Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biol Pharm Bull ; 47(6): 1231-1238, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38945844

RESUMEN

Porcine placental extract (PPE) is commonly used in various health foods and cosmetics. PPE use in cosmetics predominantly consist of the water-soluble fraction derived from the entire placenta. In this report, we examined the effect of the hydrophobic constituents of the PPE, specifically the sphingolipid-enriched fraction designated as the sphingolipid-enriched porcine placental extract (SLPPE), on the expression of genes associated with skin function in cultured normal human epidermal keratinocytes. Using quantitative RT-PCR (qRT-PCR) analysis, we found that SLPPE concentrations ranging from 25 to 100 µg/mL upregulated the gene expression of key components associated with the cornified envelope structure (filaggrin (FLG), involucrin (IVL) and loricrin (LOR)), cornification enzymes (transglutaminase 1 (TGM1) and TGM5) and the desquamation enzymes (kallikrein 5 (KLK5) and KLK7). Additionally, KLK5p and FLG protein (FLGp) were detected in the culture supernatants of keratinocytes treated with SLPPE at these concentrations. These findings suggest that SLPPE is possible to promote the cornification and desquamation in epidermal keratinocytes, and it may offer potential benefits in cosmetics.


Asunto(s)
Proteínas Filagrina , Calicreínas , Queratinocitos , Esfingolípidos , Transglutaminasas , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Humanos , Animales , Transglutaminasas/metabolismo , Transglutaminasas/genética , Porcinos , Esfingolípidos/metabolismo , Calicreínas/metabolismo , Calicreínas/genética , Extractos Placentarios/farmacología , Células Cultivadas , Femenino , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Embarazo
2.
Biol Pharm Bull ; 46(9): 1338-1342, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37661412

RESUMEN

Growth hormone receptor (GHR)-deficient pigs were generated using the CRISPR/Cas9 system to investigate the involvement of GHR-mediated growth hormone (GH) signaling in androgen-associated gene expression of hepatic drug metabolizing enzymes (DMEs) and drug transporters. We initially confirmed that no wild-type GHR mRNA was present in GHR-/- (GHR-KO) pigs; in addition, as previously reported, those pigs exhibited decreases in body weight and serum insulin-like growth factor-1 concentration and an increase in serum GH concentration compared with the levels in GHR-/+ and GHR+/+ pigs with a wild-type GHR mRNA. The real-time RT-PCR results on the mRNA levels of hepatic DMEs and drug transporters in the GHR-KO pigs and the pigs with a wild-type GHR mRNA revealed that, among the examined hepatic DMEs, the mRNA levels of CYP1A2, CYP2A19, sulfotransferase (SULT) 1A1, and SULT2A1 were higher in GHR-KO pigs than in the pigs with a wild-type GHR mRNA, whereas the opposite trend was observed for the mRNA level of uridine 5'-diphospho-glucuronosyltransferase 1A6. No such significant differences in the mRNA levels of three hepatic drug transporters including multidrug resistance protein 1 were observed. In addition, the mRNA level of hepatic cut-like homeobox 2 (CUX2), which is expressed in an androgen-dependent manner and associated with the hepatic mRNA expression of several DMEs, was significantly decreased in GHR-KO pigs. The present findings strongly suggest that not only serum androgen but also GHR-mediated GH signaling contributes to the mRNA expression of several DMEs and CUX2, but not transporters, in the pig liver.


Asunto(s)
Andrógenos , Síndrome de Laron , Animales , Porcinos , Proteínas de Transporte de Membrana , Fibrinolíticos , Expresión Génica
3.
Biol Pharm Bull ; 46(3): 482-487, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36858577

