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










Base de datos
Intervalo de año de publicación
1.
Ann Clin Biochem ; : 45632231213746, 2023 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-37903217

RESUMEN

BACKGROUND: Delayed time from collection to centrifugation may cause erroneously high lactate levels in vitro (from continued blood cell metabolism under anaerobic conditions in the collection tube) if not collected in appropriate collection devices, consequently increasing the risk for inappropriate patient care or harm. We undertook a study to determine the turnaround time for lactate testing in a tertiary care setting and also performed short- and long-term lactate stability studies in blood collected in sodium fluoride/potassium oxalate (NaF/KOx) collection tubes. METHODS: The hospital lab information system was mined for 6 months to determine patient samples that may have exceeded the time from collection-to-receival in lab of 15-min. Lactate stability was evaluated in unspun NaF/KOx collection tubes at 15 min intervals for to 2 h; and separately at 2, 6, 12, 24, and 48-h post-collection. RESULTS: A total of 8,929 plasma samples were collected in 6 months, and 1/3 were not received in the lab within 15 min from collection. In NaF/KOx additive, lactate levels had minor increases over 2 h, and incremental increases at an average rate of 0.0035 mmol/L/h over 48 h with maximum increase of 9.8% at 48 h. However, the average change across all time points were within local allowable performance goals (at ≤4 mmol/L ± 0.5 mmol/L; at >4 mmol/L ± 12%). CONCLUSION: A small proportion of lactate specimens may experience delay in processing. Although lactate levels may incrementally increase over 48-h at room temperature in unspun NaF/KOx collection tubes, the changes may not be clinically impactful.

2.
Front Oncol ; 13: 1217847, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37746289

RESUMEN

Phenobarbital (PB) is an archetypal substance used as a mouse hepatocellular carcinoma (HCC) promotor in established experimental protocols. Our previous results showed CAR is the essential factor for PB induced HCC promotion. Subsequent studies suggested Gadd45ß, which is induced by PB through CAR activation, is collaborating with CAR to repress TNF-α induced cell death. Here, we used Gadd45ß null mice (Gadd45ß KO) treated with N-diethylnitrosamine (DEN) at 5 weeks of age and kept the mice with PB supplemented drinking water from 7 to 57 weeks old. Compared with wild type mice, Gadd45ß KO mice developed no HCC in the PB treated group. Increases in liver weight were more prominent in wild type mice than KO mice. Microarray analysis of mRNA derived from mouse livers found multiple genes specifically up or down regulated in wild type mice but not null mice in DEN + PB groups. Further qPCR analysis confirmed two genes, Tgfbr2 and irisin/Fndc5, were up-regulated in PB treated wild type mice but no significant increase was observed in Gadd45ß KO mice. We focused on these two genes because previous reports showed that hepatic Irisin/Fndc5 expression was significantly higher in HCC patients and that irisin binds to TGF-ß receptor complex that includes TGFBR2 subunit. Our results revealed irisin peptide in cell culture media increased the growth rate of mouse hepatocyte-derived AML12 cells. Microarray analysis revealed that irisin-regulated genes in AML12 cells showed a significant association with the genes in the TGF-ß pathway. Expression of irisin/Fndc5 and Tgfbr2 induced growth of human HCC cell line HepG2. Thus, Gadd45ß plays an indispensable role in mouse HCC development regulating the irisin/Fndc5 and Tgfbr2 genes.

