fed a control low-fat (10E%) or a high-fat diet (45E%) for 2 weeks. During the intervention gross energy intake and energy excretion in the feces were measured. After sacrifice the small and large intestine were isolated and the small intestine was divided in three equal parts. Swiss rolls were prepared of each of the isolated segments for histological analysis and the luminal content was isolated to examine alterations in the microflora with 16S rRNA Q-PCR. Furthermore, mucosal scrapings were isolated from each segment to determine differential gene expression by microarray analysis and global DNA methylation by pyrosequencing. RESULTS: Digestible energy intake was similar between the two age groups on both the control and the high-fat diet. Microarray analysis on RNA from intestinal scrapings showed no marked changes in expression of genes involved in metabolic processes. Decreased expression of Cubilin was observed in the intestine of 21-month-old mice, which might contribute to aging-induced vitamin B12 deficiency. Furthermore, microarray data analysis revealed enhanced expression of a large number of genes involved in immune response and inflammation in the colon, but not in the small intestine of the 21-month-old mice. Aging-induced global hypomethylation was observed in the colon and the distal part of the small intestine, but not in the first two sections of the small intestine. CONCLUSION: In 21-month old mice the most pronounced effects of aging were observed in the colon, whereas very few changes were observed in the small intestine.


Asunto(s)
Envejecimiento/genética , Colon/crecimiento & desarrollo , Colon/metabolismo , Intestino Delgado/crecimiento & desarrollo , Intestino Delgado/metabolismo , Animales , Peso Corporal/genética , Colon/inmunología , Colon/patología , Metilación de ADN/genética , Dieta Alta en Grasa , Ingestión de Energía/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Inmunidad/genética , Inflamación/genética , Intestino Delgado/patología , Masculino , Ratones , Ratones Endogámicos C57BL
17.
J Nutr Biochem ; 23(11): 1425-33, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22226334

RESUMEN

Oncoprotein-induced transcript 1 (Oit1) was previously identified as a dietary fat-induced gene in the small intestine of C57Bl/6J mice. In this study, we further characterized Oit1 and its human ortholog family with sequence similarity 3, member D (Fam3D), on the messenger RNA as well as the protein level. Oit1 and Fam3D were found to be predominantly expressed in the gastrointestinal tract of mice and humans, respectively. Dietary fat induced a clear and acute up-regulation of Oit1, especially in the jejunum, whereas fasting led to a reduced gene expression in the small intestine. Regarding protein expression, we found a remarkable pattern of Oit1 along the longitudinal axis of the intestine, a predominant villus-restricted expression in the proximal small intestine and a more pronounced crypt expression in the distal parts of the intestine. Using transfection experiments, we confirmed secretion of the Oit1 protein, as was predicted by a signal peptide sequence. Detection of Oit1 and Fam3D in plasma samples indicated that both proteins are secreted to the basolateral site of enterocytes. Moreover, in human plasma samples, we also found an effect of nutritional status on Fam3D levels, with a postprandial elevation and a reduction after fasting. In conclusion, Oit1 and Fam3D are gut-derived proteins that are expressed and secreted in a nutritional status-dependent manner.


Asunto(s)
Citocinas/metabolismo , Intestino Delgado/metabolismo , Estado Nutricional/fisiología , Adolescente , Adulto , Anciano , Animales , Colon/metabolismo , Citocinas/sangre , Citocinas/genética , Dieta Alta en Grasa/efectos adversos , Ayuno , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , PPAR alfa/genética , PPAR alfa/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA