Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Cell ; 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38942017

RESUMEN

Recent advances in human genetics have shed light on the genetic factors contributing to inflammatory diseases, particularly Crohn's disease (CD), a prominent form of inflammatory bowel disease. Certain risk genes associated with CD directly influence cytokine biology and cell-specific communication networks. Current CD therapies primarily rely on anti-inflammatory drugs, which are inconsistently effective and lack strategies for promoting epithelial restoration and mucosal balance. To understand CD's underlying mechanisms, we investigated the link between CD and the FGFR1OP gene, which encodes a centrosome protein. FGFR1OP deletion in mouse intestinal epithelial cells disrupted crypt architecture, resulting in crypt loss, inflammation, and fatality. FGFR1OP insufficiency hindered epithelial resilience during colitis. FGFR1OP was crucial for preserving non-muscle myosin II activity, ensuring the integrity of the actomyosin cytoskeleton and crypt cell adhesion. This role of FGFR1OP suggests that its deficiency in genetically predisposed individuals may reduce epithelial renewal capacity, heightening susceptibility to inflammation and disease.

2.
Sci Transl Med ; 15(719): eadg5252, 2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37878672

RESUMEN

Effective tissue repair requires coordinated intercellular communication to sense damage, remodel the tissue, and restore function. Here, we dissected the healing response in the intestinal mucosa by mapping intercellular communication at single-cell resolution and integrating with spatial transcriptomics. We demonstrated that a risk variant for Crohn's disease, hepatocyte growth factor activator (HGFAC) Arg509His (R509H), disrupted a damage-sensing pathway connecting the coagulation cascade to growth factors that drive the differentiation of wound-associated epithelial (WAE) cells and production of a localized retinoic acid (RA) gradient to promote fibroblast-mediated tissue remodeling. Specifically, we showed that HGFAC R509H was activated by thrombin protease activity but exhibited impaired proteolytic activation of the growth factor macrophage-stimulating protein (MSP). In Hgfac R509H mice, reduced MSP activation in response to wounding of the colon resulted in impaired WAE cell induction and delayed healing. Through integration of single-cell transcriptomics and spatial transcriptomics, we demonstrated that WAE cells generated RA in a spatially restricted region of the wound site and that mucosal fibroblasts responded to this signal by producing extracellular matrix and growth factors. We further dissected this WAE cell-fibroblast signaling circuit in vitro using a genetically tractable organoid coculture model. Collectively, these studies exploited a genetic perturbation associated with human disease to disrupt a fundamental biological process and then reconstructed a spatially resolved mechanistic model of tissue healing.


Asunto(s)
Enfermedad de Crohn , Ratones , Humanos , Animales , Enfermedad de Crohn/genética , Enfermedad de Crohn/metabolismo , Transducción de Señal , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Diferenciación Celular
3.
Mol Psychiatry ; 27(3): 1405-1415, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35260802

RESUMEN

A missense mutation (A391T) in SLC39A8 is strongly associated with schizophrenia in genomic studies, though the molecular connection to the brain is unknown. Human carriers of A391T have reduced serum manganese, altered plasma glycosylation, and brain MRI changes consistent with altered metal transport. Here, using a knock-in mouse model homozygous for A391T, we show that the schizophrenia-associated variant changes protein glycosylation in the brain. Glycosylation of Asn residues in glycoproteins (N-glycosylation) was most significantly impaired, with effects differing between regions. RNAseq analysis showed negligible regional variation, consistent with changes in the activity of glycosylation enzymes rather than gene expression. Finally, nearly one-third of detected glycoproteins were differentially N-glycosylated in the cortex, including members of several pathways previously implicated in schizophrenia, such as cell adhesion molecules and neurotransmitter receptors that are expressed across all cell types. These findings provide a mechanistic link between a risk allele and potentially reversible biochemical changes in the brain, furthering our molecular understanding of the pathophysiology of schizophrenia and a novel opportunity for therapeutic development.


