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










Base de datos
Intervalo de año de publicación
1.
J Appl Glycosci (1999) ; 70(3): 59-66, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38143569

RESUMEN

Pectin, a type of soluble fiber, promotes morphological changes in the small intestinal villi. Although its physiological significance is unknown, we hypothesized that changes in villus morphology enhance the efficiency of nutrient absorption in the small intestine and investigated the effect of pectin derived from persimmon on calcium absorption using polarized Caco-2 cells. In polarized Caco-2 cells, pectin altered the mRNA expression levels of substances involved in calcium absorption and the regulation of intracellular calcium concentration and significantly reduced calcium absorption. Although this was comparable to the results of absorption and permeability associated with the addition of active vitamin D, the simultaneous action of pectin and active vitamin D did not show any additive effects. Furthermore, as active vitamin D significantly increases the activity of intestinal alkaline phosphatase (ALP), which is known to be involved in the regulation of intestinal absorption of calcium and lipids, we also investigated the effect of pectin on intestinal ALP activity. As a result, it was found that, unlike the effect of active vitamin D, pectin significantly reduced intestinal ALP activity. These results suggest that pectin stimulates polarized Caco-2 cells through a mechanism distinct from the regulation of calcium absorption by vitamin D, modulating total calcium absorption from the elongated villi through morphological changes in the small intestine by suppressing it at the cellular level.

2.
Biosci Biotechnol Biochem ; 85(7): 1729-1736, 2021 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-33877300

RESUMEN

Pectin in Diospyros kaki (persimmon) is a complex polysaccharide and is classified as a dietary fiber. Pectin is characterized by the presence of side chains of neutral sugars, such as galactose residues; however, the structure and properties of these sugars vary greatly depending on the plant species from which it is derived. Here, we report the structural features of pectin extracted from persimmon. The polysaccharide was low-methoxy pectin with a degree of methyl esterification <50% and ratio of side chain galactan to arabinan in the rhamnogalacturonan-I region of pectin of 3-20. To investigate the physiological function of pectin from persimmon, we performed a coculture assay using Caco-2 cells. As a result, it was shown that the proliferation of undifferentiated Caco-2 cells was promoted, and further, the importance of arabinogalactan among the pectin structures was shown.


Asunto(s)
Diospyros/química , Galactanos/química , Intestino Delgado/metabolismo , Pectinas/química , Células CACO-2 , Células Epiteliales/citología , Humanos , Intestino Delgado/citología
3.
Exp Gerontol ; 142: 111099, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33011215

RESUMEN

Impairment of gastrointestinal function and reduction of nutrient absorption associated with aging contribute to increased risk of malnutrition in the elderly population, resulting in physical weakness and vulnerability to disease. The present study was performed to examine the relationships between aging-associated morphological changes of the small intestine and nutrient malabsorption using senescence-accelerated mouse prone 8 (SAMP8) mice. Comparison of the morphology of the small intestine of young (22-week-old) and senescent (43-week-old) SAMP8 mice showed no significant changes in villus length, while the mRNA expression levels of secretory cell marker genes were significantly reduced in senescent mice. In addition, crypts recovered from the small intestine of senescent mice showed a good capacity to form intestinal organoids ex vivo, suggesting that the regenerative capacity of intestinal stem cells (ISCs) was unaffected by accelerated senescence. These results indicated that changes induced by accelerated senescence in the small intestine of SAMP8 mice are different from changes reported previously in normal aging mouse models. Biochemical analyses of serum before and during senescence also indicated that senescent SAMP8 mice are not in a malabsorption state. Furthermore, a diet supplemented with persimmon pectin had a mild effect on the small intestine of senescent SAMP8 mice. Intestinal villus length was slightly increased in the medial part of the small intestine of pectin-fed mice. In contrast, intestinal crypt formation capacity was enhanced by the pectin diet. Organoid culture derived from the small intestine of mice fed pectin exhibited a greater number of lobes per organoid compared with those from mice fed a control diet, and Lyz1 and Olfm4 mRNA levels were significantly increased. In conclusion, accelerated senescence induced exclusive changes in the small intestine, which were not related to nutrient malabsorption. Therefore, the SAMP8 strain may not be a suitable model to evaluate the effects of aging on intestinal homeostasis and nutrient absorption impairment.


Asunto(s)
Envejecimiento , Pectinas , Anciano , Animales , Dieta Alta en Grasa , Suplementos Dietéticos , Humanos , Intestino Delgado , Ratones
4.
Cell Commun Signal ; 18(1): 117, 2020 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-32727504

RESUMEN

BACKGROUND: Estrogen receptor α (ERα) has been suggested to regulate anti-inflammatory signaling in brain microglia, the only resident immune cells in the brain. ERα conserves the phosphorylation motif at Ser216 within the DNA binding domain. Previously, Ser216 was found to be phosphorylated in neutrophils infiltrating into the mouse uterus and to enable ERα to regulate migration. Given the implication of this phosphorylation in immune regulation, ERα was examined in mouse microglia to determine if Ser216 is phosphorylated and regulates microglia's inflammation. It was found that Ser216 was constitutively phosphorylated in microglia and demonstrated that in the absence of phosphorylated ERα in ERα KI brains microglia inflamed, confirming that phosphorylation confers ERα with anti-inflammatory capability. ERα KI mice were obese and weakened motor ability. METHODS: Mixed glia cells were prepared from brains of 2-days-old neonates and cultured to mature and isolate microglia. An antibody against an anti-phospho-S216 peptide of ERα (αP-S216) was used to detect phosphorylated ERα in double immunofluorescence staining with ERα antibodies and a microglia maker Iba-1 antibody. A knock-in (KI) mouse line bearing the phosphorylation-blocked ERα S216A mutation (ERα KI) was generated to examine inflammation-regulating functions of phosphorylated ERα in microglia. RT-PCR, antibody array, ELISA and FACS assays were employed to measure expressions of pro- or anti-inflammatory cytokines at their mRNA and protein levels. Rotarod tests were performed to examine motor connection ability. RESULTS: Double immune staining of mixed glia cells showed that ERα is phosphorylated at Ser216 in microglia, but not astrocytes. Immunohistochemistry with an anti-Iba-1 antibody showed that microglia cells were swollen and shortened branches in the substantial nigra (SN) of ERα KI brains, indicating the spontaneous activation of microglia as observed with those of lipopolysaccharide (LPS)-treated ERα WT brains. Pro-inflammatory cytokines were up-regulated in the brain of ERα KI brains as well as cultured microglia, whereas anti-inflammatory cytokines were down-regulated. FACS analysis showed that the number of IL-6 producing and apoptotic microglia increased in those prepared from ERα KI brains. Times of ERα KI mice on rod were shortened in Rotarod tests. CONCLUSIONS: Blocking of Ser216 phosphorylation aggravated microglia activation and inflammation of mouse brain, thus confirming that phosphorylated ERα exerts anti-inflammatory functions. ERα KI mice enable us to further investigate the mechanism by which phosphorylated ERα regulates brain immunity and inflammation and brain diseases. Video abstract.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Fosfoserina/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Técnicas de Sustitución del Gen , Ratones , Actividad Motora , Fosforilación , Tiempo de Reacción
5.
Carbohydr Res ; 475: 27-33, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30776756

RESUMEN

An α-l-fucosidase (Pap-Alf) was purified from the pancreas of a starfish Patiria pectinifera by ammonium sulfate precipitation followed by several column chromatographies. The molecular mass of the purified enzyme was estimated to be 52.6 kDa by SDS-PAGE, although gel filtration analysis of the native enzyme suggests it exists as a homodimer in solution. The purified enzyme showed maximal activity at pH 5.0 and 70 °C. The enzyme was highly specific toward a fucosyl-monosaccharide (Fuc-α-pNP), but it also showed activity toward 2-sulfo-Fuc-α-pNP and fucosyl-α-lactosides (Fuc-α-Galß1→4Glc-ß-pNP). We determined the primary structure of the α-l-fucosidase and validated its expression level in starfish tissue. Whole genome sequence analysis of P. pectinifera was also performed in the present study. Detailed primary structural analysis using bioinformatics tools revealed Pap-Alf lacks the C-terminal region that is otherwise conserved in all previously described α-l-fucosidases. Quantitative gene expression analysis of Pap-Alf in each tissue indicated that the expression of Pap-Alf gene in pancreas was 5-fold higher than in ovary.


Asunto(s)
Clonación Molecular , Páncreas/enzimología , alfa-L-Fucosidasa/química , alfa-L-Fucosidasa/genética , Animales , Regulación Enzimológica de la Expresión Génica/genética , Estrellas de Mar , alfa-L-Fucosidasa/metabolismo
6.
Drug Metab Dispos ; 47(1): 38-44, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30389729

RESUMEN

UDP-glucuronosyltransferases (UGTs) catalyze the glucuronidation of numerous endogenous and exogenous compounds to facilitate their excretion from the body. Because rats are commonly used in nonclinical studies, information regarding UGT species differences between rats and humans would be helpful for understanding human pharmacokinetics. In this study, we determined the absolute mRNA expressions of Ugt isoforms in the liver and small intestine of male and female Sprague-Dawley, Fischer 344, and Wistar rats. The sum of the mRNA levels of Ugt isoforms expressed in the liver was significantly (P < 0.005) higher than that in the small intestine regardless of the strain and sex. Ugt2b mRNA levels represented approximately 80% of total Ugt mRNA levels in the liver, whereas Ugt1a mRNA levels accounted for almost 90% in the small intestine. Ugt2b2 mRNA was specifically expressed in Wistar rat liver, resulting in 2-fold higher expression of total hepatic Ugt mRNA in Wistar rats than that in the other strains. Wistar rats showed prominently higher Ugt2b3 and Ugt2b8 mRNA levels in the small intestine than the other strains. The difference between sexes was remarkable with regard to hepatic Ugt1a10 in any of the strains, although slight differences between sexes were also observed in multiple Ugt isoforms. Taken together, this study revealed sex and strain differences in mRNA levels of rat Ugts. The data shown here would be useful for the selection of rat strains in nonclinical studies.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Glucuronosiltransferasa/análisis , Intestino Delgado/metabolismo , Hígado/metabolismo , ARN Mensajero/análisis , Animales , Variación Biológica Poblacional/genética , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Masculino , Modelos Animales , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344/genética , Ratas Sprague-Dawley/genética , Ratas Wistar/genética , Factores Sexuales
7.
Eur J Pharm Sci ; 111: 167-176, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-28966098

RESUMEN

Carboxylesterase (CES) and arylacetamide deacetylase (AADAC) are the major enzymes responsible for the hydrolysis of various clinical drugs. Our recent study demonstrated that the identity of the responsible hydrolase can be roughly surmised based on the chemical structures of compounds in humans. Dogs are used for preclinical studies in drug development, but the substrate specificities of dog CES and AADAC remain to be clarified. The purpose of this study is to characterize their substrate specificities. We prepared recombinant dog CES1, CES2, and AADAC. p-Nitrophenyl acetate, a general substrate for esterases, was hydrolyzed by dog CES1 and AADAC, while it was not hydrolyzed by CES2. CES2 protein was not substantially detected in the recombinant system or in the dog liver and intestinal microsomes by Western blot using anti-human CES2 antibodies. In silico analyses demonstrated slight differences in the three-dimensional structures of dog CES2 and human CES2, indicating that dog CES2 might be unstable or inactive. By evaluating the hydrolase activities of 22 compounds, which are known to be substrates of human CES and/or AADAC, we found that the activities of dog recombinant CES1 and AADAC as well as dog tissue preparations for nearly all compounds were lower than those of human enzymes. The dog enzymes that were responsible for the hydrolysis of most compounds corresponded to the human enzymes, but the following differences were observed: oseltamivir, irinotecan, and rifampicin were not hydrolyzed in the dog liver or by any of the recombinant esterases and procaine, a human CES2 substrate, was hydrolyzed by dog CES1. In conclusion, the present study could provide new finding to facilitate our understanding of species differences in drug hydrolysis, which can facilitate drug development and drug safety evaluation.


Asunto(s)
Carboxilesterasa/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Microsomas/enzimología , Preparaciones Farmacéuticas/metabolismo , Secuencia de Aminoácidos , Animales , Carboxilesterasa/genética , Hidrolasas de Éster Carboxílico/genética , Perros , Humanos , Intestinos/enzimología , Hígado/enzimología , Simulación de Dinámica Molecular , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Células Sf9 , Especificidad de la Especie , Especificidad por Sustrato
8.
Cell Signal ; 40: 200-209, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28911860

RESUMEN

Low glucose stimulated phosphorylation of pregnane X receptor (PXR) at Ser350 in correlation with an increased gluconeogenesis in human hepatoma-derived HepG2 cells. Only glucose, but neither insulin nor glucagon, stimulated this phosphorylation. Here, serine/threonine kinase, vaccinia related kinase 1 (VRK1)-mediated phosphorylation of PXR is now defined as this glucose-elicited novel signal. In low glucose conditions, VRK1 directly phosphorylates PXR at Ser350, enabling PO3-PXR to scaffold protein phosphatase PP2Cα. This PP2Cα dephosphorylates serine/threonine kinase 2 (SGK2) at Thr193. This dephosphorylation dissociates SGK2 from and actives the phosphoenolpyruvate carboxykinase 1 (PCK1) gene as phosphorylated SGK2 binds and represses the gene. Conversely, VRK1 self-represses its activity to phosphorylate PXR through cyclin-dependent kinase 2 (CDK2) in high glucose conditions, resulting in the repression of the PCK1 gene. This PXR phosphorylation was also observed in fasting mouse livers. Thus, the VRK1-CDK2-PXR-PP2Cα-SGK2 pathway can be a novel physiological cell signaling that regulates gluconeogenesis in response to glucose.


Asunto(s)
Glucosa/metabolismo , Proteínas Inmediatas-Precoces/genética , Proteína Fosfatasa 2C/genética , Proteínas Serina-Treonina Quinasas/genética , Receptores de Esteroides/genética , Animales , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Ayuno , Gluconeogénesis/genética , Células Hep G2 , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Hígado/metabolismo , Ratones , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Fosforilación , Receptor X de Pregnano , Unión Proteica , Proteína Fosfatasa 2C/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Esteroides/metabolismo , Transducción de Señal/genética
9.
Biochem Pharmacol ; 140: 150-160, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28606603

RESUMEN

Nitrazepam (NZP) is a hypnotic agent that rarely causes liver injuries in humans and teratogenicity in rodents. In humans, NZP is primarily metabolized to 7-aminonitrazepam (ANZP) by reduction and subsequently to 7-acetylamino nitrazepam (AANZP) by acetylation. ANZP can be regenerated from AANZP by hydrolysis in rodents, but it is still unclear whether this reaction occurs in humans. In rodents, AANZP may be associated with teratogenicity, while in humans, it is known that drug-induced liver injuries may be caused by NZP reactive metabolite(s). In this study, we attempted to identify the enzymes responsible for NZP metabolism to obtain a basic understanding of this process and the associated metabolite toxicities. We found that the NZP reductase activity in human liver cytosol (HLC) was higher than that in human liver microsomes (HLM). We purified the responsible enzyme(s) from HLC and found that the NZP reductase was aldehyde oxidase 1 (AOX1). The role of AOX1 was confirmed by an observed increase in the NZP reductase activity upon addition of N1-methylnicotinamide, an electron donor of AOX1, as well as inhibition of this activity in HLC in the presence of AOX1 inhibitors. ANZP was acetylated to form AANZP by N-acetyltransferase (NAT) 2. An experiment using recombinant esterases in an inhibition study using HLM revealed that AANZP is hydrolyzed by arylacetamide deacetylase (AADAC) in the human liver. N-Hydroxylamino NZP, which is suspected to be a reactive metabolite, was detected as a conjugate with N-acetyl-l-cysteine through NZP reduction and ANZP hydroxylation reactions. In the latter reaction, the conjugate was readily formed by recombinant CYP3A4 among the various P450 isoforms tested. In sum, we found that AOX1, NAT2, AADAC, and CYP3A4 are the determinants for the pharmacokinetics of NZP and that they confer interindividual variability in sensitivity to NZP side effects.


Asunto(s)
Aldehído Oxidasa/metabolismo , Arilamina N-Acetiltransferasa/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Citocromo P-450 CYP3A/metabolismo , Hepatocitos/metabolismo , Hipnóticos y Sedantes/metabolismo , Nitrazepam/metabolismo , Acetilación , Aldehído Oxidasa/antagonistas & inhibidores , Aldehído Oxidasa/química , Aldehído Oxidasa/aislamiento & purificación , Arilamina N-Acetiltransferasa/genética , Biotransformación , Hidrolasas de Éster Carboxílico/genética , Citocromo P-450 CYP3A/genética , Citosol/enzimología , Citosol/metabolismo , Inhibidores Enzimáticos/farmacología , Hepatocitos/enzimología , Humanos , Hidrólisis/efectos de los fármacos , Hidroxilación , Hipnóticos y Sedantes/efectos adversos , Cinética , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Nitrazepam/efectos adversos , Nitrazepam/análogos & derivados , Oxidación-Reducción , Proteínas Recombinantes/metabolismo
10.
J Biol Chem ; 292(12): 4873-4884, 2017 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-28188287

RESUMEN

Dihydrofolate reductase (DHFR) plays a key role in folate metabolism and is a target molecule of methotrexate. An increase in the cellular expression level of DHFR is one of the mechanisms of tumor resistance to methotrexate. The present study investigated the possibility that adenosine-to-inosine RNA editing, which causes nucleotide conversion by adenosine deaminase acting on RNA (ADAR) enzymes, might modulate DHFR expression. In human breast adenocarcinoma-derived MCF-7 cells, 26 RNA editing sites were identified in the 3'-UTR of DHFR. Knockdown of ADAR1 decreased the RNA editing levels of DHFR and resulted in a decrease in the DHFR mRNA and protein levels, indicating that ADAR1 up-regulates DHFR expression. Using a computational analysis, miR-25-3p and miR-125a-3p were predicted to bind to the non-edited 3'-UTR of DHFR but not to the edited sequence. The decrease in DHFR expression by the knockdown of ADAR1 was restored by transfection of antisense oligonucleotides for these miRNAs, suggesting that RNA editing mediated up-regulation of DHFR requires the function of these miRNAs. Interestingly, we observed that the knockdown of ADAR1 decreased cell viability and increased the sensitivity of MCF-7 cells to methotrexate. ADAR1 expression levels and the RNA editing levels in the 3'-UTR of DHFR in breast cancer tissues were higher than those in adjacent normal tissues. Collectively, the present study demonstrated that ADAR1 positively regulates the expression of DHFR by editing the miR-25-3p and miR-125a-3p binding sites in the 3'-UTR of DHFR, enhancing cellular proliferation and resistance to methotrexate.


Asunto(s)
Adenosina Desaminasa/genética , Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Edición de ARN , Proteínas de Unión al ARN/genética , Tetrahidrofolato Deshidrogenasa/genética , Regiones no Traducidas 3' , Adenosina/genética , Mama/metabolismo , Proliferación Celular , Femenino , Humanos , Inosina/genética , Células MCF-7 , MicroARNs/genética , Regulación hacia Arriba
11.
J Biol Chem ; 291(2): 894-903, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26601943