RESUMEN

We previously identified androgen-dependent sex differences in the mRNA expression of drug metabolizing enzymes (DMEs), including CYPs, sulfotransferases and uridine 5'-diphospho-glucuronosyltransferases, and drug transporters in the pig liver and kidney. To elucidate the mechanism for such sex differences in pigs, we herein focused on the key regulators cut-like homeobox 2 (Cux2), B-cell lymphoma 6 (Bcl6), and signal transducer and activator of transcription 5b (Stat5b), which are reported to be responsible for the sex-biased gene expression of Cyps in the mouse liver. We used real-time RT-PCR to examine androgen-dependent sex differences in the mRNA levels of these regulators in the liver and kidney basically using Meishan and Landrace pigs. Significant sex differences (male > female) in the level of CUX2 mRNA were detected in the liver of both breeds, and levels were significantly decreased in males by castration and increased in castrated males and intact females by administering testosterone propionate. No such clear androgen-dependent sex differences in hepatic BCL6 or STAT5B mRNA expression were observed in either breed. In the kidney, androgen-dependent gene expression of these regulators was not observed. In the liver, CUX2 mRNA expression closely correlated with that of DMEs and drug transporters, which were previously shown to have androgen-dependent expression. Together, these findings demonstrate that hepatic CUX2 mRNA is expressed in an androgen-dependent manner, and strongly suggest that CUX2 plays a key role in the androgen-dependent gene expression of hepatic DMEs and drug transporters.


Asunto(s)
Andrógenos , Proteínas de Homeodominio , Hígado , Animales , Femenino , Masculino , Ratones , Riñón , Proteínas de Transporte de Membrana , ARN Mensajero , Porcinos , Proteínas de Homeodominio/metabolismo
4.
Int J Mol Sci ; 23(20)2022 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-36292946

RESUMEN

Cancer stem cells (CSCs) contribute to the drug resistance, recurrence, and metastasis of breast cancers. Recently, we demonstrated that HER2 overexpression increases mammosphere formation via the activation of aryl hydrocarbon receptor (AHR). In this study, the objective was to identify the mechanism underlying mammosphere maintenance mediated by HER2 signaling-activated AHR. We compared the chromatin structure of AHR-knockout (AHRKO) HER2-overexpressing MCF-7 (HER2-5) cells with that of wild-type HER2-5 cells; subsequently, we identified TP63, a stemness factor, as a potential target gene of AHR. ΔNp63 mRNA and protein levels were higher in HER2-5 cells than in HER2-5/AHRKO cells. Activation of HER2/HER3 signaling by heregulin treatment increased ΔNp63 mRNA levels, and its induction was decreased by AHR knockdown in HER2-5 cells. The results of the chromatin immunoprecipitation assay revealed an interaction between AHR and the intronic region of TP63, which encodes ΔNp63. A luciferase reporter gene assay with the intronic region of TP63 showed that AHR expression increased reporter activity. Collectively, our findings suggest that HER2-activated AHR upregulates ΔNp63 expression and that this signaling cascade is involved in CSC maintenance in HER2-expressing breast cancers.


Asunto(s)
Neoplasias de la Mama , Receptores de Hidrocarburo de Aril , Humanos , Femenino , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Neurregulina-1/metabolismo , Regulación hacia Arriba , Neoplasias de la Mama/metabolismo , ARN Mensajero/genética , Luciferasas/metabolismo , Cromatina , Línea Celular Tumoral
5.
Exp Cell Res ; 419(2): 113333, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36030969

RESUMEN

Recently, selective androgen receptor modulators (SARMs), which bind to AR and act in a tissue/effect-specific manner, have been developed, but the selective mechanism is not well understood. In this study, we investigated the selective mechanism using the synthetic steroid YK11, which showed AR-mediated gene-selective transactivation. In the AR-positive human breast cancer MDA-MB-453 cells, different patterns of AR-mediated target gene expression and AR recruitment to their enhancer regions were observed between DHT and YK11. A docking study suggested the helices 11 and 12 was moved by the sterically hindered C17-group of YK11. Furthermore, the mutational studies of AR Gln902 and mammalian two-hybrid assays suggested different cofactor recruitment between DHT and YK11. The results of this study suggest that gene selective regulation by SARMs results from differential DNA-binding and/or cofactor recruitment by ligands. These results provide novel insights into the mechanism of action of SARMs.