Asunto(s)
Proteínas de Transporte de Catión , Esquizofrenia , Animales , Encéfalo/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Glicosilación , Manganeso/metabolismo , Ratones , Esquizofrenia/genética
4.
Science ; 371(6524)2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33384352

RESUMEN

Tissue homeostasis is perturbed in a diversity of inflammatory pathologies. These changes can elicit endoplasmic reticulum (ER) stress, protein misfolding, and cell death. ER stress triggers the unfolded protein response (UPR), which can promote recovery of ER proteostasis and cell survival or trigger programmed cell death. Here, we leveraged single-cell RNA sequencing to define dynamic transcriptional states associated with the adaptive versus terminal UPR in the mouse intestinal epithelium. We integrated these transcriptional programs with genome-scale CRISPR screening to dissect the UPR pathway functionally. We identified QRICH1 as a key effector of the PERK-eIF2α axis of the UPR. QRICH1 controlled a transcriptional program associated with translation and secretory networks that were specifically up-regulated in inflammatory pathologies. Thus, QRICH1 dictates cell fate in response to pathological ER stress.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Estrés del Retículo Endoplásmico/genética , Regulación de la Expresión Génica , Inflamación/metabolismo , Proteostasis/genética , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada/genética , Animales , Apoptosis , Células Cultivadas , Proteínas de Unión al ADN/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Inflamación/genética , Inflamación/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Organoides , RNA-Seq , Análisis de la Célula Individual , Factores de Transcripción/genética , Transcripción Genética , eIF-2 Quinasa/metabolismo
5.
Proc Natl Acad Sci U S A ; 117(46): 28930-28938, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33139556

RESUMEN

Common genetic variants interact with environmental factors to impact risk of heritable diseases. A notable example of this is a single-nucleotide variant in the Solute Carrier Family 39 Member 8 (SLC39A8) gene encoding the missense variant A391T, which is associated with a variety of traits ranging from Parkinson's disease and neuropsychiatric disease to cardiovascular and metabolic diseases and Crohn's disease. The remarkable extent of pleiotropy exhibited by SLC39A8 A391T raises key questions regarding how a single coding variant can contribute to this diversity of clinical outcomes and what is the mechanistic basis for this pleiotropy. Here, we generate a murine model for the Slc39a8 A391T allele and demonstrate that these mice exhibit Mn deficiency in the colon associated with impaired intestinal barrier function and epithelial glycocalyx disruption. Consequently, Slc39a8 A391T mice exhibit increased sensitivity to epithelial injury and pathological inflammation in the colon. Taken together, our results link a genetic variant with a dietary trace element to shed light on a tissue-specific mechanism of disease risk based on impaired intestinal barrier integrity.


Asunto(s)
Proteínas de Transporte de Catión/genética , Enfermedad de Crohn/genética , Manganeso/metabolismo , Alelos , Animales , Proteínas de Transporte de Catión/metabolismo , Técnicas de Sustitución del Gen/métodos , Homeostasis/genética , Humanos , Inflamación/genética , Mucosa Intestinal/metabolismo , Intestinos/fisiología , Manganeso/fisiología , Ratones , Mutación Missense/genética , Fenotipo , Factores de Riesgo
6.
Cell Host Microbe ; 25(5): 668-680.e7, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-31071294

RESUMEN

Sphingolipids are structural membrane components and important eukaryotic signaling molecules. Sphingolipids regulate inflammation and immunity and were recently identified as the most differentially abundant metabolite in stool from inflammatory bowel disease (IBD) patients. Commensal bacteria from the Bacteroidetes phylum also produce sphingolipids, but the impact of these metabolites on host pathways is largely uncharacterized. To determine whether bacterial sphingolipids modulate intestinal health, we colonized germ-free mice with a sphingolipid-deficient Bacteroides thetaiotaomicron strain. A lack of Bacteroides-derived sphingolipids resulted in intestinal inflammation and altered host ceramide pools in mice. Using lipidomic analysis, we described a sphingolipid biosynthesis pathway and revealed a variety of Bacteroides-derived sphingolipids including ceramide phosphoinositol and deoxy-sphingolipids. Annotating Bacteroides sphingolipids in an IBD metabolomic dataset revealed lower abundances in IBD and negative correlations with inflammation and host sphingolipid production. These data highlight the role of bacterial sphingolipids in maintaining homeostasis and symbiosis in the gut.