3.
Methods Mol Biol ; 2418: 63-75, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35119660

RESUMEN

Serine 216 constitutes a protein kinase C phosphorylation motif located within the DNA binding domain of estrogen receptor α (ERα). In this chapter, we present experimental procedures confirming that mouse ERα is phosphorylated at serine 216 in peripheral blood neutrophils and in neutrophils that infiltrate the uterus, as well as the role of phosphoserine 216 in neutrophil migration. A phospho-peptide antibody (αP-S216) was utilized in Western blot, immunohistochemistry, and double immunofluorescence staining to detect this phosphorylation of an endogenous ERα. Both immunohistochemistry (with αP-S216 or neutrophil marker Ly6G antibody) and double immunofluorescence staining of mouse uterine sections prepared from C3H/HeNCrIBR females revealed that phosphorylated ERα was expressed in all infiltrating neutrophils during hormonal cycles but not in any other of the other uterine cells. Neutrophils infiltrate the uterus from the bloodstream. White blood cells (WBC) were prepared from peripheral blood of C3H/HeNCrIBR females or males and double immunostained. Blood neutrophils also expressed phosphorylated ERα but in only about 20% of cells in both sexes. Only the neutrophils expressing phosphorylated ERα spontaneously migrated in in vitro Transwell migration assays and infiltrated the uterus in mice.


Asunto(s)
Receptor alfa de Estrógeno , Serina , Animales , Receptor alfa de Estrógeno/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C3H , Neutrófilos/metabolismo , Fosforilación , Serina/metabolismo
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.
Sci Rep ; 10(1): 5001, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32193417

RESUMEN

Estrogen sulfotransferase (SULT1E1) inactivates estrogen and regulates its metabolic homeostats. Whereas SULT1E1 is expressed low in the liver of adult mice, it is induced by phenobarbital (PB) treatment or spontaneously in diabetic livers via nuclear receptors. Utilizing constitutive active/androstane receptor (CAR) KO, estrogen receptor α (ERα KO, phosphorylation-blocked ERα S216A KI mice, it is now demonstrated that, after being activated by PB, CAR binds and recruits ERα onto the Sulte1 promoter for subsequent phosphorylation at Ser216. This phosphorylation tightens CAR interacting with ERα and to activates the promoter. Hepatic SULT1E1 mRNA levels are constitutively up-regulated in type 1 diabetic Akita mice; CAR spontaneously accumulates in the nucleus and activates the Sult1e1 promoter by recruiting phosphorylated ERα in the liver as observed with PB-induced livers. Thus, this CAR-phosphorylated ERα signaling enables these two nuclear receptors to communicate, activating the Sult1e1 gene in response to either PB or diabetes in mice. ERα phosphorylation may integrate CAR into estrogen actions, providing insights into understanding drug-hormone interactions in clinical therapy.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Regulación Enzimológica de la Expresión Génica/genética , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Sulfotransferasas/metabolismo , Animales , Línea Celular Tumoral , Receptor de Androstano Constitutivo , Humanos , Ratones , Fenobarbital/metabolismo , Fosforilación , Sulfotransferasas/genética
6.
Lab Invest ; 99(10): 1470-1483, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31152145

RESUMEN

Retinoid X receptor α (RXRα) has a conserved phosphorylation motif at threonine 162 (humans) and threonine 167 (mice) within the DNA-binding domain. Here we have generated RXRα knock-in mice (RxrαT167A) bearing a single mutation of Thr 167 to alanine and examined the roles of Thr 167 in the regulation of energy metabolism within adipose, muscle, and liver tissues. RxrαT167A mice exhibited down-regulation of metabolic pathways converting glucose to fatty acids, such as acetyl-CoA carboxylase in the white adipose tissue (WAT) and ATP citrate lyase in the muscle. They also reduced gene expression for genes related to fatty acid catabolism and triglyceride synthesis in WAT and controlled heat factors such as adrenergic receptor ß1 in muscles. In contrast, hepatic gluconeogenic pathways and synthetic pathways related to fatty acids remained unaffected by this mutation. Expression of multiple genes that were affected by the Thr 167 mutation in adipose tissue exhibited clear response to LG100268, a synthetic RXR agonist. Thus, the altered gene expression in mutant mice adipose appeared to be a direct effect of RXRα Thr 167 mutation and by some secondary effect of the mutation. Blood glucose levels remained normal in RxrαT167A during feeding, as observed with RXRα wild-type mice. However, RxrαT167A mice exhibited an attenuated decrease of blood glucose levels that occurred after fasting. This attenuation correlated with a concomitant down-regulation of lipid metabolism in WAT and was associated with RXRα phosphorylation at Thr 167. Thus, Thr 167 enabled RXRα to coordinate these three organs for regulation of energy metabolism and maintenance of glucose homeostasis.