RESUMEN

Adenosine to inosine (A-to-I) RNA editing is the most frequent type of post-transcriptional nucleotide conversion in humans, and it is catalyzed by adenosine deaminase acting on RNA (ADAR) enzymes. In this study we investigated the effect of RNA editing on human aryl hydrocarbon receptor (AhR) expression because the AhR transcript potentially forms double-stranded structures, which are targets of ADAR enzymes. In human hepatocellular carcinoma-derived Huh-7 cells, the ADAR1 knockdown reduced the RNA editing levels in the 3'-untranslated region (3'-UTR) of the AhR transcript and increased the AhR protein levels. The ADAR1 knockdown enhanced the ligand-mediated induction of CYP1A1, a gene downstream of AhR. We investigated the possibility that A-to-I RNA editing creates miRNA targeting sites in the AhR mRNA and found that the miR-378-dependent down-regulation of AhR was abolished by ADAR1 knockdown. These results indicated that the ADAR1-mediated down-regulation of AhR could be attributed to the creation of a miR-378 recognition site in the AhR 3'-UTR. The interindividual differences in the RNA editing levels within the AhR 3'-UTR in a panel of 32 human liver samples were relatively small, whereas the differences in ADAR1 expression were large (220-fold). In the human liver samples a significant inverse association was observed between the miR-378 and AhR protein levels, suggesting that the RNA-editing-dependent down-regulation of AhR by miR-378 contributes to the variability in the constitutive hepatic expression of AhR. In conclusion, this study uncovered for the first time that A-to-I RNA editing modulates the potency of xenobiotic metabolism in the human liver.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hígado/metabolismo , MicroARNs/genética , Edición de ARN/genética , Receptores de Hidrocarburo de Aril/genética , Regiones no Traducidas 3'/genética , Adenosina Desaminasa/metabolismo , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo/genética , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , MicroARNs/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Activación Transcripcional
12.
Sci Rep ; 5: 14076, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26392083

RESUMEN

Statin therapy is known to increase blood glucose levels in humans. Statins utilize pregnane X receptor (PXR) and serum/glucocorticoid regulated kinase 2 (SGK2) to activate phosphoenolpyruvate carboxykinase 1 (PEPCK1) and glucose-6-phosphatase (G6Pase) genes, thereby increasing glucose production in human liver cells. Here, the novel statin/PXR/SGK2-mediated signaling pathway has now been characterized for hepatic gluconeogenesis. Statin-activated PXR scaffolds the protein phosphatase 2C (PP2C) and SGK2 to stimulate PP2C to dephosphorylate SGK2 at threonine 193. Non-phosphorylated SGK2 co-activates PXR-mediated trans-activation of promoters of gluconeogenic genes in human liver cells, thereby enhancing gluconeogenesis. This gluconeogenic statin-PXR-SGK2 signal is not present in mice, in which statin treatment suppresses hepatic gluconeogenesis. These findings provide the basis for statin-associated side effects such as an increased risk for Type 2 diabetes.


Asunto(s)
Gluconeogénesis/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Proteínas Inmediatas-Precoces/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Esteroides/agonistas , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Gluconeogénesis/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Ratones , Modelos Biológicos , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Receptor X de Pregnano , Regiones Promotoras Genéticas , Unión Proteica , Proteína Fosfatasa 2C , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Transcripción Genética
13.
J Pharmacol Exp Ther ; 348(1): 131-40, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24204015

RESUMEN

Drug activation of the human nuclear pregnane X receptor (PXR) induced gluconeogenic genes and increased glucose production. In this study, we have determined that serum- and glucocorticoid-regulated kinase 2 (SGK2) is an essential factor that mediates this PXR-regulated glucose 6-phosphatase (G6Pase) induction and glucose production. Both SGK2 and G6Pase mRNAs were increased in rifampicin-treated HepG2 cells stably expressing human PXR. Reporter and chromatin immunoprecipitation assays delineated PXR activation of the SGK2 gene to a distal and proximal DNA sequence within its promoter: distal PXR response element (-2587/-2209) and proximal PXR response element (-115/-75), respectively. Small interfering RNA (siRNA) knockdown of SGK2 severely attenuated PXR-regulated induction of G6Pase as well as glucose production. SGK2 constitutes an insulin-independent signal pathway to regulate gluconeogenesis because siRNA knockdown of the insulin-responsive transcription factor forkhead box protein O1 did not affect rifampicin induction of G6Pase. Rifampicin treatment of two different samples of human primary hepatocytes revealed that PXR induces G6Pase in the presence of high levels of SGK2, whereas PXR represses G6Pase in its absence. Mediating PXR activation of the G6Pase gene is the first biological role found for hepatic SGK2 and might have therapeutic implications for side effects, such as diabetes, caused by drugs that activate PXR.