Asunto(s)
Bacteroides thetaiotaomicron/crecimiento & desarrollo , Bacteroides thetaiotaomicron/metabolismo , Interacciones Microbiota-Huesped , Intestinos/microbiología , Intestinos/fisiología , Esfingolípidos/metabolismo , Simbiosis/efectos de los fármacos , Animales , Vida Libre de Gérmenes , Homeostasis/efectos de los fármacos , Enfermedades Inflamatorias del Intestino/prevención & control , Intestinos/efectos de los fármacos , Ratones
7.
Science ; 359(6380): 1161-1166, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29420262

RESUMEN

Polymorphisms in C1orf106 are associated with increased risk of inflammatory bowel disease (IBD). However, the function of C1orf106 and the consequences of disease-associated polymorphisms are unknown. Here we demonstrate that C1orf106 regulates adherens junction stability by regulating the degradation of cytohesin-1, a guanine nucleotide exchange factor that controls activation of ARF6. By limiting cytohesin-1-dependent ARF6 activation, C1orf106 stabilizes adherens junctions. Consistent with this model, C1orf106-/- mice exhibit defects in the intestinal epithelial cell barrier, a phenotype observed in IBD patients that confers increased susceptibility to intestinal pathogens. Furthermore, the IBD risk variant increases C1orf106 ubiquitination and turnover with consequent functional impairments. These findings delineate a mechanism by which a genetic polymorphism fine-tunes intestinal epithelial barrier integrity and elucidate a fundamental mechanism of cellular junctional control.


Asunto(s)
Uniones Adherentes/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Enfermedades Inflamatorias del Intestino/genética , Fosfoproteínas/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/metabolismo , Animales , Células CACO-2 , Células HEK293 , Humanos , Inmunoprecipitación , Mucosa Intestinal/patología , Ratones , Ratones Mutantes , Fosfoproteínas/genética , Polimorfismo Genético , Proteolisis , Riesgo , Ubiquitinación/genética
8.
J Gastroenterol ; 53(9): 1035-1047, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29374777

RESUMEN

BACKGROUND: Intestinal stem cells (ISCs) play indispensable roles in the maintenance of homeostasis, and also in the regeneration of the damaged intestinal epithelia. However, whether the inflammatory environment of Crohn's disease (CD) affects properties of resident small intestinal stem cells remain uncertain. METHODS: CD patient-derived small intestinal organoids were established from enteroscopic biopsy specimens taken from active lesions (aCD-SIO), or from mucosa under remission (rCD-SIO). Expression of ISC-marker genes in those organoids was examined by immunohistochemistry, and also by microfluid-based single-cell multiplex gene expression analysis. The ISC-specific function of organoid cells was evaluated using a single-cell organoid reformation assay. RESULTS: ISC-marker genes, OLFM4 and SLC12A2, were expressed by an increased number of small intestinal epithelial cells in the active lesion of CD. aCD-SIOs, rCD-SIOs or those of non-IBD controls (NI-SIOs) were successfully established from 9 patients. Immunohistochemistry showed a comparable level of OLFM4 and SLC12A2 expression in all organoids. Single-cell gene expression data of 12 ISC-markers were acquired from a total of 1215 cells. t-distributed stochastic neighbor embedding analysis identified clusters of candidate ISCs, and also revealed a distinct expression pattern of SMOC2 and LGR5 in ISC-cluster classified cells derived from aCD-SIOs. Single-cell organoid reformation assays showed significantly higher reformation efficiency by the cells of the aCD-SIOs compared with that of cells from NI-SIOs. CONCLUSIONS: aCD-SIOs harbor ISCs with modified marker expression profiles, and also with high organoid reformation ability. Results suggest modification of small intestinal stem cell properties by unidentified factors in the inflammatory environment of CD.


Asunto(s)
Enfermedad de Crohn/genética , Enfermedad de Crohn/patología , Intestino Delgado/patología , Organoides/diagnóstico por imagen , Organoides/patología , Análisis de la Célula Individual/métodos , Células Madre/patología , Enteroscopia de Balón , Biomarcadores/metabolismo , Biopsia , Proteínas de Unión al Calcio/biosíntesis , Enfermedad de Crohn/diagnóstico por imagen , Progresión de la Enfermedad , Células Epiteliales/patología , Expresión Génica , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Factor Estimulante de Colonias de Granulocitos/genética , Humanos , Inflamación/fisiopatología , Intestino Delgado/diagnóstico por imagen , Receptores Acoplados a Proteínas G/biosíntesis , Miembro 2 de la Familia de Transportadores de Soluto 12/biosíntesis , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Transcriptoma
9.
Stem Cell Reports ; 10(1): 27-42, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29233556

RESUMEN

ATOH1 is a master transcription factor for the secretory lineage differentiation of intestinal epithelial cells (IECs). However, the comprehensive contribution of ATOH1+ secretory lineage IECs to the homeostasis, repair, and tumorigenesis of the intestinal epithelium remains uncertain. Through our ATOH1+ cell-lineage tracing, we show here that a definite number of ATOH1+ IECs retain stem cell properties and can form ATOH1+IEC-derived clonal ribbons (ATOH1+ICRs) under completely homeostatic conditions. Interestingly, colonic ATOH1+ IECs appeared to exhibit their stem cell function more frequently compared with those of the small intestine. Consistently, the formation of ATOH1+ICRs was significantly enhanced upon dextran sodium sulfate colitis-induced mucosal damage. In addition, colonic ATOH1+ IECs acquired tumor stem cell-like properties in the azoxymethane-DSS tumor model. Our results reveal an unexpected contribution of colonic ATOH1+ IECs to maintaining the stem cell population under both homeostatic and pathologic conditions and further illustrate the high plasticity of the crypt-intrinsic stem cell hierarchy.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Transformación Celular Neoplásica/metabolismo , Colon/metabolismo , Neoplasias del Colon/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Compuestos Azo/toxicidad , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Colon/lesiones , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Células Epiteliales/patología , Mucosa Intestinal/lesiones , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/patología
10.
Data Brief ; 10: 551-556, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28124021

RESUMEN

The data on the immunohistochemical analysis of conditional Notch ligand knockout mice is presented. Targeted deletion of Jag1, Dll1, Dll4, or Dll1 plus Dll4 in Lgr5+ve cells was induced by a Cre-mediated gene recombination, and differentiation or proliferation of the intestinal epithelial cells was examined by immunohistochemistry. These data are the extension of the data presented and discussed in the paper entitled "Indispensable role of non-canonical Notch signaling in the proliferation of Apc-deficient intestinal tumors" (Nakata et al., Submitted for publication) [1].

11.
Int J Biol Macromol ; 97: 173-180, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28064047

RESUMEN

Soybeans are part of the traditional food consumed in Asia countries. In this study, we investigated inhibitory effects of soybean oligosaccharides and water-soluble soybean fibre (Soyafibe) on putrefactive compounds from soy protein by gut microbiota in rats. Caecal microbial fermentation products and microbiota in rats fed 20% soy protein (SP-1) and whole soybean flour (SFL: protein content was 20%) diets were determined. The caecal environment in rats fed 20% soy protein without dietary fibre (SP-2) or with 2% Soyafibe (SFB) was also determined. Compared to SP-1 and SP-2 group, low indole content with high lactic acid was shown in SFL and SFB group, respectively. Using the 16S rRNA genes polymerase chain reaction-denaturing gradient gel electrophoresis (PCR-DGGE) and pyrosequencing. Prevotella, Gram-negative anaerobic rods, were detected as dominant in both SFL and SFB groups. Our findings indicated that fermentable polysaccharides in soybeans have inhibitory effects on the formation of putrefactive compounds generated from soy protein by the microbiota.


Asunto(s)
Ciego/metabolismo , Fibras de la Dieta , Microbioma Gastrointestinal/efectos de los fármacos , Glycine max/química , Oligosacáridos/farmacología , Proteínas de Soja/farmacología , Agua/química , Animales , Bacterias/metabolismo , Biodiversidad , Peso Corporal/efectos de los fármacos , Ciego/efectos de los fármacos , Ciego/crecimiento & desarrollo , Ciego/microbiología , Proteínas en la Dieta/farmacología , Heces/química , Harina/análisis , Concentración de Iones de Hidrógeno , Masculino , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Wistar , Solubilidad
12.
Biochem Biophys Res Commun ; 482(4): 1296-1303, 2017 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-27939883

RESUMEN

Ligand-dependent activation of Notch signaling is required to maintain the stem-cell niche of normal intestinal epithelium. However, the precise role of Notch signaling in the maintenance of the intestinal tumor stem cell niche and the importance of the RBPJ-independent non-canonical pathway in intestinal tumors remains unknown. Here we show that Notch signaling was activated in LGR5+ve cells of APC-deficient mice intestinal tumors. Accordingly, Notch ligands, including Jag1, Dll1, and Dll4, were expressed in these tumors. In vitro studies using tumor-derived organoids confirmed the intrinsic Notch activity-dependent growth of tumor cells. Surprisingly, the targeted deletion of Jag1 but not RBPJ in LGR5+ve tumor-initiating cells resulted in the silencing of Hes1 expression, disruption of the tumor stem cell niche, and dramatic reduction in the proliferation activity of APC-deficient intestinal tumors in vivo. Thus, our results highlight the importance of ligand-dependent non-canonical Notch signaling in the proliferation and maintenance of the tumor stem cell niche in APC-deficient intestinal adenomas.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Neoplasias Intestinales/metabolismo , Proteína Jagged-1/genética , Receptores Notch/metabolismo , Células Madre/citología , Adenoma/metabolismo , Animales , Proliferación Celular , Factor de Crecimiento Epidérmico/metabolismo , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Ligandos , Ratones , Microscopía Fluorescente , Células Madre Neoplásicas/citología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
13.
Sci Rep ; 6: 36795, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27827428

RESUMEN

Intestinal epithelial cells (IECs) play an indispensable role in maintaining body fluid balance partly through their ability to regulate anion/fluid secretion. Yet in various inflammatory gastrointestinal diseases, over-secretion of anions results in symptoms such as severe diarrhoea. Endogenous mediators, such as vasoactive intestinal peptide or prostaglandin E2 (PGE2), regulate intestinal anion/fluid secretion, but their direct effect on purified human IECs has never been described in detail. Based on a previously described intestinal organoid swelling model, we established a 3D-scanner-assisted quantification method to evaluate the anion/fluid secretory response of cultured human IECs. Among various endogenous secretagogues, we found that PGE2 had the lowest EC50 value with regard to the induction of swelling of the jejunal and colonic organoids. This PGE2-mediated swelling response was dependent on environmental Cl- concentrations as well as on several channels and transporters as shown by a series of chemical inhibitor studies. The concomitant presence of various inflammatory cytokines with PGE2 failed to modulate the PGE2-mediated organoid swelling response. Therefore, the present study features PGE2 as a direct and robust mediator of anion/fluid secretion by IECs in the human intestine.


Asunto(s)
Aniones/metabolismo , Líquidos Corporales/metabolismo , Colon/efectos de los fármacos , Dinoprostona/farmacología , Yeyuno/efectos de los fármacos , Células Cultivadas , Cloruros/metabolismo , Colon/citología , Colon/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Yeyuno/citología , Organoides/citología , Organoides/metabolismo
14.
Biosci Microbiota Food Health ; 35(3): 137-40, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27508115

RESUMEN

Oxidative stress is considered an etiological factor responsible for several symptoms of inflammatory bowel disease (IBD). In vitro anti-inflammatory activities of heat-killed Lactococcus lactis subsp. lactis BF3 have been reported. In this study, the anti-inflammatory effect of these cells was examined using a dextran sodium sulphate (DSS)-induced murine IBD model. Administration of heat-killed L. lactis BF3 via drinking water suppressed the IBD symptoms, such as shortening of colon length, damage to the colon mucosa as observed under the microscope, and spleen enlargement. This result suggests that heat-killed L. lactis BF3 has the potential to treat IBD.