Asunto(s)
Metabolismo Energético/genética , Privación de Alimentos/fisiología , Receptor alfa X Retinoide/genética , Animales , Glucemia/genética , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , ADN/metabolismo , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones , Fosforilación , Receptor alfa X Retinoide/metabolismo
7.
Shock ; 50(2): 248-254, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30010631

RESUMEN

Co-chaperone cytoplasmic constitutive active/androstane receptor retention protein (CCRP), a member of heat shock protein (HSP) 40, was first characterized to retain a nuclear-destined protein in the cytoplasm. Here we have used CCRP KO mice and demonstrated that CCRP suppresses lipopolysaccharide (LPS)-induced cardiac toxicity in mice. LPS treatment decreased heart rates in CCRP KO mice, but not in wild-type (WT) mice. In addition, LPS-treated KO mice showed reduced fraction shortening, an indicator of ventricular contractile function, to a greater degree than WT mice did. Rat cardiomyocyte-derived H9c2 cells, in which CCRP is not expressed, were used to examine a cell signal through which CCRP suppressed LPS-induced cardiac toxicity. Overexpression of CCRP prevented p65, a nuclear factor κB (NFκB) subunit, from accumulating in the nucleus after LPS treatment. As observed with H9c2 cells, nuclear accumulation of p65 was found to be higher in the hearts of KO mice than WT mice after LPS treatment. Furthermore, induction of TNFα by LPS was markedly suppressed by CCRP in H9c2 cells as well as in LPS-treated mouse serum. In supporting the notion that CCRP repressed the LPS-induced NFκB signaling, pretreatment with pyrrolidinedithiocarbamate, an NFκB signaling inhibitor, or anti-TNF-α antibody before LPS treatment restored heart rates decreased in KO mice after LPS treatment in a dose-dependent manner. Our present study characterized a novel physiological role of CCRP in protecting cardiac functions through the inhibition of NFκB signaling.


Asunto(s)
Cardiotoxicidad/metabolismo , Lipopolisacáridos/toxicidad , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Animales , Cardiotoxicidad/genética , Cardiotoxicidad/patología , Línea Celular , Proteínas de Choque Térmico , Ratones , Ratones Noqueados , Chaperonas Moleculares , Ratas , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal/genética , Factor de Transcripción ReIA/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
8.
FEBS Lett ; 592(16): 2760-2768, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30025153

RESUMEN

The estrogen sulfotransferase SULT1E1 sulfates and inactivates estrogen, which is reactivated via desulfation by steroid sulfatase, thus regulating estrogen homeostasis. Phenobarbital (PB), a clinical sedative, activates Sult1e1 gene transcription in mouse livers. Here, the molecular mechanism by which the nuclear receptors CAR, which is targeted by PB, and RORα communicate through phosphorylation to regulate Sult1e1 activation has been studied. RORα, a basal activity repressor of the Sult1e1 promoter, becomes phosphorylated at serine 100 and converts to an activator of the Sult1e1 promoter in response to PB. CAR regulates both the RORα phosphorylation and conversion. Our findings suggest that PB signals CAR to communicate with RORα via serine 100 phosphorylation, converting RORα from transcription repressor to activator of the Sult1e1 gene and inducing SULT1E1 expression in mouse livers.