Asunto(s)
Gluconeogénesis/fisiología , Hepatocitos/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Esteroides/agonistas , Receptores de Esteroides/metabolismo , Animales , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Gluconeogénesis/efectos de los fármacos , Células Hep G2 , Hepatocitos/efectos de los fármacos , Humanos , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Receptor X de Pregnano , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Rifampin/farmacología
14.
Exp Eye Res ; 112: 45-50, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23631845

RESUMEN

The Nakano cataract (NCT) is a recessive disorder in the mouse linked to the nct locus on chromosome 16. In this study, we positionally cloned the critical gene in the nct locus. Herein, we report that cataracts in the BALB/c-nct/nct mouse are caused by a hypomorphic mutation in the coproporphyrin oxidase gene (Cpox), encoding the enzyme responsible for catalyzing oxidative decarboxylation of the heme precursor, coproporphyrinogen III, in the heme biosynthetic pathway. BALB/c-nct/nct mice are homozygous for a G to T nucleotide substitution in the Cpox gene, which results in a p.R380L amino acid substitution in the CPOX protein. The CPOX isoform with the p.R380L substitution retained only 15% of the activity of the wild type isoform. BALB/c-nct/nct mice had excessive accumulation of coproporphyrin III in the lens. The NCT phenotype was normalized by the introduction of a wild type Cpox transgene. The mechanisms by which impairment of CPOX leads to lens opacity in the NCT are elusive. However, our data illuminate a hitherto unanticipated involvement of the heme biosynthesis pathway in lens physiology.


Asunto(s)
Catarata/genética , Coproporfirinógeno Oxidasa/genética , Modelos Animales de Enfermedad , Enfermedades Hereditarias del Ojo/genética , Mutación Missense , Sustitución de Aminoácidos , Animales , Catarata/metabolismo , Coproporfirinógeno Oxidasa/metabolismo , Coproporfirinas/metabolismo , Enfermedades Hereditarias del Ojo/metabolismo , Femenino , Hemo/metabolismo , Homocigoto , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Ratones Transgénicos , Fenotipo , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética
15.
Photochem Photobiol ; 87(5): 1138-45, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21668870

RESUMEN

δ-Aminolevulinic acid (ALA)-induced porphyrin accumulation is widely used in the treatment of cancer, as photodynamic therapy. To clarify the mechanisms of the tumor-preferential accumulation of protoporphyrin, we examined the effect of the expression of heme-biosynthetic and -degradative enzymes on the ALA-induced accumulation of protoporphyrin as well as photodamage. The transient expression of heme-biosynthetic enzymes in HeLa cells caused variations of the ALA-induced accumulation of protoporphyrin. When ALA-treated cells were exposed to white light, the extent of photodamage of the cells was dependent on the accumulation of protoporphyrin. The decrease of the accumulation of protoporphyrin was observed in the cells treated with inducers of heme oxygenase (HO)-1. The ALA-dependent accumulation of protoporphyrin was decreased in HeLa cells by transfection with HO-1 and HO-2 cDNA. Conversely, knockdown of HO-1/-2 with siRNAs enhanced the ALA-induced protoporphyrin accumulation and photodamage. The ALA effect was decreased with HeLa cells expressing mitoferrin-2, a mitochondrial iron transporter, whereas it was enhanced by the mitoferrin-2 siRNA transfection. These results indicated that not only the production of porphyrin intermediates but also the reuse of iron from heme and mitochondrial iron utilization control the ALA-induced accumulation of protoporphyrin in cancerous cells.