15.
Carbohydr Polym ; 143: 61-9, 2016 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-27083344

RESUMEN

Soybean is one of the major components of the Japanese diet. In traditional Japanese cuisine, soybean-based food items are often consumed with brown algae. In this study, we examined the effect of water-soluble and fermentable polysaccharides, laminaran and sodium alginate, from brown algae, on putrefactive compound production, by human faecal microbiota in broth containing 3% (w/v) soy protein. We also investigated the effect of 2% laminaran or alginate diet on caecal putrefactive compounds in rats maintained on diets containing 20% (w/w) soy protein. The caecal microbiota was also analysed using denaturing gradient gel electrophoresis and pyrosequencing with primers targeting the bacterial 16S rRNA gene. The polysaccharides, particularly laminaran, inhibited ammonia, phenol, and indole production by human faecal microbiota. Both the algal polysaccharides lowered the caecal indole content. Laminaran was found to increase the number of Coprobacter, whereas Helicobacter was found to decrease in the presence of both laminaran and sodium alginate.


Asunto(s)
Alginatos/farmacología , Microbioma Gastrointestinal/fisiología , Glucanos/farmacología , Indoles/antagonistas & inhibidores , Proteínas de Soja/metabolismo , Acetatos/metabolismo , Adulto , Amoníaco/antagonistas & inhibidores , Amoníaco/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Heces/microbiología , Fermentación , Ácido Glucurónico/farmacología , Ácidos Hexurónicos/farmacología , Humanos , Concentración de Iones de Hidrógeno , Indoles/metabolismo , Ácido Láctico/biosíntesis , Masculino , Tamaño de los Órganos/efectos de los fármacos , Phaeophyceae , Fenol/antagonistas & inhibidores , Fenol/metabolismo , ARN Ribosómico 16S/genética , Ratas Wistar , Proteínas de Soja/administración & dosificación
16.
Biochem Biophys Res Commun ; 469(4): 1062-8, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26724533

RESUMEN

Fetal hepatic stem/progenitor cells, called hepatoblasts, play central roles in liver development; however, the molecular mechanisms regulating the phenotype of these cells have not been completely elucidated. Matrix metalloproteinase (MMP)-14 is a type I transmembrane proteinase regulating pericellular proteolysis of the extracellular matrix and is essential for the activation of several MMPs and cytokines. However, the physiological functions of MMP-14 in liver development are unknown. Here we describe a functional role for MMP-14 in hepatic and biliary differentiation of mouse hepatoblasts. MMP-14 was upregulated in cells around the portal vein in perinatal stage liver. Formation of bile duct-like structures in MMP-14-deficient livers was significantly delayed compared with wild-type livers in vivo. In vitro biliary differentiation assays showed that formation of cholangiocytic cysts derived from MMP-14-deficient hepatoblasts was completely impaired, and that overexpression of MMP-14 in hepatoblasts promoted the formation of bile duct-like cysts. In contrast, the expression of molecules associated with metabolic functions in hepatocytes, including hepatic nuclear factor 4α and tryptophan 2,3-dioxygenase, were significantly increased in MMP-14-deficient livers. Expression of the epidermal growth factor receptor and phosphorylation of mitogen-activated protein kinases were significantly upregulated in MMP-14-deficient livers. We demonstrate that MMP-14-mediated signaling in fetal hepatic progenitor cells promotes biliary luminal formation around the portal vein and negatively controls the maturation of hepatocytes.