Asunto(s)
Hipnóticos y Sedantes/farmacología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Fenobarbital/farmacología , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Sulfotransferasas/genética , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Receptor de Androstano Constitutivo , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Hígado/metabolismo , Ratones , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/química , Fosforilación , Regiones Promotoras Genéticas/efectos de los fármacos , Serina/metabolismo , Activación Transcripcional
9.
Mol Cancer Res ; 16(8): 1309-1318, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29716964

RESUMEN

Phenobarbital, a nongenotoxic hepatocarcinogen, induces hepatic proliferation and promotes development of hepatocellular carcinoma (HCC) in rodents. Nuclear receptor constitutive active/androstane receptor (NR1I3/CAR) regulates the induction and promotion activities of phenobarbital. Here, it is demonstrated that phenobarbital treatment results in dephosphorylation of a tumor suppressor p38 MAPK in the liver of C57BL/6 and C3H/HeNCrlBR mice. The molecular mechanism entails CAR binding and inhibition of the growth arrest and DNA-damage-inducible 45 beta (GADD45B)-MAPK kinase 6 (MKK6) scaffold to repress phosphorylation of p38 MAPK. Phenobarbital-induced hepatocyte proliferation, as determined by BrdUrd incorporation, was significantly reduced in both male and female livers of GADD45B knockout (KO) mice compared with the wild-type mice. The phenobarbital-induced proliferation continued until 48 hours after phenobarbital injection in only the C57BL/6 males, but neither in males of GADD45B KO mice nor in females of C57BL/6 and GADD45B KO mice. Thus, these data reveal nuclear receptor CAR interacts with GADD45B to repress p38 MAPK signaling and elicit hepatocyte proliferation in male mice.Implications: This GADD45B-regulated male-predominant proliferation can be expanded as a phenobarbital promotion signal of HCC development in future studies.Visual Overview: http://mcr.aacrjournals.org/content/molcanres/16/8/1309/F1.large.jpg Mol Cancer Res; 16(8); 1309-18. ©2018 AACR.


Asunto(s)
Anticonvulsivantes/efectos adversos , Antígenos de Diferenciación/genética , Proliferación Celular/efectos de los fármacos , Hígado/patología , Fenobarbital/efectos adversos , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Receptor de Androstano Constitutivo , Ratones , Ratones Endogámicos C57BL
10.
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
11.
Mol Cell Biol ; 37(10)2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28265001

RESUMEN

The nuclear receptor CAR (NR1I3) regulates hepatic drug and energy metabolism as well as cell fate. Its activation can be a critical factor in drug-induced toxicity and the development of diseases, including diabetes and tumors. CAR inactivates its constitutive activity by phosphorylation at threonine 38. Utilizing receptor for protein kinase 1 (RACK1) as the regulatory subunit, protein phosphatase 2A (PP2A) dephosphorylates threonine 38 to activate CAR. Here we demonstrate that CAR undergoes homodimer-monomer conversion to regulate this dephosphorylation. By coexpression of two differently tagged CAR proteins in Huh-7 cells, mouse primary hepatocytes, and mouse livers, coimmunoprecipitation and two-dimensional gel electrophoresis revealed that CAR can form a homodimer in a configuration in which the PP2A/RACK1 binding site is buried within its dimer interface. Epidermal growth factor (EGF) was found to stimulate CAR homodimerization, thus constraining CAR in its inactive form. The agonistic ligand CITCO binds directly to the CAR homodimer and dissociates phosphorylated CAR into its monomers, exposing the PP2A/RACK1 binding site for dephosphorylation. Phenobarbital, which is not a CAR ligand, binds the EGF receptor, reversing the EGF signal to monomerize CAR for its indirect activation. Thus, the homodimer-monomer conversion is the underlying molecular mechanism that regulates CAR activation, by placing phosphorylated threonine 38 as the common target for both direct and indirect activation of CAR.


