Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Thyroid ; 33(8): 965-973, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37051697

RESUMEN

Background: Regulator of calcineurin 1.4 (RCAN1.4) is a functionally downregulated metastasis progression suppressor (MPS) in thyroid cancer; however, the mechanisms for RCAN1.4 loss in thyroid cancer have not yet been reported. The RCAN1.4 promoter and gene contain several cytosine-guanine (CG)-rich regions, some of which are reported to be hypermethylated in nonthyroid tissues. We, therefore, hypothesized that RCAN1.4 downregulation in thyroid cancer was in part due to hypermethylation. Methods: Studies were performed in 5 thyroid cancer cell lines (TPC1, FTC133, BCPAP, C643, and 8505C) with different genetic drivers, and in 18 paired normal and thyroid cancer human thyroid cancer tissues. Basal RCAN1.4 messenger RNA (mRNA) and protein levels were assessed in all of the cell lines. Cell lines with lowest RCAN1.4 expression levels were treated with the DNA methyl transferase inhibitor, decitabine. Normal/tumor tissue pairs were analyzed for methylation of three CG-rich regions both by capture of methylated DNA by MBD2 protein and by methylation-specific polymerase chain reaction (MSPCR). Results: In all assessed cell lines, RCAN1.4 mRNA and protein levels increased after decitabine treatment. In silico analysis of the RCAN1.4 gene identified 3 CG-rich regions as possible methylation targets: 1 in the proximal promoter and 2 in intron 1. Hypermethylation of the intron 1 CG-rich regions was identified by both the capture method and MSPCR. In contrast, hypermethylation of the CG-rich region of the proximal promoter was not identified. Gene expression confirmed that hypermethylation in thyroid cancer samples in intron 1 of RCAN1.4 was associated with lower levels of RCAN1.4 mRNA. Finally, the cancer samples demonstrated increased NFE2L3 expression, a downstream marker of functional RCAN1.4 loss. Conclusions: The MPS gene, RCAN1.4, is downregulated in thyroid cancer cells and human thyroid cancer in part by hypermethylation of CG-rich regions in intron 1.


Asunto(s)
Neoplasias de la Tiroides , Factores de Transcripción , Humanos , Decitabina/farmacología , Intrones , Factores de Transcripción/genética , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , ARN Mensajero/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo
2.
Am J Cancer Res ; 12(6): 2798-2816, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35812067

RESUMEN

Endocrine therapy for breast cancer often leads to drug resistance and tumor recurrence; tumor hypoxia is also associated with mortality and tumor relapse. Cytochrome P450 1B1 (CYP1B1) regulates estrogen metabolism in breast cells and is known to be overexpressed in breast cancer tissue. Although the individual association of hypoxia-induced hypoxia-inducible factor-1-alpha (HIF-1α) and CYP1B1 with tumorigenesis is well known, the association between HIF-1α and CYP1B1 leading to tumorigenesis has not been investigated. Here, we investigated the correlation between hypoxia and CYP1B1 expression in breast cancer cells for tumorigenesis-related mechanisms. Hypoxia was induced in the human breast cancer cell lines MCF-7 (Er-positive) and MDA-MB-231 (triple-negative) and the normal breast epithelial cell line MCF10A; these cell lines were then subjected to immunoblotting, transient transfection, luciferase assays, gene silencing using small interfering RNA, polymerase chain reaction analysis, chromatin immunoprecipitation, co-immunoprecipitation, and mammalian two-hybrid assays. Furthermore, immunofluorescence analysis of the tumor microarrays was performed, and the pub2015 and the Cancer Genome Atlas patient datasets were analyzed. HIF-1α expression in response to hypoxia occurred in both normal and breast cancer cells, whereas CYP1B1 was induced only in estrogen receptor α (ERα)-positive breast cancer cells under hypoxia. HIF-1α activated ERα through direct binding and in a ligand-independent manner to promote CYP1B1 expression. Therefore, we suggested the mechanism by which hypoxia and ER-positivity orchestrate breast cancer relapse.

3.
Sci Rep ; 10(1): 18316, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33110146

RESUMEN

The Akt family is comprised of three unique homologous proteins with isoform-specific effects, but isoform-specific in vivo data are limited in follicular thyroid cancer (FTC), a PI3 kinase-driven tumor. Prior studies demonstrated that PI3K/Akt signaling is important in thyroid hormone receptor ßPV/PV knock-in (PV) mice that develop metastatic thyroid cancer that most closely resembles FTC. To determine the roles of Akt isoforms in this model we crossed Akt1-/-, Akt2-/-, and Akt3-/- mice with PV mice. Over 12 months, thyroid size was reduced for the Akt null crosses (p < 0.001). Thyroid cancer development and local invasion were delayed in only the PVPV-Akt1 knock out (KO) mice in association with increased apoptosis with no change in proliferation. Primary-cultured PVPV-Akt1KO thyrocytes uniquely displayed a reduced cell motility. In contrast, loss of any Akt isoform reduced lung metastasis while vascular invasion was reduced with Akt1 or 3 loss. Microarray of thyroid RNA displayed incomplete overlap between the Akt KO models. The most upregulated gene was the dendritic cell (DC) marker CD209a only in PVPV-Akt1KO thyroids. Immunohistochemistry demonstrated an increase in CD209a-expressing cells in the PVPV-Akt1KO thyroids. In summary, Akt isoforms exhibit common and differential functions that regulate local and metastatic progression in this model of thyroid cancer.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias de la Tiroides/etiología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Isoformas de Proteínas , Receptores de Hormona Tiroidea/metabolismo , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología
4.
Endocrine ; 70(1): 24-35, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32779092

RESUMEN

Distant metastasis classically has been defined as a late-stage event in cancer progression. However, it has become clear that metastases also may occur early in the "lifetime" of a cancer and that they may remain stable at distant sites. This stability of metastatic cancer deposits has been termed "metastatic dormancy" or, as we term it, "metastatic progression dormancy" as the progression either may reflect growth of already existing metastases or new cancer spread. Biologically, dormancy is the presence of nongrowing, static metastatic cells that survive over time. Clinically, dormancy is defined by stability in tumor markers, imaging, and clinical course. Metastatic well-differentiated thyroid cancer offers an excellent tumor type to understand these processes for several reasons: (1) primary therapy often includes removal of the entire gland with ablation of residual normal tissue thereby removing one source for new metastases; (2) the presence of a sensitive biochemical and radiographic monitoring tests enabling monitoring of metastasis throughout the progression process; and (3) its tendency toward prolonged clinical dormancy that can last for years or decades be followed by progression. This latter factor provides opportunities to define therapeutic targets and/or markers of progression. In this review, we will discuss concepts of metastatic progression dormancy and the factors that drive both long-term stability and loss of dormancy with a focus on thyroid cancer.


Asunto(s)
Neoplasias de la Tiroides , Humanos , Metástasis de la Neoplasia , Neoplasias de la Tiroides/diagnóstico , Neoplasias de la Tiroides/terapia
5.
FASEB J ; 33(7): 8335-8348, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30991008

RESUMEN

Damage-induced long noncoding RNA (DINO) is a long noncoding RNA that directly interacts with p53 and thereby enhances p53 stability and activity in response to various cellular stresses. Here, we demonstrate that nuclear receptor subfamily 2 group E member 3 (NR2E3) plays a crucial role in maintaining active DINO epigenetic status for its proper induction and subsequent p53 activation. In acetaminophen (APAP)- or carbon tetrachloride-induced acute liver injuries, NR2E3 knockout (KO) mice exhibited far more severe liver injuries due to impaired DINO induction and p53 activation. Mechanistically, NR2E3 loss both in vivo and in vitro induced epigenetic DINO repression accompanied by reduced DINO chromatin accessibility. Furthermore, compared with the efficient reversal by a typical antidote N-acetylcysteine (NAC) treatment of APAP-induced liver injury in wild-type mice, the liver injury of NR2E3 KO mice was not effectively reversed, indicating that an intact NR2E3-DINO-p53-signaling axis is essential for NAC-mediated recovery against APAP-induced hepatotoxicity. These findings establish that NR2E3 is a critical component in p53 activation and a novel susceptibility factor to drug- or toxicant-induced acute liver injuries.-Khanal, T., Leung, Y.-K., Jiang, W., Timchenko, N., Ho, S.-M., Kim, K. NR2E3 is a key component in p53 activation by regulating a long noncoding RNA DINO in acute liver injuries.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Fallo Hepático Agudo/metabolismo , Receptores Nucleares Huérfanos/metabolismo , ARN Largo no Codificante/biosíntesis , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Acetaminofén/efectos adversos , Acetaminofén/farmacología , Acetilcisteína/farmacología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Epigénesis Genética/efectos de los fármacos , Células Hep G2 , Humanos , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/genética , Fallo Hepático Agudo/patología , Ratones , Ratones Noqueados , Receptores Nucleares Huérfanos/genética , ARN Largo no Codificante/genética , Proteína p53 Supresora de Tumor/genética
6.
PLoS One ; 13(10): e0205329, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30356307

RESUMEN

BACKGROUND: Myocardial infarction is the most common cause of heart failure. MI has been intricately linked to ventricular remodeling, subsequently leading to the reduction in the cardiac ejection fraction causing HF. The cumulative line of evidence suggests an important role of several biomarkers in modulating the cardiac vasculature, further contributing towards the progression of post-MI complications. Studies have demonstrated, yet not fully established, that an important biomarker, IL-10, has a causal relationship with MI and associated cardiac dysfunction. HYPOTHESIS: This study aims to establish the role of IL-10 as a prognostic marker for the cardiovascular outcomes and to develop a panel of biomarkers and circulating miRNAs that could potentially result in the early detection of HF resulting from MI, allowing for early intervention strategies. METHODS AND RESULTS: Blood was withdrawn and echocardiography assessment was performed on a total of 43 patients that were enrolled, within 24 hours of the incidence of MI. Patients were divided in three main groups, based on the ejection fraction measurement from echocardiography: control (n = 14), MI with normal EF (MI+NEF, n = 13) and MI with low EF (MI+LEF, n = 16). Our results showed that TGFß-1, TNF-α, IL-6 and MMP-9 were upregulated significantly in MI+NEF group and more so in MI+LEF group, as compared to control group (p<0.01). The circulating levels of miR-34a, miR-208b and miR-126 were positively correlated and showed elevated levels in the MI+NEF group, even higher in MI+LEF group, while levels of miR-24 and miR-29a were reduced in MI+NEF, and much lower in MI+LEF, as compared to the control group (p<0.01). Our results also demonstrated a direct correlation of IL-10 with the ejection fraction in patients with MI: IL-10 was elevated in MI+NEF group, however, the levels were significantly low in MI+LEF group suggesting an important role of IL-10 in predicting heart failure. Importantly, our study confirmed the correlation of IL-10 with EF by our follow-up echocardiography assessment that was performed 2 months after the incidence of MI. CONCLUSION: Our results support the clinical application of these serum biomarkers to develop a panel for appropriate prognosis and management of adverse cardiac remodeling and development of heart failure post-myocardial infarction.


Asunto(s)
Biomarcadores/sangre , Insuficiencia Cardíaca/sangre , Interleucina-10/sangre , Infarto del Miocardio/sangre , Anciano , Ecocardiografía , Femenino , Regulación de la Expresión Génica/genética , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/epidemiología , Insuficiencia Cardíaca/patología , Humanos , Interleucina-10/genética , Masculino , Metaloproteinasa 9 de la Matriz/sangre , MicroARNs/sangre , Persona de Mediana Edad , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Pronóstico , Volumen Sistólico/genética , Factor de Crecimiento Transformador beta/sangre , Factor de Necrosis Tumoral alfa/sangre , Función Ventricular Izquierda/genética , West Virginia/epidemiología
7.
Int J Mol Sci ; 19(9)2018 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-30201874

RESUMEN

BACKGROUND: Oxidant stress plays a key role in the development of chronic kidney disease (CKD). Experimental CKD leads to accumulation of uremic toxins (UT) in the circulation resulting in increased ROS production, which in turn, is known to activate the Na/K-ATPase/ROS amplification loop. Studies in a murine model of obesity have shown that increased oxidative stress in plasma is due to increased ROS and cytokine production from dysfunctional adipocytes. Therefore, we hypothesized that adipocytes exposed to UTs will activate the Na/K-ATPase oxidant amplification loop causing redox imbalance and phenotypic alterations in adipocytes. We also aimed to demonstrate that the Na/K-ATPase signaling antagonist, pNaKtide, attenuates these pathophysiological consequences. METHODS: In the first set of experiments, 3T3-L1 murine pre-adipocytes were treated with varying concentrations of UTs, indoxyl sulfate (IS) (50, 100 and 250 µM) and p-cresol (50, 100 and 200 µM), with or without pNaKtide (0.7 µM) for five days in adipogenic media, followed by Oil Red O staining to study adipogenesis. RT-PCR analysis was performed to study expression of adipogenic, apoptotic and inflammatory markers, while DHE staining evaluated the superoxide levels in UT treated cells. In a second set of experiments, visceral fat was obtained from the West Virginian population. MSCs were isolated and cultured in adipogenic media for 14 days, which was treated with indoxyl sulfate (0, 25, 50 and 100 µM) with or without pNaKtide (1 µM). MSC-derived adipocytes were evaluated for morphological and molecular analysis of the above markers. RESULTS: Our results demonstrated that 3T3-L1 cells and MSCs-derived adipocytes, treated with UTs, exhibited a significant decrease in adipogenesis and apoptosis through activation of the Na/K-ATPase/ROS amplification loop. The treatment with pNaKtide in 3T3-L1 cells and MSC-derived adipocytes negated the effects of UTs and restored cellular redox in adipocytes. We noted a varying effect of pNaKtide, in adipocytes treated with UTs, on inflammatory markers, adipogenic marker and superoxide levels in 3T3-L1 cells and MSC-derived adipocytes. CONCLUSIONS: This study demonstrates for the first time that the Na/K-ATPase/ROS amplification loop activated by elevated levels of UTs has varying effect on phenotypic alterations in adipocytes in various in vitro models. Thus, we propose that, if proven in humans, inhibition of Na/K-ATPase amplification of oxidant stress in CKD patients may ultimately be a novel way to combat adipocyte dysfunction and metabolic imbalance in these patients.


Asunto(s)
Adipocitos/efectos de los fármacos , Cresoles/toxicidad , Indicán/toxicidad , Células Madre Mesenquimatosas/efectos de los fármacos , Oxidantes/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Biomarcadores/análisis , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Células Cultivadas , Cresoles/orina , Humanos , Indicán/orina , Células Madre Mesenquimatosas/citología , Ratones , Estrés Oxidativo , Fragmentos de Péptidos/farmacología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/orina , ATPasa Intercambiadora de Sodio-Potasio/farmacología
8.
Sci Rep ; 7(1): 10662, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28878246

RESUMEN

The aryl hydrocarbon receptor (AHR) plays crucial roles in inflammation, metabolic disorder, and cancer. However, the molecular mechanisms regulating AHR expression remain unknown. Here, we found that an orphan nuclear NR2E3 maintains AHR expression, and forms an active transcriptional complex with transcription factor Sp1 and coactivator GRIP1 in MCF-7 human breast and HepG2 liver cancer cell lines. NR2E3 loss promotes the recruitment of LSD1, a histone demethylase of histone 3 lysine 4 di-methylation (H3K4me2), to the AHR gene promoter region, resulting in repression of AHR expression. AHR expression and responsiveness along with H3K4me2 were significantly reduced in the livers of Nr2e3rd7 (Rd7) mice that express low NR2E3 relative to the livers of wild-type mice. SP2509, an LSD1 inhibitor, fully restored AHR expression and H3K4me2 levels in Rd7 mice. Lastly, we demonstrated that both AHR and NR2E3 are significantly associated with good clinical outcomes in liver cancer. Together, our results reveal a novel link between NR2E3, AHR, and liver cancer via LSD1-mediated H3K4me2 histone modification in liver cancer development.


Asunto(s)
Reprogramación Celular/genética , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidad , Receptores Nucleares Huérfanos/deficiencia , Receptores de Hidrocarburo de Aril/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Biología Computacional/métodos , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Estimación de Kaplan-Meier , Modelos Biológicos , Pronóstico , Receptores de Hidrocarburo de Aril/metabolismo
9.
Food Chem Toxicol ; 106(Pt A): 232-241, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28571770

RESUMEN

Leptin plays a key role in the control of adipocyte formation, as well as in the associated regulation of energy intake and expenditure. The goal of this study was to determine if leptin-induced aromatase enhances estrogen production and induces tumor cell growth stimulation. To this end, breast cancer cells were incubated with leptin in the absence or presence of inhibitor pretreatment, and changes in aromatase and cyclooxygenase-2 (COX-2) expression were evaluated at the mRNA and protein levels. Transient transfection assays were performed to examine the aromatase and COX-2 gene promoter activities and immunoblot analysis was used to examine protein expression. Leptin induced aromatase expression, estradiol production, and promoter activity in breast cancer cells. Protein levels of phospho-STAT3, PKA, Akt, ERK, and JNK were increased by leptin. Leptin also significantly increased cAMP levels, cAMP response element (CRE) activation, and CREB phosphorylation. In addition, leptin induced COX-2 expression, promoter activity, and increased the production of prostaglandin E2. Finally, a COX-2 inhibitor and aromatase inhibitor suppressed leptin-induced cell proliferation in MCF-7 breast cancer cells. Together, our data show that leptin increased aromatase expression in breast cancer cells, which was correlated with COX-2 upregulation, mediated through CRE activation and cooperation among multiple signaling pathways.


Asunto(s)
Aromatasa/metabolismo , Neoplasias de la Mama/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ciclooxigenasa 2/metabolismo , Leptina/metabolismo , Aromatasa/genética , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/fisiopatología , Proliferación Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Ciclooxigenasa 2/genética , Dinoprostona/metabolismo , Estrógenos/metabolismo , Femenino , Humanos , Células MCF-7 , Fosforilación , Regiones Promotoras Genéticas
10.
Toxicol Lett ; 237(3): 228-36, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26149760

RESUMEN

We previously reported that NR2E3, an orphan nuclear receptor, plays an important role in maintaining the basal expression of estrogen receptor α (ER) and that the NR2E3 level is highly correlated with the relapse-free survival of breast cancer patients. Here, we investigated the role of NR2E3 in benzo(a)pyrene (BaP)-mediated cell injury. BaP treatment reduced NR2E3 homo-dimer formation and expression and subsequently decreased ER expression. The chromatin immunoprecipitation assay results showed that the treatment of MCF-7 breast cancer cells and the mouse liver with BaP released NR2E3 from the ER promoter to transform the transcriptionally active histone modification status into a repressive state. NR2E3 depletion in MCF-7 cells also induced a similar inactive epigenetic status in the ER promoter region, indicating that NR2E3 is an essential epigenetic player that maintains basal ER expression. Interestingly, these negative effects of BaP on the expression levels of NR2E3 and ER were rescued by antioxidant treatment. Collectively, our study provides novel evidence to show that BaP-induced oxidative stress decreases ER expression, in part by regulating NR2E3 function, which modulates the epigenetic status of the ER promoter. NR2E3 is likely an essential epigenetic player that maintains basal ER expression to protect cells from BaP-induced oxidative injury.


Asunto(s)
Benzo(a)pireno/toxicidad , Receptores Nucleares Huérfanos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Regiones Promotoras Genéticas , Receptores de Estrógenos/genética , Animales , Antioxidantes/farmacología , Regulación hacia Abajo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Histonas/genética , Histonas/metabolismo , Humanos , Células MCF-7 , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Nucleares Huérfanos/genética
11.
Arch Toxicol ; 89(11): 2039-50, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25199686

RESUMEN

Endosulfan (1,4,5,6,7,7-hexachloro-8,9,10-trinorborn-5-en-2,3-ylenebismet-hylene) is correlated with endocrine disruption, reproductive, and immune dysfunctions. Recently, endosulfan was shown to have an effect on inflammatory pathways, but its influence on cyclooxygenase-2(COX-2) expression is unclear. This study investigated the effects of COX-2 and molecular mechanisms by endosulfan in murine macrophage RAW 264.7 cells. Endosulfan significantly induced COX-2 protein and mRNA levels, as well as COX-2 promoter-driven luciferase activity and the production of prostaglandin E2, a major COX-2 metabolite. Transfection experiments with several human COX-2 promoter constructs revealed that endosulfan activated NF-κB, C/EBP, AP-1, and CREB. Moreover, Akt and mitogen-activated protein kinases (MAPK) were significantly activated by endosulfan. Moreover, endosulfan increased production of the ROS and the ROS-producing NAPDH-oxidase (NOX) family oxidases, NOX2, and NOX3. Endosulfan-induced Akt/MAPK pathways and COX-2 expression were attenuated by DPI, a specific NOX inhibitor, and the ROS scavenger N-acetylcysteine. These results demonstrate that endosulfan induces COX-2 expression via NADPH oxidase, ROS, and Akt/MAPK pathways. These findings provide further insight into the signal transduction pathways involved in the inflammatory effects of endosulfan.


Asunto(s)
Ciclooxigenasa 2/genética , Endosulfano/toxicidad , Macrófagos/efectos de los fármacos , NADPH Oxidasas/metabolismo , Acetilcisteína/farmacología , Animales , Línea Celular , Dinoprostona/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transfección
12.
Toxicol Appl Pharmacol ; 280(1): 138-48, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25110054

RESUMEN

Induction of cytochrome P450 (CYP) 1A1 and CYP1B1 by environmental xenobiotic chemicals or endogenous ligands through the activation of the aryl hydrocarbon receptor (AhR) has been implicated in a variety of cellular processes related to cancer, such as transformation and tumorigenesis. Here, we investigated the effects of the anti-diabetes drug metformin on expression of CYP1A1 and CYP1B1 in breast cancer cells under constitutive and inducible conditions. Our results indicated that metformin down-regulated the expression of CYP1A1 and CYP1B1 in breast cancer cells under constitutive and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced conditions. Down-regulation of AhR expression was required for metformin-mediated decreases in CYP1A1 and CYP1B1 expression, and the metformin-mediated CYP1A1 and CYP1B1 reduction is irrelevant to estrogen receptor α (ERα) signaling. Furthermore, we found that metformin markedly down-regulated Sp1 protein levels in breast cancer cells. The use of genetic and pharmacological tools revealed that metformin-mediated down-regulation of AhR expression was mediated through the reduction of Sp1 protein. Metformin inhibited endogenous AhR ligand-induced CYP1A1 and CYP1B1 expression by suppressing tryptophan-2,3-dioxygenase (TDO) expression in MCF-7 cells. Finally, metformin inhibits TDO expression through a down-regulation of Sp1 and glucocorticoid receptor (GR) protein levels. Our findings demonstrate that metformin reduces CYP1A1 and CYP1B1 expression in breast cancer cells by down-regulating AhR signaling. Metformin would be able to act as a potential chemopreventive agent against CYP1A1 and CYP1B1-mediated carcinogenesis and development of cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Citocromo P-450 CYP1A1/antagonistas & inhibidores , Citocromo P-450 CYP1B1/antagonistas & inhibidores , Regulación hacia Abajo/efectos de los fármacos , Metformina/farmacología , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Citocromo P-450 CYP1A1/biosíntesis , Citocromo P-450 CYP1B1/biosíntesis , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/fisiología , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Metformina/uso terapéutico , Receptores de Hidrocarburo de Aril/metabolismo
13.
Food Chem Toxicol ; 71: 51-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24930757

RESUMEN

TRAIL induces apoptosis in a variety of tumor cells. However, development of resistance to TRAIL is a major obstacle to more effective cancer treatment. Therefore, novel pharmacological agents that enhance sensitivity to TRAIL are necessary. In the present study, we investigated the molecular mechanisms by which ilimaquinone isolated from a sea sponge sensitizes human colon cancer cells to TRAIL. Ilimaquinone pretreatment significantly enhanced TRAIL-induced apoptosis in HCT 116 cells and sensitized colon cancer cells to TRAIL-induced apoptosis through increased caspase-8, -3 activation, PARP cleavage, and DNA damage. Ilimaquinone also reduced the cell survival proteins Bcl2 and Bcl-xL, while strongly up-regulating death receptor (DR) 4 and DR5 expression. Induction of DR4 and DR5 by ilimaquinone was mediated through up-regulation of CCAAT/enhancer-binding protein homologous protein (CHOP). The up-regulation of CHOP, DR4 and DR5 expression was mediated through activation of extracellular-signal regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) signaling pathways. Finally, the generation of ROS was required for CHOP and DR5 up-regulation by ilimaquinone. These results demonstrate that ilimaquinone enhanced the sensitivity of human colon cancer cells to TRAIL-induced apoptosis through ROS-ERK/p38 MAPK-CHOP-mediated up-regulation of DR4 and DR5 expression, suggesting that ilimaquinone could be developed into an adjuvant chemotherapeutic drug.


Asunto(s)
Apoptosis/fisiología , Neoplasias del Colon/patología , Quinonas/toxicidad , Receptores de Muerte Celular/efectos de los fármacos , Sesquiterpenos/toxicidad , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Activación Enzimática , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Espectroscopía de Protones por Resonancia Magnética , Quinonas/química , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Muerte Celular/metabolismo , Sesquiterpenos/química , Factor de Transcripción CHOP/metabolismo , Regulación hacia Arriba/efectos de los fármacos
14.
Phytomedicine ; 21(8-9): 1053-61, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24854572

RESUMEN

PURPOSE: The consequences of precipitously rising allergic skin inflammation rates worldwide have accelerated the risk of atopic dermatitis (AD). Natural product-based agents with good efficacy and low risk of side effects offer promising prevention and treatment strategies for inflammation-related diseases. We have already reported that Platycodon grandiflorum root-derived saponins (Changkil saponins, CKS) have many pharmacological effects, including anti-inflammatory and anti-allergic effects, but its influence on AD remains unclear. Therefore, we evaluated the inhibitory effect of CKS, mainly platycodin D, on AD-like skin symptoms in mice and the possible mechanisms in cells. METHODS: Mice were sensitized and challenged with 2,4-dinitrochlorobenzene (DNCB). Four weeks after challenge, mice were treated with oral administration of CKS for 4 weeks. In addition, cells were used to evaluate the effect of CKS, mainly platycodin D, on the TARC expression regulated mechanism. RESULTS: CKS attenuated DNCB-induced dermatitis severity, serum levels of IgE and TARC, and mRNA expression of TARC, TNF-α, IFN-γ, IL-4, IL-5, and IL-13 in mice. Histopathological examination showed reduced thickness of the epidermis/dermis and dermal infiltration of inflammatory cells and mast cells in the ears. Moreover, CKS and platycodin D inhibited TNF-α/IFN-γ-induced TARC expression through the suppression of NF-κB and STAT1 and induction of Nrf2/ARE-mediated hemeoxygenase-1 (HO-1) expression in cells. CONCLUSION: We suggest that CKS and platycodin D inhibited the development of AD-like skin symptoms by regulating cytokine mediators and may be an effective alternative therapy for AD-like skin symptoms.


Asunto(s)
Antialérgicos/farmacología , Dermatitis Atópica/tratamiento farmacológico , Extractos Vegetales/farmacología , Platycodon/química , Saponinas/farmacología , Triterpenos/farmacología , Animales , Antialérgicos/química , Antialérgicos/aislamiento & purificación , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Dermatitis Atópica/inducido químicamente , Dinitroclorobenceno/efectos adversos , Regulación de la Expresión Génica , Genes Reporteros , Hemo-Oxigenasa 1/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Humanos , Inmunoglobulina E/sangre , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Raíces de Plantas/química , Factor de Transcripción STAT1/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo , Saponinas/química , Saponinas/aislamiento & purificación , Triterpenos/química , Triterpenos/aislamiento & purificación
15.
Toxicol Appl Pharmacol ; 277(1): 39-48, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24631339

RESUMEN

Leptin, a hormone with multiple biological actions, is produced predominantly by adipose tissue. Among its functions, leptin can stimulate tumour cell growth. Oestrogen receptor α (ERα), which plays an essential role in breast cancer development, can be transcriptionally activated in a ligand-independent manner. In this study, we investigated the effect of leptin on CYP1B1 expression and its mechanism in breast cancer cells. Leptin induced CYP1B1 protein, messenger RNA expression and promoter activity in ERα-positive MCF-7 cells but not in ERα-negative MDA-MB-231 cells. Additionally, leptin increased 4-hydroxyoestradiol in MCF-7 cells. Also, ERα knockdown by siRNA significantly blocked the induction of CYP1B1 expression by leptin, indicating that leptin induced CYP1B1 expression via an ERα-dependent mechanism. Transient transfection with CYP1B1 deletion promoter constructs revealed that the oestrogen response element (ERE) plays important role in the up-regulation of CYP1B1 by leptin. Furthermore, leptin stimulated phosphorylation of ERα at serine residues 118 and 167 and increased ERE-luciferase activity, indicating that leptin induced CYP1B1 expression by ERα activation. Finally, we found that leptin activated ERK and Akt signalling pathways, which are upstream kinases related to ERα phosphorylation induced by leptin. Taken together, our results indicate that leptin-induced CYP1B1 expression is mediated by ligand-independent activation of the ERα pathway as a result of the activation of ERK and Akt in MCF-7 cells.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/biosíntesis , Neoplasias de la Mama/fisiopatología , Receptor alfa de Estrógeno/metabolismo , Leptina/farmacología , Línea Celular Tumoral , Citocromo P-450 CYP1B1 , Estrógenos de Catecol/biosíntesis , Femenino , Humanos , Células MCF-7 , Fosforilación , ARN Mensajero , ARN Interferente Pequeño , Elementos de Respuesta , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Transfección , Regulación hacia Arriba/efectos de los fármacos
16.
Nutr Cancer ; 66(1): 140-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24341702

RESUMEN

Transforming growth factor ß (TGFß) is a multifunctional cytokine that induces growth arrest, tissue fibrosis, and epithelial-mesenchymal transition (EMT) through activation of Smad and non-Smad signaling pathways. EMT is the differentiation switch by which polarized epithelial cells differentiate into contractile and motile mesenchymal cells. Our previous studies have shown that saponins from the roots of Platycodon grandiflorum (CKS) have antiinflammatory, antioxidant, antimetastatic, and hepatoprotective effects. In this study, we investigated the inhibitory effect of CKS on TGFß1-induced alterations characteristic of EMT in human lung carcinoma A549 cells. We found that CKS-treated cells displayed inhibited TGFß1-mediated E-cadherin downregulation and Vimentin upregulation and also retained epithelial morphology. Furthermore, TGFß1-increased Snail expression, a repressor of E-cadherin and an inducer of the EMT, was reduced by CKS. CKS inhibited TGFß1-induced phosphorylation of Akt, ERK1/2, and glycogen synthase kinase-3ß (GSK-3ß). Inhibition of PI3K/Akt and ERK1/2 also blocked TGFß1-induced GSK-3ß phosphorylation and Snail activation. Furthermore, TGFß1-increased Snail expression was reduced by selective inhibitors of Akt and ERK1/2. Moreover, CKS treatment attenuated TGFß1-induced Smad2/3 phosphorylation and upregulated Smad7 expression. These results indicate that pretreatment with the CKS inhibits the TGFß1-induced EMT through PI3K/Akt, ERK1/2, GSK-3ß and Smad2/3 in human lung carcinoma cells.


Asunto(s)
Represión Epigenética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Platycodon/química , Saponinas/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Regulación hacia Abajo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Extractos Vegetales/farmacología , Raíces de Plantas/química , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo
17.
Food Chem Toxicol ; 64: 126-34, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24296130

RESUMEN

Genipin is a compound found in gardenia fruit extract with diverse pharmacological activities. However, the mechanism underlying genipin-induced cyclooxygenase-2 (COX-2) expression remains unknown. In this study, we investigated the effects of genipin on COX-2 expression and determined that exposure to genipin dose-dependently enhanced the production of prostaglandin E2 (PGE2), a major COX-2 metabolite, in RAW 264.7 cells. These effects were mediated by genipin-induced activation of the COX-2 promoter, as well as AP-1 and NF-κB luciferase constructs. Phosphatidylinositol-3-kinase/Akt and MAPKs were also significantly activated by genipin, and Akt and MAPKs inhibitors (PD98059, SB20358, SP600125, and LY294002) inhibited genipin-induced COX-2 expression. Moreover, genipin increased production of the ROS and the ROS-producing NAPDH-oxidase (NOX) family oxidases, NOX2 and NOX3. Inhibition of NADPH with diphenyleneiodonium attenuated ROS production, COX-2 expression and NF-κB and AP-1 activation. These results suggest that the molecular mechanism mediating ROS-dependent COX-2 up-regulation and PGE2 production by genipin involves activation of Akt, MAPKs and AP-1/NF-κB.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Iridoides/farmacología , Macrófagos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , NADPH Oxidasas/metabolismo , FN-kappa B/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Línea Celular , Macrófagos/enzimología , Macrófagos/metabolismo , Ratones , Transducción de Señal
18.
Food Chem ; 141(3): 2591-9, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23870999

RESUMEN

Piperine is a bioactive component of black pepper, Piper nigrum Linn, commonly used for daily consumption and in traditional medicine. Here, the molecular mechanisms by which piperine exerts antitumor effects in HER2-overexpressing breast cancer cells was investigated. The results showed that piperine strongly inhibited proliferation and induced apoptosis through caspase-3 activation and PARP cleavage. Furthermore, piperine inhibited HER2 gene expression at the transcriptional level. Blockade of ERK1/2 signaling by piperine significantly reduced SREBP-1 and FAS expression. Piperine strongly suppressed EGF-induced MMP-9 expression through inhibition of AP-1 and NF-κB activation by interfering with ERK1/2, p38 MAPK, and Akt signaling pathways resulting in a reduction in migration. Finally, piperine pretreatment enhanced sensitization to paclitaxel killing in HER2-overexpressing breast cancer cells. Our findings suggest that piperine may be a potential agent for the prevention and treatment of human breast cancer with HER2 overexpression.


Asunto(s)
Alcaloides/farmacología , Antineoplásicos Fitogénicos/farmacología , Benzodioxoles/farmacología , Neoplasias de la Mama/genética , Piperidinas/farmacología , Extractos Vegetales/farmacología , Alcamidas Poliinsaturadas/farmacología , Receptor ErbB-2/genética , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/fisiopatología , Caspasa 3/genética , Caspasa 3/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Receptor ErbB-2/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Toxicol Appl Pharmacol ; 271(2): 229-38, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23707609

RESUMEN

Resistance to therapy is the major obstacle to more effective cancer treatment. Heme oxygenase-1 (HO-1) is often highly up-regulated in tumor tissues, and its expression is further increased in response to therapies. It has been suggested that inhibition of HO-1 expression is a potential therapeutic approach to sensitize tumors to chemotherapy and radiotherapy. In this study, we tested the hypothesis that the anti-tumor effects of metformin are mediated by suppression of HO-1 expression in cancer cells. Our results indicate that metformin strongly suppresses HO-1 mRNA and protein expression in human hepatic carcinoma HepG2, cervical cancer HeLa, and non-small-cell lung cancer A549 cells. Metformin also markedly reduced Nrf2 mRNA and protein levels in whole cell lysates and suppressed tert-butylhydroquinone (tBHQ)-induced Nrf2 protein stability and antioxidant response element (ARE)-luciferase activity in HepG2 cells. We also found that metformin regulation of Nrf2 expression is mediated by a Keap1-independent mechanism and that metformin significantly attenuated Raf-ERK signaling to suppress Nrf2 expression in cancer cells. Inhibition of Raf-ERK signaling by PD98059 decreased Nrf2 mRNA expression in HepG2 cells, confirming that the inhibition of Nrf2 expression is mediated by an attenuation of Raf-ERK signaling in cancer cells. The inactivation of AMPK by siRNA, DN-AMPK or the pharmacological AMPK inhibitor compound C, revealed that metformin reduced HO-1 expression in an AMPK-independent manner. These results highlight the Raf-ERK-Nrf2 axis as a new molecular target in anticancer therapy in response to metformin treatment.


Asunto(s)
Hemo-Oxigenasa 1/antagonistas & inhibidores , Hipoglucemiantes/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metformina/farmacología , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Factor 2 Relacionado con NF-E2/fisiología , Neoplasias/enzimología , Quinasas raf/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Núcleo Celular/química , Supervivencia Celular/efectos de los fármacos , Citosol/química , Galactosidasas/metabolismo , Humanos , Indicadores y Reactivos , Luciferasas/metabolismo , Plásmidos/genética , ARN/biosíntesis , ARN/genética , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transfección
20.
Food Chem Toxicol ; 56: 231-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23485615

RESUMEN

The purpose of this study was to investigate the anti-fibrotic effects of the aqueous extract of the Platycodi Radix root (Changkil: CK) on dimethylnitrosamine (DMN)-induced liver fibrosis in rats. DMN treatment for 4 weeks led to marked liver fibrosis as assessed by serum biochemistry, histopathological examination, and hepatic lipid peroxidation and collagen content. CK significantly inhibited DMN-induced increases in serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities, fibrosis score, and hepatic malondialdehyde and collagen content. CK also inhibited DMN-induced reductions in rat body and liver weights. Reverse transcription polymerase chain reaction (RT-PCR) and western blot analyses revealed that CK inhibited DMN-induced increases in matrix metalloproteinase-13 (MMP-13), tissue inhibitor of metalloproteinase-1 (TIMP-1), and tumor necrosis factor-α (TNF-α) mRNA, and collagen type I and α-smooth muscle actin protein. DMN-induced cyclooxygenase-2 (COX-2) expression and nuclear factor-kappa B (NF-κB) activation was reduced by CK treatment. Furthermore, CK induced activation of nuclear erythroid 2-related factor 2 (Nrf2)-mediated antioxidant enzymes such as γ-glutamylcysteine synthetase (γ-GCS), heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase 1 (NQO1), and glutathione-S-transferase (GST) in HepG2 cells. These results demonstrated that CK attenuates DMN-induced liver fibrosis through the activation of Nrf2-mediated antioxidant enzymes.


Asunto(s)
Antioxidantes/farmacología , Dimetilnitrosamina/efectos adversos , Cirrosis Hepática/patología , Extractos Vegetales/farmacología , Actinas/metabolismo , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Colágeno Tipo I/metabolismo , Ciclooxigenasa 2/metabolismo , Glutamato-Cisteína Ligasa/metabolismo , Glutatión Transferasa/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Células Hep G2 , Humanos , Peroxidación de Lípido/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/tratamiento farmacológico , Masculino , Malondialdehído/sangre , Metaloproteinasa 13 de la Matriz/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Raíces de Plantas/química , Platycodon , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...