Asunto(s)
Conductos Biliares/citología , Conductos Biliares/fisiología , Hígado/citología , Hígado/fisiología , Metaloproteinasa 14 de la Matriz/metabolismo , Células Madre/enzimología , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre/citología
17.
Int Immunopharmacol ; 26(2): 295-303, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25887264

RESUMEN

Six lactic acid bacteria strains (four Lactobacillus plantarum strains and one each of Lactococcus lactis subsp. lactis and Pediococcus pentosaceus) have been isolated and shown to possess anti-oxidant activity. In this study, we determined their acid, bile, salt resistance, and adhesion activity on human enterocyte-like HT-29-Luc and Caco-2 cells. An isolate Lc. lactis S-SU2 showed highest bile resistance and adhesion activity compared to type strains. S-SU2 could ferment both 10% skimmed milk and soy milk while the type strain could not ferment soy milk. Soy milk fermented with S-SU2 showed an increased nitric oxide (NO) secretion in the mouse macrophage RAW264.7 cells without bacterial lipopolysaccharide (LPS). Furthermore, the inhibitory effects of the fermented soy milk on Escherichia coli O111 LPS-induced NO secretion were higher than those of fresh soy milk. Inflammatory bowel disease (IBD) was induced in mice fed either 5% (w/v) dextran sodium sulfate (DSS) in drinking water or 50% soy milk in drinking water. Shortening of colon length, breaking of epithelial cells, lowering liver and thymus weights, and enlargement of spleen are some of the characteristics observed in the IBD, which were prevented by the use of soy milk fermented with Lc. lactis S-SU2.


Asunto(s)
Colon/patología , Productos Lácteos Cultivados/metabolismo , Enterocitos/inmunología , Escherichia coli/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Lactococcus lactis/metabolismo , Macrófagos/metabolismo , Animales , Antiinflamatorios/metabolismo , Antioxidantes/metabolismo , Células CACO-2 , Bovinos , Colitis/inducido químicamente , Colon/efectos de los fármacos , Sulfato de Dextran/administración & dosificación , Modelos Animales de Enfermedad , Enterocitos/metabolismo , Células HT29 , Humanos , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/inmunología , Lipopolisacáridos/metabolismo , Macrófagos/inmunología , Ratones , Leche/metabolismo , Óxido Nítrico/metabolismo , Leche de Soja/metabolismo
18.
PeerJ ; 2: e370, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24860699

RESUMEN

Reports have suggested that the two Notch ligands, Dll1 and Dll4, are indispensable to maintain the homeostasis of the intestinal epithelium. However, within the intestinal epithelium, the precise distribution of the cells that express those ligands at the protein level remains largely unknown. Here, we show a series of immunohistochemical analysis through which we successfully identified mice intestinal epithelial cells (IECs) that endogenously express Dll1 or Dll4. Results showed that Dll1-positive (Dll1+ve) IECs reside exclusively within the crypt, whereas Dll4-positive (Dll4+ve) IECs can locate both in the crypt and in the villus of the small intestine. Also in the colon, Dll1+ve IECs resided at the lower part of the crypt, whereas Dll4+ve IECs resided at both upper and lower part of the crypt, including the surface epithelium. Both Dll1+ve and Dll4+ve IECs were ATOH1-positive, but Hes1-negative cells, and located adjacent to Hes1-positive cells within the crypts. A sub-population of both Dll1+ve and Dll4+ve IECs appeared to co-express Muc2, but rarely co-expressed other secretory lineage markers. However, as compared to Dll1+ve IECs, Dll4+ve IECs included larger number of Muc2-postive IECs, suggesting that Dll4 is more preferentially expressed by goblet cells. Also, we identified that Dll4 is expressed in the Paneth cells of the small intestine, whereas Dll1 and Dll4 is expressed in the c-kit-positive IECs of the colon, indicating that Dll1+ve and Dll4+ve IECs may contribute to constitute the intestinal stem cell niche. Compared to the normal colon, analysis of DSS-colitis showed that number of Dll1+ve IECs significantly decrease in the elongated crypts of the inflamed colonic mucosa. In sharp contrast, number of Dll4+ve IECs showed a significant increase in those crypts, which was accompanied by the increase in number of Hes1-positive IECs. Those Dll4+ve IECs were mostly found adjacent to the Hes1-positive IECs, suggesting that Dll4 may act as a major Notch ligand in the crypts of the inflamed colonic mucosa. Our results illustrate distinct expression patterns of Dll1 and Dll4 within the intestinal epithelium, and suggest that these two ligands may have different roles in normal and inflamed mucosa.