Asunto(s)
Hepatocitos/metabolismo , Neuropéptidos/metabolismo , Proteína Fosfatasa 2/metabolismo , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Células Cultivadas , Receptor de Androstano Constitutivo , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Humanos , Ligandos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fosforilación , Multimerización de Proteína , Receptores de Cinasa C Activada
12.
Drug Metab Dispos ; 44(6): 871-6, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27074912

RESUMEN

Nuclear receptor constitutive androstane receptor (CAR, NR1I3), which regulates hepatic drug and energy metabolisms as well as cell growth and death, is sequestered in the cytoplasm as its inactive form phosphorylated at threonine 38. CAR activators elicit dephosphorylation, and nonphosphorylated CAR translocates into the nucleus to activate its target genes. CAR was previously found to require p38 mitogen-activated protein kinase (MAPK) to transactivate the cytochrome P450 2B (CYP2B) genes. Here we have demonstrated that p38 MAPK forms a complex with CAR, enables it to bind to the response sequence, phenobarbital-responsive enhancer module (PBREM), within the CYP2B promoter, and thus recruits RNA polymerase II to activate transcription. Subsequently, p38 MAPK elicited rephosphorylation of threonine 38 to inactivate CAR and exclude it from the nucleus. Thus, nuclear p38 MAPK exerted dual regulation by sequentially activating and inactivating CAR-mediated transcription through phosphorylation of threonine 38.


Asunto(s)
Núcleo Celular/metabolismo , Fosforilación/fisiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Treonina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Hidrocarburo de Aril Hidroxilasas , Línea Celular , Núcleo Celular/efectos de los fármacos , Receptor de Androstano Constitutivo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C3H , Fenobarbital/farmacología , Fosforilación/efectos de los fármacos , Factores de Transcripción/metabolismo
13.
J Pharmacol Exp Ther ; 357(2): 367-74, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26994072

RESUMEN

Phenobarbital (PB) antagonized insulin to inactivate the insulin receptor and attenuated the insulin receptor downstream protein kinase B (AKT)-forkhead box protein O1 and extracellular signal-regulated kinase 1/2 signals in mouse primary hepatocytes and HepG2 cells. Hepatic AKT began dephosphorylation in an early stage of PB treatment, and blood glucose levels transiently increased in both wild-type and constitutive androstane receptor (CAR) knockout (KO) mice. On the other hand, blood glucose levels increased in wild-type mice, but not KO mice, in later stages of PB treatment. As a result, PB, acting as an insulin receptor antagonist, elicited CAR-independent increases and CAR-dependent decreases of blood glucose levels at these different stages of treatment, respectively. Reciprocally, insulin activation of the insulin receptor repressed CAR activation and induction of its target CYP2B6 gene in HepG2 cells. Thus, PB and insulin cross-talk through the insulin receptor to regulate glucose and drug metabolism reciprocally.


Asunto(s)
Hipoglucemiantes/farmacología , Insulina/farmacología , Fenobarbital/farmacología , Receptor de Insulina/efectos de los fármacos , Receptores Acoplados a Proteínas G/agonistas , Animales , Hidrocarburo de Aril Hidroxilasas/metabolismo , Glucemia/metabolismo , Citocromo P-450 CYP2B6/efectos de los fármacos , Familia 2 del Citocromo P450 , Hepatocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Cross-Talk/efectos de los fármacos , Receptores Sensibles al Calcio , Receptores Acoplados a Proteínas G/genética , Esteroide Hidroxilasas/metabolismo , Transfección
14.
Methods Mol Biol ; 1366: 413-424, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26585153

RESUMEN

Serine 216 constitutes a protein kinase C phosphorylation motif located within the DNA binding domain of estrogen receptor α (ERα). In this chapter we present experimental procedures confirming that mouse ERα is phosphorylated at serine 216 in peripheral blood neutrophils and in neutrophils that infiltrate the uterus, as well as the role of phosphoserine 216 in neutrophil migration. A phospho-peptide antibody (αP-S216) was utilized in Western blot, immunohistochemistry, and double immunofluorescence staining to detect this phosphorylation of an endogenous ERα. Both immunohistochemistry (with αP-S216 or neutrophil marker Ly6G antibody) and double immunofluorescence staining of mouse uterine sections prepared from C3H/HeNCrIBR females revealed that phosphorylated ERα was expressed in all infiltrating neutrophils during hormonal cycles but not in any other of the other uterine cells. Neutrophils infiltrate the uterus from the blood stream. White blood cells (WBC) were prepared from peripheral blood of C3H/HeNCrIBR females or males and double immunostained. Blood neutrophils also expressed phosphorylated ERα but in only about 20 % of cells in both sexes. Only the neutrophils expressing phosphorylated ERα spontaneously migrated in in vitro Transwell migration assays and infiltrated the uterus in mice.


