Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Endocrinol Metab (Seoul) ; 36(1): 96-105, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33677931

RESUMEN

BACKGROUND: Several cancers show increased levels of lactate dehydrogenase A (LDHA), which are associated with cancer progression. However, it remains unclear whether LDHA levels are associated with papillary thyroid cancer (PTC) aggressiveness or with the presence of the PTC prognostic marker, the BRAFV600E mutation. This study aimed to evaluate the potential of LDHA as a PTC prognostic marker. METHODS: LDHA expression was examined in 83 PTC tissue specimens by immunohistochemistry. Human thyroid cell lines were genetically manipulated to overexpress BRAFV600E or were treated with a BRAF-specific short hairpin RNA (shBRAF), whose effects on LDHA expression were evaluated by Western blotting. Data from 465 PTC patients were obtained from The Cancer Genome Atlas (TCGA) database and analyzed to validate the in vitro results. RESULTS: LDHA was aberrantly overexpressed in PTC. Intense immunostaining for LDHA was observed in PTC specimens carrying mutated BRAF, whereas the intensity was less in wild-type BRAF samples. Overexpression of BRAFV600E resulted in LDHA upregulation, whereas treatment with shBRAF downregulated LDHA in human thyroid cell lines. Furthermore, LDHA mRNA expression was significantly elevated and associated with BRAFV600E expression in thyroid cancer tissues from TCGA database. Additionally, LDHA overexpression was found to be correlated with aggressive clinical features of PTC, such as lymph node metastases and advanced tumor stages. CONCLUSION: LDHA overexpression is associated with the BRAFV600E mutation and an aggressive PTC behavior. Therefore, LDHA may serve as a biomarker and therapeutic target in PTC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Papilar , L-Lactato Deshidrogenasa/metabolismo , Neoplasias de la Tiroides , Carcinoma Papilar/patología , Humanos , Lactato Deshidrogenasa 5 , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias de la Tiroides/patología
2.
Mol Oncol ; 12(10): 1811-1826, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30171795

RESUMEN

Transforming growth factor ß (TGFß) is overexpressed in advanced cancers and promotes tumorigenesis by inducing epithelial-mesenchymal transition (EMT), which enhances invasiveness and metastasis. Although we previously reported that EMT could be induced by increasing CK2 activity alone, it is not known whether CK2 also plays an essential role in TGFß-induced EMT. Therefore, in the present study, we investigated whether TGFß signaling could activate CK2 and, if so, whether such activation is required for TGFß-induced EMT. We found that CK2 is activated by TGFß treatment, and that activity peaks at 48 h after treatment. CK2 activation is dependent on TGFß receptor (TGFBR) I kinase activity, but independent of SMAD4. Inhibition of CK2 activation through the use of either a CK2 inhibitor or shRNA against CSNK2A1 inhibited TGFß-induced EMT. TGFß signaling decreased CK2ß but did not affect CK2α protein levels, resulting in a quantitative imbalance between the catalytic α and regulatory ß subunits, thereby increasing CK2 activity. The decrease in CK2ß expression was dependent on TGFBRI kinase activity and the ubiquitin-proteasome pathway. The E3 ubiquitin ligases responsible for TGFß-induced CK2ß degradation were found to be CHIP and WWP1. Okadaic acid (OA) pretreatment protected CK2ß from TGFß-induced degradation, suggesting that dephosphorylation of CK2ß by an OA-sensitive phosphatase might be required for CK2 activation in TGFß-induced EMT. Collectively, our results suggest CK2 as a therapeutic target for the prevention of EMT and metastasis of cancers.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Línea Celular Tumoral , Activación Enzimática , Células HEK293 , Humanos , Fosforilación , Proteolisis , Transducción de Señal/efectos de los fármacos , Proteína Smad4 , Ubiquitina-Proteína Ligasas/metabolismo
3.
J Cell Biochem ; 119(10): 8501-8510, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30015359

RESUMEN

Protein kinase CK2 is active in cancer cells. Previously, we reported that increased CK2 activity could induce epithelial mesenchymal transition of cancer cells. CK2 also induced epithelial mesenchymal transition in colon cancer cell lines such as HT29 and SW620, and the transitioned cells (CK2α cells) became more proliferative than the controls. We assumed that CK2 could affect cancer cell growth by modulating their energy metabolism. Here, we examined the molecular effects of CK2 on the glucose metabolism of cancer cells. We found that CK2α cells consumed more glucose and produced more lactate than control cells did. An XF glycolysis stress test showed that aerobic glycolysis was augmented up to the cancer cell's maximal glycolytic capacity in CK2α cells. Molecular analysis revealed that pyruvate kinase M1 was downregulated and pyruvate kinase M2 was nuclear localized in CK2α cells. Consequently, the expression and activity of lactate dehydrogenase A (LDHA) were upregulated. Treatment with FX11-a specific LDHA inhibitor-or clustered regularly interspaced short palindromic repeats (CRISPR)-mediated knockout of LDHA inhibited the CK2-driven proliferation of cancer cells. We conclude that CK2 augments the Warburg effect, resulting in increased proliferation of cancer cells.


Asunto(s)
Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Glucólisis , Piruvato Quinasa/genética , Piruvato Quinasa/metabolismo , Quinasa de la Caseína II/genética , Quinasa de la Caseína II/metabolismo , Proliferación Celular , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Glucosa/metabolismo , Células HEK293 , Células HT29 , Humanos , Isoenzimas/metabolismo , Lactato Deshidrogenasa 5/antagonistas & inhibidores , Lactato Deshidrogenasa 5/genética , Lactato Deshidrogenasa 5/metabolismo , Ácido Láctico/metabolismo , Naftalenos/farmacología , Activación Transcripcional
4.
Oncogenesis ; 7(3): 27, 2018 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-29540671

RESUMEN

Epithelial-mesenchymal transition (EMT) is a critical process in invasion and metastasis of cancer cells. E-cadherin to N-cadherin switching is considered a molecular hallmark of EMT. Recently, we reported that increased CK2 activity fully induces E-cadherin to N-cadherin switching, but the molecular mechanisms of N-cadherin upregulation are unknown. In this study, we examined how N-cadherin is upregulated by CK2. N-cadherin promoter analysis and ChIP analysis identified and confirmed myeloid zinc finger 1 (MZF1) as an N-cadherin transcription factor. Molecular analysis showed that MZF1 directly interacts with CK2 and is phosphorylated at serine 27. Phosphorylation stabilizes MZF1 and induces transcription of N-cadherin. MZF1 knockdown (MKD) in N-cadherin-expressing cancer cells downregulates N-cadherin expression and reverts the morphology from spindle and fibroblast-like to a rounded, epithelial shape. In addition, we showed that that MKD reduced the motility and invasiveness of N-cadherin-expressing cancer cells. Collectively, these data indicate that N-cadherin upregulation in CK2-mediated E-cadherin to N-cadherin switching is dependent on phosphorylation-mediated MZF1 stabilization. CK2 could be a good therapeutic target for the prevention of metastasis.

5.
Biochim Biophys Acta Gene Regul Mech ; 1860(9): 962-972, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28757384

RESUMEN

Expression of the POK family protein ZNF509L, and -its S1 isoform, is induced by p53 upon exposure to genotoxic stress. Due to alternative splicing of the ZNF509 primary transcript, ZNF509S1 lacks the 6 zinc-fingers and C-terminus of ZNF509L, resulting in only one zinc-finger. ZNF509L and -S1 inhibit cell proliferation by activating p21/CDKN1A and RB transcription, respectively. When cells are exposed to severe DNA damage, p53 activates PUMA (p53-upregulated modulator of apoptosis) transcription. Interestingly, apoptosis due to transcriptional activation of PUMA by p53 is attenuated by ZNF509S1. Thus we investigated the molecular mechanism(s) underlying the transcriptional attenuation and anti-apoptotic effects of ZNF509S1. We show that ZNF509S1 modulation of p53 activity is important in PUMA gene transcription by modulating post-translational modification of p53 by p300. ZNF509S1 directly interacts with p53 and inhibits p300-mediated acetylation of p53 lysine K382, with deacetylation of p53 K382 leading to decreased DNA binding at the p53 response element 1 of the PUMA promoter. ZNF509S1 may play a role not only in cell cycle arrest, by activating RB expression, but also in rescuing cells from apoptotic death by repressing PUMA expression in cells exposed to severe DNA damage.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Regulación hacia Abajo/fisiología , Puma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Animales , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN/fisiología , Proteína p300 Asociada a E1A , Células HCT116 , Células HEK293 , Humanos , Regiones Promotoras Genéticas/fisiología , Unión Proteica/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Activación Transcripcional/fisiología , Dedos de Zinc/fisiología
6.
Drug Dev Ind Pharm ; 43(9): 1557-1565, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28554216

RESUMEN

OBJECTIVES: Nanoparticulation using fat and supercritical fluid (NUFSTM) is a drug delivery platform technology enabling efficient and effective formulation of poorly soluble drugs. We performed experiments to examine whether NUFS™ could improve poor bioavailability and reduce fed-fasted bioavailability variances of erlotinib (Ert). METHODS: NUFS-Ert was prepared using NUFS™ technology; its physical properties were characterized, and drug release was measured. Furthermore, in vitro and in vivo efficacy tests and pharmacokinetic analysis were performed. RESULTS: NUFS-Ert nanoparticles had an average size of 250 nm and were stable for 2 months at 40 °C, 4 °C, and room temperature. The dissolution rate of NUFS-Ert increased in bio-relevant dissolution media. NUFS-Ert was more potent in inhibiting EGF signaling and in suppressing the proliferation of A549, a human non-small cell lung cancer cell line. Furthermore, A549 xenografts in BALB/c nude mice treated with NUFS-Ert regressed more efficiently than those in the mice treated with vehicle or Tarceva®. In addition, experimental lung metastasis was more efficiently inhibited by NUFS-Ert than by Tarceva®. The relative bioavailability of NUFS-Ert compared with that of Tarceva® was 550% and the ratio of the area under the concentration-time curve (AUC) of fed state to the AUC of fasted state was 1.8 for NUFS-Ert and 5.8 for Tarceva®. CONCLUSIONS: NUFS-Ert could improve poor bioavailability and reduce fed-fasted bioavailability variances of Ert. NUFS-Ert was more efficacious than Tarceva®.


Asunto(s)
Antineoplásicos/farmacocinética , Disponibilidad Biológica , Clorhidrato de Erlotinib/farmacocinética , Excipientes/química , Neoplasias Pulmonares/tratamiento farmacológico , Nanopartículas/química , Animales , Antineoplásicos/química , Química Farmacéutica , Clorhidrato de Erlotinib/química , Clorhidrato de Erlotinib/farmacología , Humanos , Ratones Desnudos , Solubilidad
7.
Oncotarget ; 7(50): 83001-83016, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27792998

RESUMEN

Recent research has focused on the impact of long noncoding RNA (lncRNA) in cervical carcinogenesis. However, whether HOXA11 antisense (HOXA11-AS) is involved in cervical cancer remains to be elucidated. In the present study, we examined HOXA11-AS expression levels in cervical cancer patients and determined the relationships between HOXA11-AS expression and clinicopathological factors. We also investigated the bio-functional consequences of HOXA11-AS overexpression both in vitro and in vivo. HOXA11-AS expression was significantly greater in tissues from patients with cervical cancer than in control patients (P<0.001). Multivariate analysis showed that high HOXA11-AS was an independent prognosticator of overall survival (Hazard ratio=2.450, P=0.032). HOXA11-AS overexpression enhanced cell proliferation, migration, and tumor invasion in vitro, whereas HOXA11-AS knockdown inhibited these biologic aggressive features. These adverse changes were accompanied by characteristics of epithelial-mesenchymal transition (EMT). In vivo xenograft experiments using the siHOXA11-AS-transfected HeLa cells revealed that HOXA11-AS strongly induced tumor growth. Furthermore, we found that HOXA11-AS knockdown decreased cancer stemness and triggered the EMT program. In conclusion, HOXA11-AS overexpression correlated with poor survival in patients with cervical cancer. Thus, HOXA11-AS may be a pivotal target for exploring novel cervical cancer therapeutics.


Asunto(s)
Células Madre Neoplásicas/metabolismo , ARN Largo no Codificante/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Adulto , Animales , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos BALB C , Persona de Mediana Edad , Análisis Multivariante , Invasividad Neoplásica , Células Madre Neoplásicas/patología , Interferencia de ARN , ARN Largo no Codificante/genética , Factores de Riesgo , Transducción de Señal , Factores de Tiempo , Transfección , Carga Tumoral , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/mortalidad , Neoplasias del Cuello Uterino/patología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Biochim Biophys Acta ; 1859(11): 1429-1439, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27646874

RESUMEN

Matrix metalloproteinases (MMPs) are zinc-containing endopeptidases that play roles in cell proliferation, migration, differentiation, angiogenesis, and apoptosis. The expression of MMP gene is tightly regulated and shows cell- and tissue-specific expression patterns. Despite their differential expression, MMP genes have AP-1 (activator protein-1) binding elements within their promoters. Interestingly, c-JUN phosphorylation by cytokine signaling decreased its interaction with NCoR, but increased its interaction with p300, resulting in activation of MMP gene transcription. Here, we found that Zbtb7c (Kr-pok) is a critical component of a transcriptional repressor complex containing c-Jun and NCoR. c-Jun, bound at AP-1, interacts with Zbtb7c, which in turn recruits an NCoR/Hdac3 complex to repress several Mmp (-8, -10, -13, and -16) genes. The molecular interaction between c-Jun and Zbtb7c also prevents phosphorylation of c-Jun by p-Jnk, However, Zbtb7c phosphorylation by p-Jnk (induced by TNFα), and its (Zbtb7c) subsequent degradation by the ubiquitin-mediated proteasomal pathway, leads to c-Jun phosphorylation by p-Jnk. Promoter-bound p-c-Jun then recruits the coactivator p300 to upregulate Mmp gene. Overall, these findings show that Zbtb7c is a key molecule that recruits an NCoR/Hdac3 complex to inhibit phosphorylation of c-Jun, and thereby repress Mmp gene expression.


Asunto(s)
Metaloproteinasas de la Matriz/genética , Proteínas/genética , Transcripción Genética , Secuencia de Aminoácidos , Animales , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas , Proteínas/química , Proteolisis , Homología de Secuencia de Aminoácido , Factor de Necrosis Tumoral alfa/administración & dosificación , Ubiquitinación
9.
Biomed Res Int ; 2016: 8432759, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26989693

RESUMEN

Diabetes and obesity represent the major health problems and the most age-related metabolic diseases. Protein-tyrosine phosphatase 1B (PTP1B) has emerged as an important regulator of insulin signal transduction and is regarded as a pharmaceutical target for metabolic disorders. To find novel natural materials presenting therapeutic activities against diabetes and obesity, we screened various herb extracts using a chip screening allowing the determination of PTP1B inhibitory effects of the tested compounds using insulin receptor (IR) as the substrate. Cudrania tricuspidata leaves (CTe) had a strong inhibitory effect on PTP1B activity and substantially inhibited fat accumulation in 3T3-L1 cells. CTe was orally administrated to diet-induced obesity (DIO) mice once daily for 3 weeks after which changes in glucose, insulin metabolism, and fat accumulation were examined. Hepatic enzyme markers (aspartate aminotransferase, AST, and alanine aminotransferase, ALT) and total fat mass and triglyceride levels decreased in CTe-treated mice, whereas body weight and total cholesterol concentration slightly decreased. CTe increased the phosphorylation of IRS-1 and Akt in liver tissue. Furthermore, CTe treatment significantly lowered blood glucose levels and improved insulin secretion in DIO mice. Our results strongly suggest that CTe may represent a promising therapeutic substance against diabetes and obesity.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/biosíntesis , Células 3T3-L1 , Animales , Diabetes Mellitus/genética , Diabetes Mellitus/patología , Humanos , Insulina/sangre , Resistencia a la Insulina/genética , Hígado/efectos de los fármacos , Hígado/patología , Ratones , Ratones Obesos , Moraceae/química , Obesidad/genética , Obesidad/patología , Extractos Vegetales/química , Transducción de Señal/efectos de los fármacos
11.
Nucleic Acids Res ; 43(3): 1609-25, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25609694

RESUMEN

The NF-κB is found in almost all animal cell types and is involved in a myriad of cellular responses. Aberrant expression of NF-κB has been linked to cancer, inflammatory diseases and improper development. Little is known about transcriptional regulation of the NF-κB family member gene RelA/p65. Sp1 plays a key role in the expression of the RelA/p65 gene. ZBTB2 represses transcription of the gene by inhibiting Sp1 binding to a Sp1-binding GC-box in the RelA/p65 proximal promoter (bp, -31 to -21). Moreover, recent studies revealed that RelA/p65 directly binds to the peroxisome proliferator-activated receptor-γ coactivator1α (PGC1α) to decrease transcriptional activation of the PGC1α target gene PDK4, whose gene product inhibits pyruvate dehydrogenase (PDH), a key regulator of TCA cycle flux. Accordingly, we observed that RelA/p65 repression by ZBTB2 indirectly results in increased PDK4 expression, which inhibits PDH. Consequently, in cells with ectopic ZBTB2, the concentrations of pyruvate and lactate were higher than those in normal cells, indicating changes in glucose metabolism flux favoring glycolysis over the TCA cycle. Knockdown of ZBTB2 in mouse xenografts decreased tumor growth. ZBTB2 may increase cell proliferation by reprogramming glucose metabolic pathways to favor glycolysis by upregulating PDK4 expression via repression of RelA/p65 expression.


Asunto(s)
Proteínas Serina-Treonina Quinasas/genética , Proteínas Represoras/fisiología , Factor de Transcripción ReIA/genética , Transcripción Genética , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Regiones Promotoras Genéticas , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción Sp1/metabolismo
12.
Bioorg Med Chem ; 22(17): 4553-65, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25131958

RESUMEN

Protein kinase casein kinase 2 (PKCK2) is a constitutively active, growth factor-independent serine/threonine kinase, and changes in PKCK2 expression or its activity are reported in many cancer cells. To develop a novel PKCK2 inhibitor(s), we first performed cell-based phenotypic screening using 4000 chemicals purchased from ChemDiv chemical libraries (2000: randomly selected; 2000: kinase-biased) and performed in vitro kinase assay-based screening using hits found from the first screening. We identified compound 24 (C24)[(Z)-ethyl 5-(4-chlorophenyl)-2-(3,4-dihydroxybenzylidene)-7-methyl-3-oxo-3,5-dihydro-2H-thiazolo[3,2-a] pyrimidine-6-carboxylate] as a novel inhibitor of PKCK2 that is more potent and selective than 4,5,6,7-tetrabromobenzotriazole (TBB). In particular, compound 24 [half maximal inhibitory concentration (IC50)=0.56µM] inhibited PKCK2 2.2-fold more efficiently than did TBB (IC50=1.24µM), which is quite specific toward PKCK2 with respect to ATP binding, in a panel of 31 human protein kinases. The Ki values of compound 24 and TBB for PKCK2 were 0.78µM and 2.70µM, respectively. Treatment of cells with compound 24 inhibited endogenous PKCK2 activity and showed anti-proliferative and pro-apoptotic effects against stomach and hepatocellular cancer cell lines more efficiently than did TBB. As expected, compound 24 also enabled tumor necrosis factor-related apoptosis inducing ligand (TRAIL)-resistant cancer cells to be sensitive toward TRAIL. In comparing the molecular docking of compound 24 bound to PKCK2α versus previously reported complexes of PKCK2 with other inhibitors, our findings suggest a new scaffold for specific PKCK2α inhibitors. Thus, compound 24 appears to be a selective, cell-permeable, potent, and novel PKCK2 inhibitor worthy of further characterization.


Asunto(s)
Antineoplásicos/farmacología , Quinasa de la Caseína II/antagonistas & inhibidores , Catecoles/farmacología , Compuestos Heterocíclicos con 2 Anillos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Quinasa de la Caseína II/metabolismo , Catecoles/síntesis química , Catecoles/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células Hep G2 , Compuestos Heterocíclicos con 2 Anillos/síntesis química , Compuestos Heterocíclicos con 2 Anillos/química , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
13.
Toxicology ; 322: 23-33, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24793912

RESUMEN

The epithelial-mesenchymal transition (EMT) is a pivotal cellular process during which epithelial polarized cells become motile mesenchymal-appearing cells, which, in turn, promotes the metastatic potential of cancer. Ginseng is a perennial plant belonging to the genus Panax that exhibits a wide range of pharmacological and physiological activities. Ginsenosides 20-Rg3, which is the active component of ginseng, has various medical effects, such as anti-tumorigenic, anti-angiogenesis, and anti-fatiguing activities. In addition, ginsenosides 20(S)-Rg3 and 20(R)-Rg3 are epimers, and this epimerization is produced by steaming. However, the possible role of 20(S)-Rg3 and 20(R)-Rg3 in the EMT is unclear. We investigated the effect of 20(S)-Rg3 and 20(R)-Rg3 on the EMT. Transforming growth factor-beta 1 (TGF-ß1) induces the EMT to promote lung adenocarcinoma migration, invasion, and anoikis resistance. To understand the repressive role of 20(S)-Rg3 and 20(R)-Rg3 in lung cancer migration, invasion, and anoikis resistance, we investigated the potential use of 20(S)-Rg3 and 20(R)-Rg3 as inhibitors of TGF-ß1-induced EMT development in A549 lung cancer cells in vitro. Here, we show that 20(R)-Rg3, but not 20(S)-Rg3, markedly increased expression of the epithelial marker E-cadherin and repressed Snail upregulation and expression of the mesenchymal marker vimentin during initiation of the TGF-ß1-induced EMT. 20(R)-Rg3 also inhibited the TGF-ß1-induced increase in cell migration, invasion, and anoikis resistance of A549 lung cancer cells. Additionally, 20(R)-Rg3 markedly inhibited TGF-ß1-regulated matrix metalloproteinase-2 and activation of Smad2 and p38 mitogen activated protein kinase. Taken together, our findings provide new evidence that 20(R)-Rg3 suppresses lung cancer migration, invasion, and anoikis resistance in vitro by inhibiting the TGF-ß1-induced EMT.


Asunto(s)
Anoicis/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Ginsenósidos/farmacología , Neoplasias Pulmonares/patología , Invasividad Neoplásica/prevención & control , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Western Blotting , Cadherinas/biosíntesis , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Ginsenósidos/química , Humanos , Neoplasias Pulmonares/inducido químicamente , Metaloproteinasa 2 de la Matriz/metabolismo , ARN/biosíntesis , ARN/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína Smad2/metabolismo , Estereoisomerismo , Factor de Crecimiento Transformador beta1/farmacología , Vimentina/biosíntesis , Cicatrización de Heridas/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
J Biol Chem ; 289(27): 18625-40, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24821727

RESUMEN

Promyelocytic leukemia zinc finger (PLZF) is a transcription repressor that was initially isolated as a fusion protein with retinoic acid receptor α. PLZF is aberrantly overexpressed in various human solid tumors, such as clear cell renal carcinoma, glioblastoma, and seminoma. PLZF causes cellular transformation of NIH3T3 cells and increases cell proliferation in several cell types. PLZF also increases tumor growth in the mouse xenograft tumor model. PLZF may stimulate cell proliferation by controlling expression of the genes of the p53 pathway (ARF, TP53, and CDKN1A). We found that PLZF can directly repress transcription of CDKN1A encoding p21, a negative regulator of cell cycle progression. PLZF binds to the proximal Sp1-binding GC-box 5/6 and the distal p53-responsive elements of the CDKN1A promoter to repress transcription. Interestingly, PLZF interacts with Sp1 or p53 and competes with Sp1 or p53. PLZF interacts with corepressors, such as mSin3A, NCoR, and SMRT, thereby deacetylates Ac-H3 and Ac-H4 histones at the CDKN1A promoter, which indicated the involvement of the corepressor·HDACs complex in transcription repression by PLZF. Also, PLZF represses transcription of TP53 and also decreases p53 protein stability by ubiquitination. PLZF may act as a potential proto-oncoprotein in various cell types.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Represoras/metabolismo , Acetilación , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Regiones Promotoras Genéticas/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Factor de Transcripción Sp1/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo
15.
J Biol Chem ; 289(27): 18641-56, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24821728

RESUMEN

Promyelocytic leukemia zinc finger-retinoic acid receptor α (PLZF-RARα) is an oncogene transcriptional repressor that is generated by a chromosomal translocation between the PLZF and RARα genes in acute promyelocytic leukemia (APL-type) patients. The molecular interaction between PLZF-RARα and the histone deacetylase corepressor was proposed to be important in leukemogenesis. We found that PLZF-RARα can repress transcription of the p21WAF/CDKN1A gene, which encodes the negative cell cycle regulator p21 by binding to its proximal promoter Sp1-binding GC-boxes 3, 4, 5/6, a retinoic acid response element (RARE), and distal p53-responsive elements (p53REs). PLZF-RARα also acts as a competitive transcriptional repressor of p53, RARα, and Sp1. PLZF-RARα interacts with co-repressors such as mSin3A, NCoR, and SMRT, thereby deacetylating histones Ac-H3 and Ac-H4 at the CDKN1A promoter. PLZF-RARα also interacts with the MBD3-NuRD complex, leading to epigenetic silencing of CDKN1A through DNA methylation. Furthermore, PLZF-RARα represses TP53 and increases p53 protein degradation by ubiquitination, further repressing p21 expression. Resultantly, PLZF-RARα promotes cell proliferation and significantly increases the number of cells in S-phase.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética/genética , Proteína p53 Supresora de Tumor/genética , Unión Competitiva , Línea Celular Tumoral , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/deficiencia , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Silenciador del Gen , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Regiones Promotoras Genéticas/genética , Estabilidad Proteica , Fase S , Factor de Transcripción Sp1/metabolismo
16.
Life Sci ; 95(2): 101-7, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24231449

RESUMEN

AIMS: Neobavaisoflavone (NBIF), an isoflavone isolated from Psoralea corylifolia (Leguminosae), has striking anti-inflammatory and anti-cancer effects. NBIF inhibits the proliferation of prostate cancer in vitro and in vivo. MAIN METHODS: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a key endogenous molecule that selectively induces apoptosis in cancer cells with little or no toxicity in normal cells. However, some cancer cells, including U373MG cells, are resistant to TRAIL-mediated apoptosis. We demonstrated that the cell viability, migration and invasion assay were used in U373MG glioma cells. KEY FINDINGS: In this study, we found that NBIF sensitizes human U373MG glioma cells to TRAIL-mediated apoptosis. Co-treatment of TRAIL and NBIF effectively induced Bid cleavage and activated caspases 3, 8, and 9. Importantly, DR5 expression was upregulated by NBIF. We also observed that the combination NBIF and TRAIL increased expression of BAX. We further demonstrate that NBIF induced TRAIL-mediated apoptosis in human glioma cells by suppressing migration and invasion, and by inhibiting anoikis resistance. SIGNIFICANCE: Taken together, our results suggest that NBIF reduces the resistance of cancer cells to TRAIL and that the combination of NBIF and TRAIL may be a new therapeutic strategy for treating TRAIL-resistant glioma cells.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Glioma/patología , Isoflavonas/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Western Blotting , Caspasas/genética , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Glioma/tratamiento farmacológico , Humanos , Metástasis de la Neoplasia , Regulación hacia Arriba/efectos de los fármacos
17.
Mol Cancer Res ; 10(8): 1032-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22767590

RESUMEN

Previously, we reported that high PKCK2 activity could protect cancer cells from death receptor-mediated apoptosis through phosphorylation of procaspase-2. Because anoikis is another form of apoptosis, we asked whether PKCK2 could similarly confer resistance to anoikis on cancer cells. Human esophageal squamous cancer cell lines with high PKCK2 activity (HCE4 and HCE7) were anoikis-resistant, whereas cell lines with low PKCK2 activity (TE2 and TE3) were anoikis-sensitive. Because the cells showed different sensitivity to anoikis, we compared the expression of cell adhesion molecules between anoikis-sensitive TE2 and anoikis-resistant HCE4 cells using cDNA microarray. We found that E-cadherin is expressed only in TE2 cells; whereas N-cadherin is expressed instead of E-cadherin in HCE4 cells. To examine whether PKCK2 activity could determine the type of cadherin expressed, we first increased intracellular PKCK2 activity in TE2 cells by overexpressing the PKCK2α catalytic subunit using lentivirus and found that high PKCK2 activity could switch cadherin expression from type E to N and confer anoikis resistance. Conversely, a decrease in PKCK2 activity in HCE4 cells by knockdown of PKCK2α catalytic subunit using shRNA induced N- to E-cadherin switching and the anoikis-resistant cells became sensitive. In addition, N-cadherin expression correlated with PKB/Akt activation and increased invasiveness. We conclude that high intracellular PKCK2 activity confers anoikis resistance on esophageal cancer cells by inducing E- to N-cadherin switching.


Asunto(s)
Anoicis , Antígenos CD , Cadherinas , Carcinoma , Neoplasias Esofágicas , Anoicis/genética , Anoicis/fisiología , Antígenos CD/genética , Cadherinas/genética , Cadherinas/metabolismo , Carcinoma/tratamiento farmacológico , Carcinoma/metabolismo , Carcinoma/patología , Quinasa de la Caseína II/genética , Quinasa de la Caseína II/metabolismo , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Invasividad Neoplásica , Fosforilación , Transducción de Señal
18.
Mol Biol Cell ; 23(15): 2943-54, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22675025

RESUMEN

Aberrant expression of casein kinase 2 (CK2) is associated with tumor progression; however, the molecular mechanism by which CK2 modulates tumorigenesis is incompletely understood. In this paper, we show that CK2α phosphorylates the C-terminal domain of the nuclear receptor corepressor (NCoR) at Ser-2436 to stabilize the NCoR against the ubiquitin-dependent proteasomal degradation pathway. Importantly, NCoR promoted the invasion of esophageal cancer cells in a CK2-dependent manner. By using cyclic DNA microarray analysis, we identified CXCL10/IP-10 as a novel CK2α-NCoR cascade-regulated gene. The depletion of both NCoR and HDAC3 commonly derepressed IP-10 transcription, demonstrating the functional engagement of the NCoR-HDAC3 axis in IP-10 transcriptional repression. Furthermore, chromatin immunoprecipitation assays showed that c-Jun recruits NCoR-HDAC3 corepressor complexes to the (AP1 site of IP-10, leading to histone hypoacetylation and IP-10 down-regulation. Collectively these data suggest that the CK2α-NCoR cascade selectively represses the transcription of IP-10 and promotes oncogenic signaling in human esophageal cancer cells.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Quimiocina CXCL10/metabolismo , Neoplasias Esofágicas , Invasividad Neoplásica/genética , Co-Represor 1 de Receptor Nuclear , Sitios de Unión , Quinasa de la Caseína II/genética , Línea Celular Tumoral , Transformación Celular Neoplásica , Quimiocina CXCL10/genética , Proteínas Co-Represoras , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Histona Desacetilasas/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteolisis , Transducción de Señal , Transcripción Genética
19.
Sci Signal ; 4(197): ra71, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22045851

RESUMEN

Although loss of p53 function and activation of canonical Wnt signaling cascades are frequently coupled in cancer, the links between these two pathways remain unclear. We report that p53 transactivated microRNA-34 (miR-34), which consequently suppressed the transcriptional activity of ß-catenin-T cell factor and lymphoid enhancer factor (TCF/LEF) complexes by targeting the untranslated regions (UTRs) of a set of conserved targets in a network of genes encoding elements of the Wnt pathway. Loss of p53 function increased canonical Wnt signaling by alleviating miR-34-specific interactions with target UTRs, and miR-34 depletion relieved p53-mediated Wnt repression. Gene expression signatures reflecting the status of ß-catenin-TCF/LEF transcriptional activity in breast cancer and pediatric neuroblastoma patients were correlated with p53 and miR-34 functional status. Loss of p53 or miR-34 contributed to neoplastic progression by triggering the Wnt-dependent, tissue-invasive activity of colorectal cancer cells. Further, during development, miR-34 interactions with the ß-catenin UTR affected Xenopus body axis polarity and the expression of Wnt-dependent patterning genes. These data provide insight into the mechanisms by which a p53-miR-34 network restrains canonical Wnt signaling cascades in developing organisms and human cancer.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , Proteína p53 Supresora de Tumor/genética , Vía de Señalización Wnt/genética , Regiones no Traducidas 3'/genética , Animales , Secuencia de Bases , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Células Cultivadas , Embrión de Pollo , Niño , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Ratones , MicroARNs/metabolismo , Mutación , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Interferencia de ARN , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Xenopus laevis , beta Catenina/genética , beta Catenina/metabolismo
20.
PLoS One ; 6(4): e19163, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21559479

RESUMEN

Despite the numerous studies of protein kinase CK2, little progress has been made in understanding its function in chondrocyte death. Our previous study first demonstrated that CK2 is involved in apoptosis of rat articular chondrocytes. Recent studies have suggested that CK2 downregulation is associated with aging. Thus examining the involvement of CK2 downregulation in chondrocyte death is an urgently required task. We undertook this study to examine whether CK2 downregulation modulates chondrocyte death. We first measured CK2 activity in articular chondrocytes of 6-, 21- and 30-month-old rats. Noticeably, CK2 activity was downregulated in chondrocytes with advancing age. To build an in vitro experimental system for simulating tumor necrosis factor (TNF)-α-induced cell death in aged chondrocytes with decreased CK2 activity, chondrocytes were co-treated with CK2 inhibitors and TNF-α. Viability assay demonstrated that CK2 inhibitors facilitated TNF-α-mediated chondrocyte death. Pulsed-field gel electrophoresis, nuclear staining, flow cytometry, TUNEL staining, confocal microscopy, western blot and transmission electron microscopy were conducted to assess cell death modes. The results of multiple assays showed that this cell death was mediated by apoptosis. Importantly, autophagy was also involved in this process, as supported by the appearance of a punctuate LC3 pattern and autophagic vacuoles. The inhibition of autophagy by silencing of autophage-related genes 5 and 7 as well as by 3-methyladenine treatment protected chondrocytes against cell death and caspase activation, indicating that autophagy led to the induction of apoptosis. Autophagic cells were observed in cartilage obtained from osteoarthritis (OA) model rats and human OA patients. Our findings indicate that CK2 down regulation facilitates TNF-α-mediated chondrocyte death through apoptosis and autophagy. It should be clarified in the future if autophagy observed is a consequence versus a cause of the degeneration in vivo.


Asunto(s)
Quinasa de la Caseína II/biosíntesis , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Osteoartritis/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Autofagia , Condrocitos/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...