Asunto(s)
Neoplasias de la Mama , Receptores Androgénicos , Andrógenos/farmacología , Animales , Neoplasias de la Mama/genética , ADN , Femenino , Expresión Génica , Humanos , Mamíferos/genética , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Esteroides
6.
Biochem Biophys Res Commun ; 570: 131-136, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34280616

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates various toxicological and biological functions. We reported previously that 3-methylcholanthrene (3MC), an exogenous AhR agonist, inhibited tumorsphere (mammosphere) formation from breast cancer cell lines, while the endogenous AhR agonist, indirubin, very weakly inhibited this process. However, the difference in inhibition mechanism of mammosphere formation by 3MC or indirubin is still unknown. In this study, we established AhR-re-expressing (KOTR-AhR) cells from AhR knockout MCF-7 cells using the tetracycline (Tet)-inducible gene expression systems. To identify any difference in inhibition of mammosphere formation by 3MC or indirubin, RNA-sequencing (RNA-seq) experiments were performed using KOTR-AhR cells. RNA-seq experiments revealed that cell division cycle 20 (CDC20), which regulates the cell cycle and mitosis, was decreased by 3MC, but not by indirubin, in the presence of AhR expression. Furthermore, the mRNA and protein levels of CDC20 were decreased by 3MC in MCF-7 cells via the AhR. In addition, mammosphere formation was suppressed by small interfering RNA-mediated CDC20 knockdown compared to the negative control in MCF-7 cells. These results suggest that AhR activation by 3MC suppresses mammosphere formation via downregulation of CDC20 expression in breast cancer cells. This study provides useful information for the development of AhR-targeted anti-cancer drugs.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Cdc20/metabolismo , Regulación hacia Abajo , Metilcolantreno/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Esferoides Celulares/patología , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/farmacología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Transcriptoma/genética
7.
Biol Pharm Bull ; 44(4): 571-578, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33790107

RESUMEN

We had previously reported that treatment with the aryl hydrocarbon receptor (AHR) agonist ß-naphthoflavone (ßNF) suppressed mammosphere formation derived from cancer stem cells in human breast cancer MCF-7 cells (Cancer Lett., 317, 2012, Zhao et al.). Here, using several AHR agonists, we have investigated the association of this suppression with the classical ability to induce AHR-mediated gene transcription in the xenobiotic response element (XRE). The mammosphere formation assays were performed using wild-type and AHR-knockout MCF-7 cells in the presence of AHR agonists including 3-methylcholanthrene (3MC), benzo[a]pyrene (BaP), 7,12-dimethylbenz[a]anthracene (DMBA), 6-formylindolo[3,2-b]carbazole (FICZ), indirubin, indole-3-carbinol (I3C), indole-3-acetic acid (IAA), and kynurenine (KYN), followed by the XRE-reporter gene assays of the agonists. We showed that treatments with 3MC, BaP, and DMBA strongly suppressed mammosphere formation of the stem cells in an AHR-dependent manner, while other agonists showed weaker suppression. In reporter gene assays, the strength or duration of AHR/XRE-mediated gene transcription was found to be dependent on the agonist. Although strong transcriptional activation was observed with 3MC, FICZ, indirubin, I3C, IAA, or KYN after 6 h of treatment, only weak activation was seen with BaP or DMBA. While transcriptional activation was sustained or increased at 24 h with 3MC, BaP, or DMBA, appreciable reduction was observed with the other agonists. In conclusions, the results demonstrated that the suppressive effects of AHR agonists on mammosphere formation do not necessarily correlate with their abilities to induce AHR-mediated gene transcription. Hence, different AHR functions may be differentially induced in an agonist-dependent manner.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Neoplasias de la Mama/genética , Indoles/farmacología , Quinurenina/farmacología , Hidrocarburos Policíclicos Aromáticos/farmacología , Receptores de Hidrocarburo de Aril/agonistas , Transcripción Genética/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Supervivencia Celular/efectos de los fármacos , Técnicas de Inactivación de Genes , Genes Reporteros , Humanos , Células MCF-7 , Receptores de Hidrocarburo de Aril/genética
8.
J Toxicol Sci ; 46(1): 25-29, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33408298