19.
Cancer ; 120(2): 229-37, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24122122

RESUMEN

BACKGROUND: A new predictive biomarker for determining prognosis in patients with hepatocellular carcinoma (HCC) who receive sorafenib is required, because achieving a reduction in tumor size with sorafenib is rare, even in patients who have a favorable prognosis. Vascular endothelial growth factor (VEGF) receptor is a sorafenib target. In the current study, the authors examined changes in plasma VEGF concentrations during sorafenib treatment and determined the clinical significance of VEGF as a prognostic indicator in patients with HCC. METHODS: Plasma VEGF concentrations were serially measured in 63 patients with advanced HCC before and during sorafenib treatment. A plasma VEGF concentration that decreased >5% from the pretreatment level at 8 weeks was defined as a "VEGF decrease." An objective tumor response was determined using modified Response Evaluation Criteria in Solid Tumors 1 month after the initiation of therapy and every 3 months thereafter. RESULTS: Patients who had a VEGF decrease at week 8 (n=14) had a longer median survival than those who did not have a VEGF decrease (n=49; 30.9 months vs 14.4 months; P=.038). All patients who had a VEGF decrease survived for >6 months, and the patients who had both a VEGF decrease and an α-fetoprotein response (n=6) survived during the observation period (median, 19.7 months; range, 6.5-31.0 months). In univariate analyses, a VEGF decrease, radiologic findings classified as progressive disease, and major vascular invasion were associated significantly with 1-year survival; and, in multivariate analysis, a VEGF decrease was identified as an independent factor associated significantly with survival. CONCLUSIONS: A plasma VEGF concentration decrease at 8 weeks after starting sorafenib treatment may predict favorable overall survival in patients with advanced HCC.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/mortalidad , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/mortalidad , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/sangre , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/sangre , Carcinoma Hepatocelular/metabolismo , Femenino , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Persona de Mediana Edad , Análisis Multivariante , Niacinamida/efectos adversos , Niacinamida/uso terapéutico , Compuestos de Fenilurea/efectos adversos , Valor Predictivo de las Pruebas , Pronóstico , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/uso terapéutico , Sorafenib , Resultado del Tratamiento , alfa-Fetoproteínas/análisis
20.
Biochem Biophys Res Commun ; 443(3): 840-6, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24342613

RESUMEN

Notch signaling plays an essential role in the proliferation and differentiation of intestinal epithelial cells (IECs). We have previously shown that Notch signaling is up-regulated in the inflamed mucosa of ulcerative colitis (UC) and thereby plays an indispensable role in tissue regeneration. Here we show that in addition to Notch signaling, STAT3 signaling is highly activated in the inflamed mucosa of UC. Forced expression of the Notch target gene Hes1 dramatically enhanced the IL-22-mediated STAT3-dependent transcription in human IECs. This enhancement of STAT3-dependent transcription was achieved by the extended phosphorylation of STAT3 by Hes1. Microarray analysis revealed that Hes1-mediated enhancement of IL-22-STAT3 signaling significantly increased the induction of genes encoding antimicrobial peptides, such as REG1A, REG3A and REG3G, in human IECs. Conversely, the reduction of Hes1 protein levels with a γ-secretase inhibitor significantly down-regulated the induction of those genes in IECs, resulting in a markedly poor response to IL-22. Our present findings identify a new role for the molecular function of Hes1 in which the protein can interact with cytokine signals and regulate the immune response of IECs.


Asunto(s)
Antiinfecciosos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Enterocitos/metabolismo , Proteínas de Homeodominio/metabolismo , Interleucinas/farmacología , Factor de Transcripción STAT3/metabolismo , Transcripción Genética/efectos de los fármacos , Antígenos de Neoplasias/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Línea Celular , Colitis Ulcerosa/genética , Colitis Ulcerosa/patología , Enterocitos/efectos de los fármacos , Enterocitos/patología , Proteínas de Homeodominio/genética , Humanos , Inflamación/patología , Proteínas Asociadas a Pancreatitis , Fosforilación/efectos de los fármacos , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Transcripción HES-1 , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Interleucina-22
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...