Asunto(s)
Ácido Aminolevulínico/metabolismo , Hierro/metabolismo , Fotoquimioterapia , Fármacos Fotosensibilizantes/metabolismo , Protoporfirinas/biosíntesis , Neoplasias del Cuello Uterino/tratamiento farmacológico , Ácido Aminolevulínico/farmacología , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Femenino , Silenciador del Gen/efectos de los fármacos , Células HeLa , Hemo/metabolismo , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Luz , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Fármacos Fotosensibilizantes/farmacología , Plásmidos , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
16.
Gene ; 472(1-2): 28-36, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21044876

RESUMEN

δ-Aminolevulinic acid synthase 1 (ALAS1) is the first and rate-limiting enzyme in the heme biosynthesis. It has been well known that heme exerts a negative feedback control over the transcription of ALAS1 gene to maintain intracellular heme at appropriate level. To clarify the mechanisms by which heme regulates the expression of ALAS1, we examined the promoter activity of the gene and identified the heme-responsive element (HRE) located in the proximal promoter of the mouse ALAS1 gene. Reporter and EMSA assays revealed the sequence (GCGGGGGCG), as the site of repression by heme, at -301/-293bp of the ALAS1 promoter. Subsequently, EMSA and ChIP assays showed that a transcription factor, early growth response 1 (Egr-1) and its major corepressors, NAB1 and NAB2 were found to bind to the ALAS1-HRE, and these bindings increased dependent on the level of intracellular heme. When Egr-1 and NAB1 in combination were expressed in the cells, decreases of the level of ALAS1 mRNA and intracellular level of heme were observed. These results suggest that Egr-1-NABs complex is involved in the regulation of the transcription of ALAS1 by heme, leading to the regulation of the heme biosynthesis.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Regulación Enzimológica de la Expresión Génica , Hemo/metabolismo , Proteínas Represoras/metabolismo , 5-Aminolevulinato Sintetasa/metabolismo , Animales , Células Cultivadas , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Ensayo de Cambio de Movilidad Electroforética , Ratones , Células 3T3 NIH , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , ARN Mensajero/metabolismo , Proteínas Represoras/genética
17.
Gene ; 423(2): 207-14, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18675890

RESUMEN

Heme acts as a ligand for transcription factors and regulates the expression of several genes. The nuclear receptor retinoid X receptor alpha (RXRalpha) plays important roles in various nuclear receptor-dependent signaling pathways. We here show that heme binds to RXRalpha and impairs its DNA-binding activity. Deletion and mutation studies of RXRalpha revealed that the binding region of hemin corresponded to the ligand binding domain of mouse RXRalpha and cysteine 374 was involved in the binding. The DNA-binding activity using the DR-1 consensus sequence of RXRalpha in electrophoretic mobility shift assays was inhibited by heme. The reporter assay also showed a decrease of RXRalpha-dependent transcriptional activity. It was reported that hemin enhanced the adipocyte differentiation of mouse 3T3-L1 cells, where the functions of several nuclear receptors including RXRalpha and peroxisome proliferator-activated receptor-gamma (PPAR-gamma) are activated. However, the inductions of adipogenic factor mRNAs including PPAR-gamma, fatty acid binding protein-4 and glucose transporter-4 were markedly repressed by heme during adipocyte differentiation. These results suggest that heme causes the impairment of RXRalpha-dependent signal pathways and inhibits the adipocyte differentiation of 3T3-L1 cells.


Asunto(s)
Hemo/metabolismo , Receptor alfa X Retinoide/genética , Receptor alfa X Retinoide/metabolismo , Transcripción Genética , Células 3T3-L1 , Adipocitos/citología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Diferenciación Celular , Núcleo Celular/metabolismo , ADN/metabolismo , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica , Hemina/metabolismo , Ratones , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Receptor alfa X Retinoide/química , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...