RESUMEN

The aryl hydrocarbon receptor (AhR) regulates expression of genes encoding drug/xenobiotic metabolizing enzymes. Cytochrome P450 (CYP) 3A5 is involved in drug metabolism. However, regulation of CYP3A5 gene expression is not yet well understood. In this study, we aimed to investigate the effect of the ligands of AhR on CYP3A5 gene expression. CYP3A5 mRNA expression was induced by the polycyclic aromatic hydrocarbons (PAHs) such as 3-methylcholanthrene (3MC) and benzo[a]pyrene in HepG2 cells. To determine whether the PAHs induced CYP3A5 gene expression via AhR, we generated AhR knockout (AhR KO) HepG2 cells. CYP3A5 mRNA expression was not induced by 3MC treatment in AhR KO cells. In addition, we generated AhR rescue cells from AhR KO cells and evaluated CYP3A5 mRNA expression. We found that CYP3A5 mRNA expression was induced by 3MC treatment in AhR rescue cells. Taken together, these results demonstrated that CYP3A5 mRNA expression was induced by PAHs via AhR in HepG2 cells. Our findings suggest that ligand-activated AhR affects CYP3A5-mediated drug metabolism.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Hidrocarburos Policíclicos Aromáticos/toxicidad , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Células Hep G2 , Humanos , Ligandos , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
Regul Toxicol Pharmacol ; 114: 104670, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32371103

RESUMEN

Nobiletin and tangeretin are major components of polymethoxylated flavones in the peels of citrus fruits such as Citrus reticulata. Because nobiletin and tangeretin have attracted attention due to their beneficial health properties, citrus peel extracts, in which they are concentrated, have the potential to serve as a functional food ingredient to prevent diseases. In this study, a series of toxicological studies on the peel extract of Ponkan cultivar 'Ohta ponkan' (Citrus reticulata Blanco), was conducted. No mutagenic activity was observed in a bacterial reverse mutation test, whereas chromosomal aberrations were induced in an in vitro mammalian chromosomal aberration test. No genotoxicity was observed in an in vivo mammalian micronucleus test. In a 90-day study at daily doses of 54, 180, or 540 mg/kg body weight (bw)/day, hyaline droplet nephropathy, which specifically occurs in adult male rats, was observed in males of 540 mg/kg bw/day group. No other adverse effects were observed in the 90-day study. The no adverse effect level in the 90-day study was considered to be 540 mg/kg bw/day for female rats and less than 540 mg/kg bw/day for male rats.


Asunto(s)
Citrus/química , Flavonas/toxicidad , Nootrópicos/toxicidad , Extractos Vegetales/toxicidad , Plantas Medicinales/toxicidad , Administración Oral , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Peso Corporal/efectos de los fármacos , Aberraciones Cromosómicas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Flavonas/administración & dosificación , Flavonas/química , Alimentos Funcionales/efectos adversos , Alimentos Funcionales/toxicidad , Masculino , Pruebas de Micronúcleos , Nootrópicos/administración & dosificación , Nootrópicos/química , Enfermedad de Parkinson/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Plantas Medicinales/química , Ratas , Ratas Sprague-Dawley
10.
Biochem Biophys Res Commun ; 516(3): 693-698, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31253396

RESUMEN

Triple-negative breast cancer (TNBC) is associated with poor prognosis, because of no effective targeted therapy. In the present study, we demonstrated the crucial role of the aryl hydrocarbon receptor (AhR) in mediating the effects of the chemotherapeutic agent doxorubicin (DOX) in the chemotherapeutic sensitivity of TNBC. Firstly, we established AhR knockout (KO) MDA-MB 231 TNBC cells. The cytotoxic effects of DOX were more pronounced in AhR KO cells than in parental cells. In addition, our results indicated that AhR KO cells showed downregulated expression of DOX-metabolism enzyme, aldo-keto reductase (AKR) 1C3, relative to those of parental cells. Furthermore, AhR was found to enhance AKR1C3 promoter reporter activity, suggesting that AKR1C3 mRNA transcription is activated by AhR. Additionally, our findings confirmed that the downregulation of AKR1C3 expression enhanced DOX sensitivity in MDA-MB 231 cells. Finally, AhR and AKR1C3 expression were positively correlated in human breast cancer. Taken together, our results suggested that AhR is involved in DOX sensitivity by regulating AKR1C3 expression in TNBC cells.


Asunto(s)
Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas/genética , Doxorrubicina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Receptores de Hidrocarburo de Aril/genética , Neoplasias de la Mama Triple Negativas/genética , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Femenino , Técnicas de Inactivación de Genes , Humanos , Receptores de Hidrocarburo de Aril/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo
11.
Bioorg Chem ; 88: 102977, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31100617

RESUMEN

Selective estrogen receptor modulators (SERMs) act as either agonist or antagonist of estrogen receptor (ER) in a tissue selective manner and have been used in several diseases such as breast cancer, postmenopausal syndrome, osteoporosis, and cardiovascular diseases. However, current SERMs may also increase the risk of serious side effects and trigger drug resistance. Herein, a screening program, that was designed to search for novel SERMs, resulted in the identification of a series of 2-arylbenzofuran-containing compounds that are ligands for ERα, when applying the Gaussia-luciferase reporter assay. One of these compounds, 10-dehydrooxyglycyuralin E (T9) was chemically synthesized. T9 showed anti-estrogenic/proliferative activity in ERα-positive breast cancer cells. Pretreatment of T9 prevented the mRNA expression of GREB1, which is an estrogen response gene. Furthermore, by an in silico docking simulation study we demonstrated that T9 showed interactions directly to ERα. Taken together, these results demonstrated that T9 is a candidate of SERMs and a useful seed compound for the foundation of the selective activity of SERMs.


Asunto(s)
Benzofuranos/farmacología , Receptor alfa de Estrógeno/agonistas , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Benzofuranos/síntesis química , Benzofuranos/química , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Moduladores Selectivos de los Receptores de Estrógeno/síntesis química , Moduladores Selectivos de los Receptores de Estrógeno/química , Relación Estructura-Actividad
12.
Mol Pharmacol ; 95(1): 120-126, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30397001

RESUMEN

The constitutive active/androstane receptor (CAR) controls genes involved in xenochemical metabolism. Although numerous cofactors have been reported to be involved in CAR-mediated transactivation, unknown and poorly defined proteins recruited by CAR have yet to be characterized. In this study, a novel CAR-interacting protein, cell cycle and apoptosis regulator 1 (CCAR1), was identified by coimmunoprecipitation analysis using human hepatocarcinoma HepG2 cells expressing FLAG epitope-tagged CAR. We demonstrated that CCAR1 can act as an enhancer-dependent coactivator of CAR. First, we showed that overexpression of CCAR1 enhanced CAR-induced reporter gene activity with triplicate consensus direct repeat 4 motif (DR4-Luc), xenobiotic-responsive enhancer module (XREM)-enhancer of CYP3A4 (XREM-Luc), and phenobarbital-responsive enhancer module of UDP-glucuronosyltransferases 1A1 (UGT1A1) (gtPBREM)-enhancer of UGT1A1 (gtPBREM-Luc)-driven reporter plasmids but not PBREM-enhancer of CYP2B6 (PBREM-Luc)-driven reporter activity. Furthermore, we showed that knockdown of CCAR1 suppressed CAR-induced UGT1A1 mRNA expression but did not affect CAR-induced CYP2B6 mRNA expression in HepTR/CAR and HepaRG cells. Moreover, CCAR1 could be recruited to the gtPBREM of the UGT1A1 enhancer by CAR but not to the PBREM of the CYP2B6 enhancer. Moreover, we showed that CCAR1 can act as a secondary coactivator by cooperating with the p160 family of steroid receptor coactivators (SRCs). These findings demonstrated CCAR1 to be a novel transcriptional cofactor for CAR and provided insight regarding the mechanism of CAR-mediated gene-selective transactivation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Apoptosis/genética , Proteínas de Ciclo Celular/genética , Ciclo Celular/genética , Elementos de Facilitación Genéticos/genética , Receptores Citoplasmáticos y Nucleares/genética , Activación Transcripcional/genética , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Receptor de Androstano Constitutivo , Citocromo P-450 CYP2B6/genética , Citocromo P-450 CYP3A/genética , Elementos de Facilitación Genéticos/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Genes Reporteros/genética , Glucuronosiltransferasa/genética , Células Hep G2 , Humanos , Reactores Nucleares , Fenobarbital/farmacología , ARN Mensajero/genética , Receptores de Esteroides/genética , Activación Transcripcional/efectos de los fármacos , Xenobióticos/farmacología
13.
Biol Pharm Bull ; 41(3): 394-398, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29491216

RESUMEN

Androgens are key regulators that play a critical role in the male reproductive system and have anabolic effects on bone mineral density and skeletal muscle mass. We have previously reported that YK11 is a novel selective androgen receptor modulator (SARM) and induces myogenic differentiation and selective gene regulation. In this study, we show that treatment of YK11 and dihydrotestosterone (DHT) accelerated cell proliferation and mineralization in MC3T3-E1 mouse osteoblast cells. Further, YK11-treated cells increased osteoblast specific differentiation markers, such as osteoprotegerin and osteocalcin, compared to untreated cells. These observations were attenuated by androgen receptor (AR) antagonist treatment. To clarify the effect of YK11, we investigated rapid non-genomic signaling by AR. The phosphorylated Akt protein level was increased by YK11 and DHT treatment, suggesting that YK11 activates Akt-signaling via non-genomic signaling of AR. Because it is known Akt-signaling is a key regulator of androgen-mediated osteoblast differentiation, YK11 has osteogenic activity as well as androgen.


Asunto(s)
Andrógenos/farmacología , Norpregnadienos/farmacología , Osteoblastos/efectos de los fármacos , Células 3T3 , Animales , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ratones , Proteína Oncogénica v-akt/biosíntesis , Proteína Oncogénica v-akt/genética , Osteogénesis/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
14.
Exp Cell Res ; 366(1): 34-40, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29501698

RESUMEN

HER2 overexpression accounts for approximately 15-20% of all breast cancers. We have shown that HER2 overexpression leads to elevated expression of the aryl hydrocarbon receptor (AhR) in breast cancer cells. In this study, firstly, we showed that AhR expression was up-regulated by treatment with the HER3 ligand heregulin (HRG) in HER2-overexpressing breast cancer cell lines. Induction of AhR was mediated by transcriptional activation of the region of AhR promoter corresponding to - 190 to - 100 bp. In addition, HRG treatment elicited nuclear translocation of AhR. To investigate the role of AhR in HRG-HER2/HER3 signaling in HER2-overexpressing cells, we established AhR knockout (KO) HER2-overexpressing cells to perform wound-healing assays. HRG-induced cell migration was markedly attenuated by AhR KO. HRG-induced cell migration was associated with increased expression of the inflammatory cytokines interleukin (IL)-6 and IL-8 in wild type cells, but not in AhR KO cells. These results elucidate that AhR is an important factor for the malignancy in HER2 overexpressing breast cancers.


Asunto(s)
Neoplasias de la Mama/genética , Movimiento Celular/genética , Neurregulina-1/genética , Receptor ErbB-2/genética , Receptores de Hidrocarburo de Aril/genética , Línea Celular Tumoral , Núcleo Celular/genética , Femenino , Humanos , Interleucina-6/genética , Interleucina-8/genética , Regiones Promotoras Genéticas/genética , Receptor ErbB-3/genética , Transducción de Señal/genética , Regulación hacia Arriba/genética , Cicatrización de Heridas/genética
15.
Drug Metab Dispos ; 46(1): 46-52, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29101097

RESUMEN

The constitutive androstane receptor (CAR) is a nuclear receptor that acts as a transcription factor for a variety of genes, including genes encoding xenobiotic, steroid, and drug-metabolizing enzymes and transporters. Transactivation of a target gene by a transcription factor is generally mediated through the concerted and stepwise recruitment of various proteins termed coregulators, including coactivators and corepressors. In this study, TRIM24 (also known as transcriptional intermediary factor 1 alpha) was found to interact with the CAR. TRIM24 enhanced the CAR-dependent transactivation in reporter assays using the direct repeat-4 motif, a binding site of the CAR. This enhancement was synergistically augmented in the presence of steroid receptor coactivator (SRC) 1 or SRC2, both of which are coactivators of the CAR. In addition, TRIM24 was recruited to the CAR-binding element of the CYP2B6 promoter together with the CAR. We also noted that knockdown of TRIM24 suppressed CAR-induced CYP2B6 mRNA expression in HepTR/CAR and HepaRG cells and suppressed CAR-induced CYP3A4 mRNA expression in HepaRG cells but not HepTR/CAR cells. From these results, we suggest that TRIM24 is a novel coactivator of the CAR that is involved in cell- and/or promoter- selective transactivation.


Asunto(s)
Proteínas Portadoras/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Activación Transcripcional , Proteínas Portadoras/genética , Receptor de Androstano Constitutivo , Citocromo P-450 CYP2B6/genética , Citocromo P-450 CYP2B6/metabolismo , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Coactivador 1 de Receptor Nuclear/genética , Coactivador 1 de Receptor Nuclear/metabolismo , Coactivador 2 del Receptor Nuclear/genética , Coactivador 2 del Receptor Nuclear/metabolismo , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores Citoplasmáticos y Nucleares/genética
16.
Bioorg Med Chem Lett ; 27(11): 2608-2612, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28427809

RESUMEN

Targeted therapy is unavailable for treating patients with triple-negative breast cancer (TNBC), which accounts for approximately 15% of all breast cancers. Overexpression of epidermal growth factor receptor (EGFR) is observed in approximately 30-60% of TNBCs. Therefore, developing novel strategies for inhibiting EGFR signaling is required. In the present study, a natural compound library was screened to identify molecules that target TNBCs that overexpress EGFR. Picrasidine G (PG), a naturally occurring dimeric alkaloid produced by Picrasma quassioides, decreased the viability of the MDA-MB 468 cell line (TNBCEGFR+) compared with other breast cancer cell lines. PG treatment increased markers of apoptosis, including chromatin condensation, sub-G1 population, cleavage of caspase 3 and cleavage of poly (ADP-ribose) polymerase (PARP). PG inhibited the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and inhibited transcription of the STAT3-target gene encoding survivin. Further, PG inhibited EGF-induced STAT3 phosphorylation but not interleukin-6 (IL-6)-induced STAT3 phosphorylation. These results suggest that PG may contribute to the development of targeted therapy of patients with EGFR-overexpressing TNBC.


Asunto(s)
Alcaloides/química , Carbolinas/toxicidad , Receptores ErbB/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Alcaloides/toxicidad , Apoptosis/efectos de los fármacos , Carbolinas/química , Caspasa 3/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Interleucina-6/farmacología , Fosforilación/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
17.
Phytomedicine ; 26: 22-27, 2017 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-28257661

RESUMEN

BACKGROUND: Pregnane X receptor (PXR) is a key regulator of the induction of drug metabolizing enzymes. PXR has been studied for its importance in drug-drug or herb-drug interactions, and it is also a molecular target for the treatment of inflammatory and metabolic diseases. PURPOSE: This study aims to determine new natural PXR-ligands from traditional plant medicines. METHODS: The PXR activation activity was measured by a mammalian one hybrid assay of PXR. Identification of the active compound from Alisma rhizome (the rhizomes of Alisma orientale) was carried out by bioassay-guided fractionation method. The transcriptional activity of the liver-enriched nuclear receptors was measured by the luciferase reporter assay. The interaction between the SRC-1 and PXR was measured by a mammalian 2-hybrid assay. The expression of endogenous CYP3A4 mRNA in both cultured hPXR-overexpressing hepatoma cells and human primary hepatocytes were measured by quantitative RT-PCR method. RESULTS: The extract of Alisma rhizome showed the most potent activation activity by screening of a library of medicinal plant extracts. Alisol B 23-acetate (ABA) was identified to be the active compound of Alisma rhizome. ABA caused a concentration-dependent increase on the PXR-dependent transactivation of a luciferase reporter gene, but did not affect the ligand binding activity of the liver-enriched nuclear receptors, such as CAR, LXR, FXR, PPARα, PPARδ and PPARγ, emphasizing that ABA is a potent and specific agonist of PXR. With ABA treatment, the direct interaction between the ligand-binding domain of PXR and the receptor interaction domain of SRC1 was observed. ABA also induced the expression of endogenous CYP3A4 mRNA in both cultured hPXR-overexpressing hepatoma cells and human primary hepatocytes. CONCLUSION: Since the rhizomes of Alisma orientale are used for a wide range of ailments in traditional Chinese medicine and Japanese Kampo medicine, this study could possibly extend into the clinical usage of these medicines via the mechanism of PXR activation.


Asunto(s)
Alisma/química , Colestenonas/farmacología , Extractos Vegetales/farmacología , Receptores de Esteroides/agonistas , Rizoma/química , Animales , China , Humanos , Medicina Tradicional China , Receptor X de Pregnano
18.
J Nat Prod ; 79(12): 3127-3133, 2016 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-27958735

RESUMEN

Picrasidine C (1), a dimeric ß-carboline-type alkaloid isolated from the root of Picrasma quassioides, was identified to have PPARα agonistic activity by a mammalian one-hybrid assay from a compound library. Among the PPAR subtypes, 1 selectively activated PPARα in a concentration-dependent manner. Remarkably, 1 also promoted PPARα transcriptional activity by a peroxisome proliferator response element-driven luciferase reporter assay. Furthermore, 1 induced the expression of PPARα-regulated genes involved in lipid, glucose, and cholesterol metabolism, such as CPT-1, PPARα, PDK4, and ABCA1, which was abrogated by the PPARα antagonist MK-886, indicating that the effect of 1 was dependent on PPARα activation. This is the first report to demonstrate 1 to be a subtype-selective PPARα agonist with potential application in treating metabolic diseases, such as hyperlipidemia, atherosclerosis, and hypercholesterolemia.


Asunto(s)
Alcaloides/aislamiento & purificación , Alcaloides/farmacología , Carbolinas/aislamiento & purificación , Carbolinas/farmacología , PPAR alfa/agonistas , Picrasma/química , Alcaloides/química , Animales , Aterosclerosis/tratamiento farmacológico , Carbolinas/química , Colesterol/metabolismo , Glucosa/metabolismo , Hipercolesterolemia/tratamiento farmacológico , Indoles/farmacología , Metabolismo de los Lípidos , Lípidos , Ratones , Estructura Molecular , PPAR alfa/genética , Factores de Transcripción/metabolismo
19.
Molecules ; 21(9)2016 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-27607997

RESUMEN

An efficient Pd(II)/Pd°-p-benzoquinone/hydroquinone-CuCl2/CuCl catalyst system was developed that uses environmentally friendly molecular oxygen as the terminal oxidant to catalyze the cyclization-carbonylation-cyclization coupling reaction (CCC-coupling reaction) of (o-alkynyl phenyl) (methoxymethyl) sulfides.


Asunto(s)
Oxígeno/química , Paladio/química , Sulfuros/química , Sulfuros/síntesis química , Catálisis , Oxidación-Reducción
20.
Drug Metab Dispos ; 44(8): 1158-63, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27197997

RESUMEN

The constitutive androstane receptor (CAR) and pregnane X receptor (PXR) regulate various genes involved in xenobiotics and drug metabolism. In many cases, CAR/PXR share ligands termed dual ligands of CAR/PXR. It is difficult to investigate the effect of CAR/PXR dual ligands in cell lines because CAR and PXR expression is scarcely detected in cultured cell lines. Here, we established a tetracycline-inducible human CAR and stably human PXR-overexpressing HepG2 cell line (HepTR/hCAR/hPXR) to examine CAR/PXR dual ligands. In the present study, we investigated the regulation of CYP2B6, CYP2C9, CYP3A4, and UDP-glucuronosyl transferase, which are target genes of CAR/PXR, by dual ligands of CAR/PXR in two transfectants. Activation of CAR and PXR in cells treated with a high dose of CITCO [6-(4-chlorophenyl)-imidazo(2,1-b)thiazole-5-carbaldehyde] or cotreated with rifampicin and tetracycline resulted in synergistic enhancement of CYP3A4, but not CYP2B6, CYP2C9, or UGT1A1, mRNA expression in HepTR/hCAR/hPXR cells. In contrast, this synergistic effect was not observed in HepTR/hCAR cells. These observations were also demonstrated in human primary hepatocytes. Taken together, our results suggest that dual ligands of CAR/PXR show distinct gene regulation patterns by cross-talk between CAR and PXR. Furthermore, the two newly established cell lines are useful tools to investigate dual ligands of CAR/PXR.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Isoquinolinas/farmacología , Oximas/farmacología , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores de Esteroides/agonistas , Rifampin/farmacología , Tiazoles/farmacología , Receptor de Androstano Constitutivo , Citocromo P-450 CYP2B6/genética , Citocromo P-450 CYP2B6/metabolismo , Citocromo P-450 CYP2C9/genética , Citocromo P-450 CYP2C9/metabolismo , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Agonismo Parcial de Drogas , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Células Hep G2 , Humanos , Ligandos , Receptor X de Pregnano , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Cross-Talk , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...