Asunto(s)
Movimiento Celular , Receptor alfa de Estrógeno/metabolismo , Infiltración Neutrófila , Neutrófilos/metabolismo , Útero/metabolismo , Animales , Western Blotting , Ensayos de Migración Celular , Receptor alfa de Estrógeno/sangre , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C3H , Fosforilación , Proteína Quinasa C/metabolismo , Serina , Transducción de Señal , Flujo de Trabajo
15.
PLoS One ; 9(12): e115663, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25542016

RESUMEN

Cytoplasmic constitutive active/androstane receptor (CAR) retention protein (CCRP and also known as DNAJC7) is a co-chaperone previously characterized to retain nuclear receptor CAR in the cytoplasm of HepG2 cells. Here we have produced CCRP knockout (KO) mice and demonstrated that CCRP regulates CAR at multiple steps in activation of the cytochrome (Cyp) 2b10 gene in liver: nuclear accumulation, RNA polymerase II recruitment and epigenetic modifications. Phenobarbital treatment greatly increased nuclear CAR accumulation in the livers of KO males as compared to those of wild type (WT) males. Despite this accumulation, phenobarbital-induced activation of the Cyp2b10 gene was significantly attenuated. In ChIP assays, a CAR/retinoid X receptor-α (RXRα) heterodimer binding to the Cyp2b10 promoter was already increased before phenobarbital treatment and further pronounced after treatment. However, RNA polymerase II was barely recruited to the promoter even after phenobarbital treatment. Histone H3K27 on the Cyp2b10 promoter was de-methylated only after phenobarbital treatment in WT but was fully de-methylated before treatment in KO males. Thus, CCRP confers phenobarbital-induced de-methylation capability to the promoter as well as the phenobarbital responsiveness of recruiting RNA polymerase II, but is not responsible for the binding between CAR and its cognate sequence, phenobarbital responsive element module. In addition, KO males developed steatotic livers and increased serum levels of total cholesterol and high density lipoprotein in response to fasting. CCRP appears to be involved in various hepatic regulations far beyond CAR-mediated drug metabolism.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Hígado Graso/metabolismo , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Esteroide Hidroxilasas/metabolismo , Animales , Hidrocarburo de Aril Hidroxilasas/genética , Familia 2 del Citocromo P450 , Epigénesis Genética , Hígado Graso/genética , Proteínas de Choque Térmico , Histonas/metabolismo , Masculino , Ratones , Chaperonas Moleculares , Fenobarbital/farmacología , Unión Proteica , Transporte de Proteínas , ARN Polimerasa II/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Elementos de Respuesta/efectos de los fármacos , Receptor alfa X Retinoide/metabolismo , Esteroide Hidroxilasas/genética , Activación Transcripcional
16.
Toxicol Sci ; 137(2): 292-302, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24218150

RESUMEN

Polybrominated diphenyl ether BDE-47 (2,2',4,4'-tetrabromodiphenyl ether) is a thyroid hormone disruptor in mice; hepatic induction of various metabolic enzymes and transporters has been suggested as the mechanism for this disruption. Utilizing Car (-/-) and Pxr (-/-) mice as well as human primary hepatocytes, here we have demonstrated that BDE-47 activated both mouse and human nuclear receptor constitutive activated/androstane receptor (CAR). In mouse livers, CAR, not PXR, was responsible for Cyp2b10 mRNA induction by BDE-47. In human primary hepatocytes, BDE-47 was able to induce translocation of YFP-tagged human CAR from the cytoplasm to the nucleus andCYP2B6 and CYP3A4 mRNAs expressions. BDE-47 activated human CAR in a manner akin to the human CAR ligand CITCO (6-(4-Chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime) in luciferase-reporter assays using Huh-7 cells. In contrast, mouse CAR was not potently activated by BDE-47 in the same reporter assays. Furthermore, human pregnane X receptor (PXR) was effectively activated by BDE-47 while mouse PXR was weakly activated in luciferase-reporter assays. Our results indicate that BDE-47 induces CYP genes through activation of human CAR in addition to the previously identified pathway through human PXR.


Asunto(s)
Disruptores Endocrinos/toxicidad , Retardadores de Llama/toxicidad , Éteres Difenilos Halogenados/toxicidad , Hepatocitos/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Western Blotting , Células Cultivadas , Receptor de Androstano Constitutivo , Sistema Enzimático del Citocromo P-450/genética , Expresión Génica/efectos de los fármacos , Genes Reporteros , Hepatocitos/enzimología , Hepatocitos/metabolismo , Humanos , Ratones , Ratones Noqueados , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Receptor X de Pregnano , Cultivo Primario de Células , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Transfección
17.
Sci Signal ; 6(274): ra31, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23652203

RESUMEN

Phenobarbital is a central nervous system depressant that also indirectly activates nuclear receptor constitutive active androstane receptor (CAR), which promotes drug and energy metabolism, as well as cell growth (and death), in the liver. We found that phenobarbital activated CAR by inhibiting epidermal growth factor receptor (EGFR) signaling. Phenobarbital bound to EGFR and potently inhibited the binding of EGF, which prevented the activation of EGFR. This abrogation of EGFR signaling induced the dephosphorylation of receptor for activated C kinase 1 (RACK1) at Tyr(52), which then promoted the dephosphorylation of CAR at Thr(38) by the catalytic core subunit of protein phosphatase 2A. The findings demonstrated that the phenobarbital-induced mechanism of CAR dephosphorylation and activation is mediated through its direct interaction with and inhibition of EGFR.


Asunto(s)
Receptores ErbB/metabolismo , Fenobarbital/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Receptor de Androstano Constitutivo , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/química , Receptores ErbB/genética , Moduladores del GABA/metabolismo , Moduladores del GABA/farmacología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Modelos Moleculares , Fenobarbital/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Estructura Terciaria de Proteína , Interferencia de ARN , Receptores de Cinasa C Activada , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Citoplasmáticos y Nucleares/genética
18.
Drug Metab Dispos ; 41(6): 1170-3, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23539296

RESUMEN

The constitutive active/androstane receptor (CAR) regulates hepatic drug metabolism by activating genes, such as cytochrome P450, and certain transferases. p38 Mitogen-activated protein kinase (MAPK) is highly activated in human primary hepatocytes but barely in human hepatoma cell lines including HepG2 cells. Liganded-CAR induced CYP2B6 mRNA in human primary hepatocytes far more effectively than in HepG2 cells ectopically expressing CAR. In the present study, we found that activation of p38 MAPK by anisomycin potentiated induction of CYP2B6 mRNA by CAR ligand in HepG2 cells to levels observed in ligand-treated human primary hepatocytes. siRNA knockdown of p38 MAPK abrogated the ability of anisomycin to synergistically induce CYP2B6 mRNA. In addition to CYP2B6, anisomycin cotreatment potentiated an increase in CYP2A7 and CYP2C9 mRNAs but not CYP3A4 or UDP-glucuronosyltransferase 1A1 mRNAs. Thus, activated p38 MAPK is required for liganded-CAR to selectively activate a set of genes that encode drug-metabolizing enzymes. Our present results suggest that CAR-mediated induction of these enzymes cannot be understood by ligand binding alone because the specificity and magnitude of induction are codetermined by a given cell signaling, such as p38 MAPK; both physiologic and pathophysiological states of cell signaling may have a strong impact in hepatic drug-metabolizing capability during treatments.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Hidrocarburo de Aril Hidroxilasas/genética , Células Cultivadas , Receptor de Androstano Constitutivo , Citocromo P-450 CYP2B6 , Activación Enzimática/fisiología , Femenino , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/genética
19.
Toxicol Sci ; 132(1): 151-61, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23291559

RESUMEN

KCNK1, a member of the family of two-pore K(+) ion channels, is specifically induced in the livers of male mice after phenobarbital treatment. Here, we have determined the molecular mechanism of this male-specific activation of the Kcnk1 gene and characterized KCNK1 as a phenobarbital-inducible antihyperplasia factor. Upon activation by phenobarbital, nuclear receptor CAR binds the 97-bp response element (-2441/-2345) within the Kcnk1 promoter. This binding is observed in the livers of male mice, but not in the livers of female mice and requires the pituitary gland, because hypophysectomy abrogates it. Hyperplasia further progressed in the livers of Kcnk1 ( -/- ) male mice compared with those of Kcnk1 ( +/+ ) males after phenobarbital treatment. Thus, KCNK1 suppresses phenobarbital-induced hyperplasia. These results indicate that phenobarbital treatment induces KCNK1 to elicit a male-specific and growth-suppressing signal. Thus, KCNK1 and Kcnk1 ( -/- ) mice provide an experimental tool for further investigation into the molecular mechanism of CAR-mediated promotion of the development of hepatocellular carcinoma in mice.


Asunto(s)
Hiperplasia/inducido químicamente , Hígado/efectos de los fármacos , Fenobarbital/toxicidad , Canales de Potasio de Dominio Poro en Tándem/genética , Receptores Citoplasmáticos y Nucleares/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Inmunoprecipitación de Cromatina , Receptor de Androstano Constitutivo , Cartilla de ADN , Femenino , Hiperplasia/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Reacción en Cadena en Tiempo Real de la Polimerasa
20.
PLoS One ; 8(12): e84462, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24386386

RESUMEN

BACKGROUND: Whereas estrogen receptors are present in immune cells, it is not known if they are phosphorylated to regulate immune cell functions. Here we determined the phosphorylation status of estrogen receptor α (ERα) at residue serine 216 in mouse neutrophils and examined its role in migration and infiltration. Serine 216 is the conserved phosphorylation site within the DNA binding domains found in the majority of nuclear receptors. METHODOLOGY/PRINCIPAL FINDINGS: A phospho-peptide antibody specific to phosphorylated serine 216 and ERα KO mice were utilized in immunohistochemistry, double immuno-staining or Western blot to detect phosphorylation of ERα in peripheral blood as well as infiltrating neutrophils in the mouse uterus. Transwell assays were performed to examine migration of neutrophils. An anti-Ly6G antibody identified neutrophils. About 20% of neutrophils expressed phosphorylated ERα at serine 216 in peripheral white blood cells (WBC) from C3H/HeNCrIBR females. Phosphorylation was additively segregated between C3H/HeNCrIBR and C57BL/6 females. Only neutrophils that expressed phosphorylated ERα migrated in Transwell assays as well as infiltrated the mouse uterus during normal estrous cycles. CONCLUSIONS/SIGNIFICANCE: ERα was phosphorylated at serine 216 in about 20% of mouse peripheral blood neutrophils. Only those that express phosphorylated ERα migrate and infiltrate the mouse uterus. This phosphorylation was the first to be characterized in endogenous ERα found in normal tissues and cells. Phosphorylated ERα may have opened a novel research direction for biological roles of phosphorylation in ERα actions and can be developed as a drug target for treatment of immune-related diseases.


Asunto(s)
Receptor alfa de Estrógeno/inmunología , Infiltración Neutrófila , Neutrófilos/inmunología , Útero/inmunología , Animales , Femenino , Ratones , Neutrófilos/citología , Fosforilación/inmunología , Serina/inmunología , Útero/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA