Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Small ; 15(20): e1805086, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30925031

RESUMEN

Emerging advances in iron oxide nanoparticles exploit their high magnetization for various applications, such as bioseparation, hyperthermia, and magnetic resonance imaging. In contrast to their excellent magnetic performance, the harmonic generation and luminescence properties of iron oxide nanoparticles have not been thoroughly explored, thus limiting their development as a tool in photomedicine. In this work, a seed/growth-inspired synthesis is developed combined with primary mineralization and a ligand-assisted secondary growth strategy to prepare mesostructured α-FeOOH nanorods (NRs). The sub-wavelength heterogeneity of the refractive index leads to enhanced third-harmonic generation (THG) signals under near-infrared excited wavelengths at 1230 nm. The as-prepared NRs exhibit an 11-fold stronger THG intensity compared to bare α-FeOOH NRs. Using these unique nonlinear optical properties, it is demonstrated that mesostructured α-FeOOH NRs can serve as biocompatible and nonbleaching contrast agents in THG microscopy for long-term labeling of cells as well as in angiography in vivo by modifying lectin to enhance the binding efficiency to the glycocalyx layers on the wall of blood vessels. These results provide a new insight into Fe-based nanoplatforms capable of emitting coherent light as molecular probes in optical microscopy, thus establishing a complementary microscopic imaging method for macroscopic magnetic imaging systems.


Asunto(s)
Imagenología Tridimensional , Compuestos de Hierro/química , Minerales/química , Nanotubos/química , Células A549 , Animales , Supervivencia Celular , Oído/anatomía & histología , Humanos , Ratones Endogámicos BALB C , Nanotubos/ultraestructura , Dinámicas no Lineales
2.
Toxicol Appl Pharmacol ; 290: 98-106, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26589485

RESUMEN

Gemcitabine and cisplatin (GC) has been widely used for advanced and metastatic urothelial carcinoma (UC). However, resistance to this remedy has been noticed. We have demonstrated that increase of TG-interacting factor (TGIF) in specimens is associated with worse prognosis of upper tract UC (UTUC) patients. The roles of TGIF in the gemcitabine resistance of UC were explored. Specimens of 23 locally advanced/advanced stage UTUC patients who received GC systemic chemotherapy after radical nephroureterectomy were collected to evaluate the alterations of TGIF in the resistance to the remedy by using immunohistochemistry. In vitro characterizations of mechanisms mediating TGIF in gemcitabine resistance were conducted by analyzing NTUB1 cells and their gemcitabine-resistant subline, NGR cells. Our results show that increased TGIF is significantly associated with chemo-resistance, poor progression-free survival, and higher cancer-related deaths of UTUC patients. Higher increases of TGIF, p-AKT(Ser473) and invasive ability were demonstrated in NGR cells. Overexpression of TGIF in NTUB1 cells upregulated p-AKT(Ser473) activation, enhanced migration ability, and attenuated cellular sensitivity to gemcitabine. Knockdown of TGIF in NGR cells downregulated p-AKT(Ser473) activation, declined migration ability, and enhanced cellular sensitivity to gemcitabine. In addition, histone deacetylases inhibitor trichostatin A (TSA) inhibited TGIF, p-AKT(Ser473) expression and migration ability. Synergistic effects of gemcitabine and TSA on NGR cells were also demonstrated. Collectively, TGIF contributes to the gemcitabine resistance of UC via AKT activation. Combined treatment with gemcitabine and TSA might be a promising therapeutic remedy to improve the gemcitabine resistance of UC.


Asunto(s)
Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Anciano , Desoxicitidina/farmacología , Regulación hacia Abajo , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/patología , Gemcitabina
3.
Toxicol Appl Pharmacol ; 285(1): 41-50, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25791921

RESUMEN

Arsenic trioxide (ATO) is a multi-target drug approved by the Food and Drug Administration as the first-line chemotherapeutic agent for the treatment of acute promyelocytic leukemia. In addition, several clinical trials are being conducted with arsenic-based drugs for the treatment of other hematological malignancies and solid tumors. However, ATO's modest clinical efficacy on some cancers, and potential toxic effects on humans have been reported. Determining how best to reduce these adverse effects while increasing its therapeutic efficacy is obviously a critical issue. Previously, we demonstrated that the JNK-induced complex formation of phosphorylated c-Jun and TG-interacting factor (TGIF) antagonizes ERK-induced cyclin-dependent kinase inhibitor CDKN1A (p21(WAF1/CIP1)) expression and resultant apoptosis in response to ATO in A431 cells. Surprisingly, at low-concentrations (0.1-0.2 µM), ATO increased cellular proliferation, migration and invasion, involving TGIF expression, however, at high-concentrations (5-20 µM), ATO induced cell apoptosis. Using a promoter analysis, TGIF was transcriptionally regulated by ATO at the FOXO3A binding site (-1486 to -1479bp) via the c-Src/EGFR/AKT pathway. Stable overexpression of TGIF promoted advancing the cell cycle into the S phase, and attenuated 20 µM ATO-induced apoptosis. Furthermore, blockage of the AKT pathway enhanced ATO-induced CDKN1A expression and resultant apoptosis in cancer cells, but overexpression of AKT1 inhibited CDKN1A expression. Therefore, we suggest that TGIF is transcriptionally regulated by the c-Src/EGFR/AKT pathway, which plays a role as a negative regulator in antagonizing ATO-induced CDKN1A expression and resultant apoptosis. Suppression of these antagonistic effects might be a promising therapeutic strategy toward improving clinical efficacy of ATO.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Factores de Transcripción Forkhead/metabolismo , Proteínas de Homeodominio/metabolismo , Neoplasias/enzimología , Neoplasias/patología , Óxidos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética/efectos de los fármacos , Antineoplásicos/toxicidad , Trióxido de Arsénico , Sitios de Unión , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Proteínas de Homeodominio/genética , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Células MCF-7 , Invasividad Neoplásica , Neoplasias/genética , Óxidos/toxicidad , Fosforilación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Represoras/genética , Transducción de Señal/efectos de los fármacos , Transfección
4.
Arch Toxicol ; 89(12): 2229-41, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25537191

RESUMEN

Inorganic arsenic is well known as a carcinogen in human beings. Chronic exposure to inorganic arsenic increases risks of developing some cancers and non-carcinogenic diseases, such as skin lesions in humans. However, the modes of action are not well elucidated. In the present study, HaCaT cells, an immortalized non-tumorigenic human keratinocyte, were continuously exposed to low-dose trivalent arsenic (arsenic trioxide, 0.1 and 0.2 µM) for at least 4 weeks. We proved that low-dose arsenic could stimulate malignant transformation of HaCaT cells, including increase of cellular proliferation, epithelial-to-mesenchymal transition markers alteration, matrix metalloproteinases activation, invadopodia formation, migration/invasion activities, and anchorage-independent growth. Surprisingly, low-dose arsenic could also transcriptionally increase TG-interacting factor (TGIF) expression via c-Src/EGFR/AKT/FOXO3A signaling involving superoxide production from NADPH oxidase. Moreover, stable overexpression of TGIF could also induce malignant transformation of HaCaT cells. Knockdown of TGIF with its specific shRNA abolished the arsenic-induced effects. Taken together, we suggest that TGIF plays an important role in low-dose arsenic-induced malignant transformation of HaCaT cells, which is regulated by c-Src/EGFR/AKT/FOXO3A pathway and redox signaling.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Queratinocitos/efectos de los fármacos , Óxidos/toxicidad , Proteínas Represoras/metabolismo , Animales , Trióxido de Arsénico , Arsenicales/administración & dosificación , Proteína Tirosina Quinasa CSK , Línea Celular , Relación Dosis-Respuesta a Droga , Receptores ErbB/metabolismo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Humanos , Queratinocitos/patología , Ratones , Células 3T3 NIH , Oxidación-Reducción/efectos de los fármacos , Óxidos/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/genética , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/metabolismo
5.
J Neurochem ; 131(6): 816-24, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25319900

RESUMEN

Traumatic brain injury (TBI) is a complex injury involving several physiological alterations, potentially leading to neurological impairment. Previous mouse studies using high-density oligonucleotide array analysis have confirmed the upregulation of transforming growth-interacting factor (TGIF) mRNA in TBI. TGIF is a transcriptional corepressor of transforming growth factor beta (TGF-ß) signaling which plays a protective role in TBI. However, the functional roles of TGIF in TBI are not well understood. In this study, we used confocal microscopy after immunofluorescence staining to demonstrate the increase of TGIF levels in the activated microglia of the pericontusional cortex of rats with TBI. Intracerebral knockdown of TGIF in the pericontusional cortex significantly downregulated TGIF expression, attenuated microglial activation, reduced the volume of damaged brain tissue, and facilitated recovery of limb motor function. Collectively, our results indicate that TGIF is involved in TBI-induced microglial activation, resulting in secondary brain injury and motor dysfunction. This study investigated the roles of transforming growth-interacting factor (TGIF) in a traumatic brain injury (TBI)-rat model. We demonstrated the increase of TGIF levels in the activated microglia of the pericontusional cortex of rats with TBI. Intracerebral knockdown of TGIF in the pericontusional cortex of the TBI rats significantly attenuated micoglial activation, reduced the volume of damaged brain tissue, and facilitated recovery of limb motor function. We suggest that inhibition of TGIF might provide a promising therapeutic strategy for TBI.


Asunto(s)
Lesiones Encefálicas/metabolismo , Corteza Cerebral/metabolismo , Microglía/metabolismo , Activación Transcripcional/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Lesiones Encefálicas/tratamiento farmacológico , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen/métodos , Masculino , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/genética
6.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1869(5): 159492, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38575107

RESUMEN

Obesity is one of the significant health challenges in the world and is highly associated with abnormal adipogenesis. TG-interacting factor 1 (TGIF1) is essential for differentiating murine adipocytes and human adipose tissue-derived stem cells. However, the mode of action needs to be better elucidated. To investigate the roles of TGIF1 in differentiation in-depth, CRISPR/Cas9 knockout technology was performed to generate TGIF1-silenced preadipocytes. The absence of TGIF1 in 3 T3-F442A preadipocytes abolished lipid accumulation throughout the differentiation using Oil Red O staining. Conversely, we established 3 T3-F442A preadipocytes stably expressing TGIF1 and doxycycline-inducible TGIF1 in TGIF1-silenced 3 T3-F442A preadipocytes. Remarkably, the induction of TGIF1 by doxycycline during the initial differentiation phase successfully promoted lipid accumulation in TGIF1-silenced 3 T3-F442A cells. We further explored the mechanisms of TGIF1 in early differentiation. We demonstrated that TGIF1 promoted the mitotic clonal expansion via upregulation of CCAAT/enhancer-binding proteins ß expression, interruption with peroxisome proliferators activated receptor γ downstream regulation, and inhibition of p27kip1 expression. In conclusion, we strengthen the pivotal roles of TGIF1 in early differentiation, which might contribute to resolving obesity-associated metabolic syndromes.


Asunto(s)
Adipocitos , Adipogénesis , Diferenciación Celular , Proteínas de Homeodominio , Proteínas Represoras , Animales , Humanos , Ratones , Adipocitos/metabolismo , Adipocitos/citología , Adipogénesis/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Mitosis/genética , PPAR gamma/metabolismo , PPAR gamma/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
7.
FEBS Lett ; 598(8): 945-955, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38472156

RESUMEN

TG-interacting factor 1 (TGIF1) contributes to the differentiation of murine white preadipocyte and human adipose tissue-derived stem cells; however, its regulation is not well elucidated. Insulin is a component of the adipogenic cocktail that induces ERK signaling. TGIF1 phosphorylation and sustained stability in response to insulin were reduced through the use of specific MEK inhibitor U0126. Mutagenesis at T235 or T239 residue of TGIF1 in preadipocytes led to dephosphorylation of TGIF1. The reduced TGIF1 stability resulted in an increase in p27kip1 expression, a decrease in phosphorylated Rb expression and cellular proliferation, and a reduced accumulation of lipids compared to the TGIF1-overexpressed cells. These findings highlight that insulin/ERK-driven phosphorylation of the T235 or T239 residue at TGIF1 is crucial for adipocyte differentiation.


Asunto(s)
Células 3T3-L1 , Adipocitos , Adipogénesis , Diferenciación Celular , Proteínas de Homeodominio , Insulina , Animales , Ratones , Fosforilación/efectos de los fármacos , Insulina/metabolismo , Adipocitos/metabolismo , Adipocitos/citología , Adipocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Humanos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Proliferación Celular/efectos de los fármacos , Butadienos/farmacología
8.
Am J Pathol ; 181(3): 1044-55, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22771156

RESUMEN

Prognostic outcome prediction would be useful for the treatment of patients with upper urinary tract urothelial carcinoma (UC). However, its prognostic biomarkers are not well established so far. According to the results of analysis of 168 human upper urinary tract UC specimens, overexpressed TG-interacting factor (TGIF) in nuclei of tumor tissues is significantly correlated with poor progression-free survival and higher cancer-related death. When both TGIF and p21 expression are altered, these patients had an even worse prognosis than those with one or no marker altered. Furthermore, to elucidate the role of TGIF in the progression of UC, overexpression of TGIF in RT4 or TSGH8301 cells was performed, and the results revealed that TGIF can significantly increase migration/invasion ability, matrix metalloproteinase expression, and invadopodia formation via the phosphatidylinositol 3-kinase-AKT pathway. In contrast, knockdown of TGIF with its specific short hairpin RNA inhibited the invasion ability of T24 cells. Besides, TGIF could inhibit p21(WAF/CIP1) expression, up-regulate cyclin D1 expression, and phosphorylate retinoblastoma to promote G1-S transition and cellular proliferation. In conclusion, we demonstrated that TGIF contributes to the progression of urothelial carcinoma via the phosphatidylinositol 3-kinase-AKT pathway. It may serve as an attractive therapeutic or prognostic target for selected patients with upper urinary tract UC.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Proteínas Represoras/metabolismo , Neoplasias Urológicas/metabolismo , Neoplasias Urológicas/patología , Urotelio/metabolismo , Urotelio/patología , Anciano , Movimiento Celular , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Supervivencia sin Enfermedad , Femenino , Fase G1 , Humanos , Masculino , Análisis Multivariante , Invasividad Neoplásica , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Seudópodos/metabolismo , Fase S , Neoplasias Urológicas/enzimología , Urotelio/enzimología
9.
PLoS One ; 18(3): e0281873, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36862682

RESUMEN

Urine-based cytology is non-invasive and widely used for clinical diagnosis of urothelial carcinoma (UC), but its sensitivity is less than 40% for low-grade UC detection. As such, there is a need for new diagnostic and prognostic biomarkers of UC. CUB domain containing protein 1 (CDCP1) is a type I transmembrane glycoprotein highly expressed in various cancers. Using tissue array analysis, we demonstrated that CDCP1 expression in UC patients (n = 133), especially in those with low-grade UC, was significantly higher than in 16 normal persons. In addition, CDCP1 expression in urinary UC cells could also be detected by using immunocytochemistry method (n = 11). Furthermore, in 5637-CD cells, overexpression of CDCP1 affected the expression of epithelial mesenchymal transition-related markers and increased matrix metalloproteinase 2 expression and migration ability. Conversely, the knockdown of CDCP1 in T24 cells had the opposite effects. Using specific inhibitors, we demonstrated the involvement of c-Src/PKCδ signaling in the CDCP1-regulated migration of UC. In conclusion, our data suggest that CDCP1 contributes to the malignant progression of UC and may have the potential as a urine-based biomarker for detecting low-grade UC. However, a cohort study needs to be conducted.


Asunto(s)
Líquidos Corporales , Carcinoma de Células Transicionales , Neoplasias de la Vejiga Urinaria , Humanos , Neoplasias de la Vejiga Urinaria/diagnóstico , Metaloproteinasa 2 de la Matriz , Biomarcadores , Antígenos de Neoplasias , Moléculas de Adhesión Celular/genética
10.
J Biol Chem ; 286(9): 7043-51, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21189260

RESUMEN

The innate immune system elicits the first wave of immune responses against pathogen infection. Its operational modes are complex and have yet to be defined. Here, we report the identification of an innate immune regulator termed TAPE (TBK1-associated protein in endolysosomes), previously known as CC2D1A/Freud-1/Aki-1, which modulates the TLR3 and TLR4 pathways. We found that TAPE activated the TBK1, NF-κB, and ERK pathways leading to IFN-ß and inflammatory cytokine induction. TAPE was shown to colocalize with endosomal marker Rab5 and lysosomal marker LAMP1 in mammalian cells, suggesting that TAPE resided in endolysosomes. Knockdown of TAPE selectively impaired the TLR3 and endocytic TLR4 pathways to IFN-ß induction. Furthermore, TAPE interacted and synergized with Trif to activate IFN-ß. TAPE knockdown failed to block Trif-mediated IFN-ß induction, whereas Trif knockdown impaired the TLR3 and TAPE cooperation on IFN-ß induction, suggesting that TAPE acts upstream of Trif. Together, our data demonstrate a central role for TAPE in linking TLR3 and TLR4 to innate immune defenses at an early step.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factores Inmunológicos/metabolismo , Transducción de Señal/inmunología , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Proteínas de Unión al ADN/inmunología , Endosomas/inmunología , Endosomas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Humanos , Factores Inmunológicos/inmunología , Interferón beta/inmunología , Interferón beta/metabolismo , Lisosomas/inmunología , Lisosomas/metabolismo , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 4/inmunología
11.
Arch Toxicol ; 86(6): 935-45, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22532027

RESUMEN

Arsenic is a well-known poison and carcinogen in humans. However, it also has been used to effectively treat some human cancers and non-carcinogenic ailments. Previously, we demonstrated in keratinocytes that arsenic trioxide (ATO)-induced p21(WAF1/CIP1) (p21) expression leading to cellular cytotoxicity through the c-Src/EGFR/ERK pathway and generation of reactive oxygen species (ROS). In this study, we found that EGFR-Y845 and EGFR-Y1173 could be phosphorylated by ATO. Using confocal microscopy and flow cytometry, we found that pretreatment with apocynin, DPI, and tiron could remove ATO-induced ROS production. Furthermore, to increase NADPH oxidase activity, ATO could induce cytosolic p67(phox) expression and translocation to membrane. In addition, knockdown of p67(phox) could abolish ATO-induced ROS production. Therefore, we suggest that NADPH oxidase-produced superoxide was a major source of ATO-induced ROS production. Conversely, ATO-induced NADPH oxidase activation and superoxide generation could be inhibited by the c-Src inhibitor PP1, but not by the EGFR inhibitor PD153035. In addition, overexpression of c-Src as well as treatment with ATO could stimulate EGFR-Y845/ERK phosphorylation, p21 expression, and cellular arrest/apoptosis, which could be attenuated by pretreatment with apocynin or knockdown of p67(phox). Collectively, we suggest that NADPH oxidase was involved in the ATO-induced arrest/apoptosis of keratinocytes, which was regulated by c-Src activation.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Queratinocitos/metabolismo , NADPH Oxidasas/metabolismo , Óxidos/toxicidad , Superóxidos/metabolismo , Familia-src Quinasas/metabolismo , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Arsenicales , Línea Celular , Inhibidores Enzimáticos/metabolismo , Humanos , Fosforilación/efectos de los fármacos , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo
12.
Biochem J ; 438(2): 349-58, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21649584

RESUMEN

HCC (hepatocellular carcinoma) is among the most common and lethal cancers worldwide with a poor prognosis mainly due to a high recurrence rate and chemotherapy resistance. ATO (arsenic trioxide) is a multi-target drug that has been effectively used as an anticancer drug in acute promyelocytic leukaemia. However, a Phase II trial involving patients with HCC indicates that the use of arsenic as a single agent is not effective against HCC. TGIF (TG-interacting factor) is a transcriptional co-repressor that interferes with TGF-ß (transforming growth factor-ß) signalling which plays a growth-inhibitory role in HCC. In the present study, we demonstrated that ATO induced hepatocellular apoptosis via TGF-ß/Smad signalling and led to downstream induction of p21(WAF1/CIP1) (p21). However, ATO could also induce TGIF expression via a post-transcriptional regulation mechanism to antagonize this effect. Using a biotin-labelled RNA probe pull-down assay and in vivo RNA immunoprecipitation analysis, we identified that HuR (human antigen R) bound to the TGIF mRNA 3'-UTR (3'-untranslated region) and prevented it from degradation. ATO treatment increased the interaction between HuR and TGIF mRNA, and reduction of HuR expression inhibited ATO-induced TGIF expression. Moreover, the EGFR (epidermal growth factor receptor)/PI3K (phosphoinositide 3-kinase)/Akt pathway was shown to mediate the post-transcriptional regulation of TGIF in response to ATO. Finally, we also demonstrated that the down-regulation of TGIF could sensitize ATO-induced HepG2 cell apoptosis. Collectively, we propose that the EGFR/PI3K/Akt pathway may regulate the post-transcriptional regulation of TGIF expression to antagonize ATO-induced apoptosis in HCC. Blockage of the PI3K/Akt pathway or TGIF expression combined with ATO treatment may be a promising strategy for HCC therapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Carcinoma Hepatocelular/patología , Proteínas de Homeodominio/metabolismo , Neoplasias Hepáticas/patología , Óxidos/farmacología , Proteínas Represoras/metabolismo , Regiones no Traducidas 3'/genética , Antígenos de Superficie/metabolismo , Trióxido de Arsénico , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas ELAV , Proteína 1 Similar a ELAV , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/genética , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
13.
PLoS One ; 16(7): e0254011, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34237099

RESUMEN

Patients with urothelial carcinoma (UC) experience gemcitabine resistance is a critical issue. The role of hedgehog pathway in the problem was explored. The expressions of phospho-AKTser473, phospho-GSK3ßser9 and Gli2 were up-regulated in gemcitabine-resistant NTUB1 (NGR) cells. Without hedgehog ligands, Gli proteins can be phosphorylated by GSK3ß kinase to inhibit their downstream regulations. Furthermore, the GSK3ß kinase can be phosphorylated by AKT at its Ser9 residue to become an inactive kinase. Therefore, overexpression of AKT1, Flag-GSKS9D (constitutively inactive form) or active Gli2 (GLI2ΔN) in NTUB1 cells could activate Gli2 pathway to enhance migration/invasion ability and increase gemcitabine resistance, respectively. Conversely, overexpression of Flag-GSKS9A (constitutively active form) or knockdown of Gli2 could suppress Gli2 pathway, and then reduce gemcitabine resistance in NGR cells. Therefore, we suggest gemcitabine-activated AKT/GSK3ß pathway can elicit Gli2 activity, which leads to enhanced migration/invasion ability and resistance to gemcitabine therapy in UC patients. The non-canonical hedgehog pathway should be evaluated in the therapy to benefit UC patients.


Asunto(s)
Desoxicitidina/análogos & derivados , Proteínas Hedgehog/metabolismo , Transducción de Señal , Proteína Gli2 con Dedos de Zinc/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Desoxicitidina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Modelos Biológicos , Invasividad Neoplásica , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Urotelio/metabolismo , Urotelio/patología , Gemcitabina
14.
Toxicol Appl Pharmacol ; 244(2): 234-41, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20074581

RESUMEN

Arsenic is well known as a carcinogen predisposing humans to some severe diseases and also as an effective medicine for treating acute promyelocytic leukemia, syphilis, and psoriasis. Multiple active mechanisms, including cell cycle arrest and apoptosis, have been proposed in therapy; however, the opposing effects of arsenic remain controversial. Our previous study found that arsenic trioxide (ATO)-induced activation of p21(WAF1/CIP1) (p21) led to A431 cell death through the antagonistic effects of the signaling of ERK1/2 and JNK1. In the current study, the inhibitory effects of JNK1 on ATO-induced p21 expression were explored. Over-expression of JNK1 in A431 cells could inhibit p21 expression, which was associated with HDAC1 and TGIF. Using the GST pull-down assay and fluorescence resonance energy transfer analysis, N-terminal domain (amino acids 1-108) of TGIF, critical to its binding with c-Jun, was found. Using reporter assays, requirement of the C-terminal domain (amino acids 138-272) of TGIF to suppress ATO-induced p21 expression was observed. Thus, the domains of TGIF that carried out its inhibitory effects on p21 were identified. Finally, treatment with JNK inhibitor SP600125 could enhance ATO-induced apoptosis of HaCaT keratinocytes by using flow cytometry.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Queratinocitos/efectos de los fármacos , Queratinocitos/enzimología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Óxidos/toxicidad , Antracenos/farmacología , Apoptosis/fisiología , Trióxido de Arsénico , Arsenicales , Línea Celular Tumoral , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Queratinocitos/patología , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/fisiología
15.
Toxicol Appl Pharmacol ; 233(2): 297-307, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18822310

RESUMEN

An infamous poison, arsenic also has been used as a drug for nearly 2400 years; in recently years, arsenic has been effective in the treatment of acute promyelocytic leukemia. Increasing evidence suggests that opposite effects of arsenic trioxide (ATO) on tumors depend on its concentrations. For this reason, the mechanisms of action of the drug should be elucidated, and it should be used therapeutically only with extreme caution. Previously, we demonstrated the opposing effects of ERK1/2 and JNK on p21(WAF1/CIP1) (p21) expression in response to ATO in A431 cells. In addition, JNK phosphorylates c-Jun (Ser(63/73)) to recruit TGIF/HDAC1 to suppress p21 gene expression. Presently, we demonstrated that a high concentration of ATO sustains ERK1/2 phosphorylation, and increases c-Fos biosynthesis and stability, which enhances p21 gene expression. Using site-directed mutagenesis, a DNA affinity precipitation assay, and functional assays, we demonstrated that phosphorylation of the C-terminus of c-Fos (Thr(232), Thr(325), Thr(331), and Ser(374)) plays an important role in its binding to the p21 promoter, and in conjunction with N-terminus phosphorylation of c-Fos (Ser(70)) to transactivate p21 promoter expression. In conclusion, a high concentration of ATO can sustain ERK1/2 activation to enhance c-Fos expression, then dimerize with dephosphorylated c-Jun (Ser(63/73)) and recruit p300/CBP to the Sp1 sites (-84/-64) to activate p21 gene expression in A431 cells.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Óxidos/toxicidad , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Trióxido de Arsénico , Arsenicales/administración & dosificación , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mutagénesis Sitio-Dirigida , Óxidos/administración & dosificación , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo
16.
J Biomed Sci ; 15(3): 333-42, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18210215

RESUMEN

Although arsenic is an infamous carcinogen, it has been effectively used to treat acute promyelocytic leukemia, and can induce cell cycle arrest or apoptosis in human solid tumors. Previously, we had demonstrated that opposing effects of ERK1/2 and JNK on p21 expression in response to arsenic trioxide (As(2)O(3)) are mediated through the Sp1 responsive elements of the p21 promoter in A431 cells. Presently, we demonstrate that Sp1, and c-Jun functionally cooperate to activate p21 promoter expression through Sp1 binding sites (-84/-64) by using DNA affinity binding, chromatin immunoprecipitation, and promoter assays. Surprisingly, As(2)O(3)-induced c-Jun(Ser63/73) phosphorylation can recruit TGIF/HDAC1 to the Sp1 binding sites and then suppress p21 promoter activation. We suggest that, after As(2)O(3 )treatment, the N-terminal domain of c-Jun phosphorylation by JNK recruits TGIF/HDAC1 to the Sp1 sites and then represses p21 expression. That is, TGIF is involved in As(2)O(3)-inhibited p21 expression, and then blocks the cell cycle arrest.


Asunto(s)
Arsenicales/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Regulación de la Expresión Génica/efectos de los fármacos , Óxidos/farmacología , Trióxido de Arsénico , Secuencia de Bases , Sitios de Unión , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Cartilla de ADN , Ensayo de Cambio de Movilidad Electroforética , Histona Desacetilasas/metabolismo , Humanos , Fosforilación , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción Sp1/metabolismo
17.
Cell Signal ; 18(2): 244-55, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15961274

RESUMEN

Arsenic has been effectively used to treat acute promyelocytic leukemia, and can induce cell cycle arrest or apoptosis in human solid tumors. Previously, we have demonstrated that As2O3 can induce p21WAF1/CIP1 (p21) expression in A431 cells and then due to cellular cytotoxicity. Presently, we have clarified these signaling events and compared them with EGF. Using reporter assay, RT-PCR and Western blotting, we show that c-Src activation might be a prerequisite for As2O3-induced EGFR/Ras/Raf/ERK signaling. Furthermore, with the aids of 5'-deletion and site-directed mutagenesis, we demonstrate that Sp1 binding sites, ranging from -64 to -84 bp, are essential for As2O3- or EGF-regulated p21 expression. Finally, our experiments utilizing cycloheximide prompt the suggestion that the stability of mRNA or protein also contributes to As2O3- or EGF-induced p21 expression. Taken together, we conclude that the Sp1 binding sites are required for As2O3-induced p21 gene transcription through c-Src/EGFR/Ras/Raf/ERK pathway. Furthermore, post-transcriptional or post-translational stabilization mechanism is also essential for As2O3-induced p21 expression. EGF-induced p21 expression may involve similar mechanisms as those that operate in the As2O3-mediated reactions in A431 cells.


Asunto(s)
Antineoplásicos/farmacología , Arsenicales/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Óxidos/farmacología , Trióxido de Arsénico , Arsenicales/antagonistas & inhibidores , Sitios de Unión , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Óxidos/antagonistas & inhibidores , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Factor de Transcripción Sp1/metabolismo , Activación Transcripcional , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
J Agric Food Chem ; 55(24): 9784-91, 2007 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-17960877

RESUMEN

Pycnogenol, polyphenolic compounds extracted from the pine bark, is beneficial for human health. To understand more of its effects, the present study is to explore the protein expression pattern induced by pycnogenol in RAW 264.7 cells. Global analysis using two-dimensional gel electrophoresis indicated that treatment with pycnogenol induces upregulation of four proteins, whose identities were revealed by mass spectrometry as cathepsin D, keratinocyte lipid-binding protein, proteasome subunit alpha type 1, and annexin IV. The pycnogenol effect displayed a time- and concentration-dependent manner. Unlike pycnogenol, N-acetyl cysteine and vitamin C had no effect on cathepsin D expression. Further studies showed that cathepsin D induction is correlated with an increase of lysosomal staining and enhancement of phagocytosis. These results reveal the novel effects of pycnogenol on protein expression and phagocytic functions and illustrate the advantage of proteomics-based strategy in unveiling the molecular basis of phytochemicals.


Asunto(s)
Catepsina D/metabolismo , Flavonoides/farmacología , Fagocitosis/efectos de los fármacos , Proteómica , Animales , Anexina A4 , Relación Dosis-Respuesta a Droga , Electroforesis en Gel Bidimensional/métodos , Proteínas de Unión a Ácidos Grasos , Expresión Génica , Macrófagos , Espectrometría de Masas , Ratones , Proteínas de Neoplasias , Fagocitosis/fisiología , Extractos Vegetales , Subunidades de Proteína , Factores de Tiempo
19.
Oncotarget ; 8(8): 13832-13845, 2017 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-28099155

RESUMEN

Metformin, as an AMP-activated protein kinase (AMPK) activator, can activate autophagy. A study showed that metformin decreased the risk of hepatocellular carcinoma (HCC) in diabetic patients. However, the detailed mechanism in the metformin-mediated anticancer effect remains an open question. Transcription factor CCAAT/enhancer-binding protein delta (CEBPD) has been suggested to serve as a tumor suppressor and is responsive to multiple anticancer drugs in HCC. In this study, we found that CEBPD and autophagy are involved in metformin-induced cell apoptosis in Huh7 cells. The underlying mechanisms in this process included a reduction in Src-mediated CEBPD protein degradation and an increase in CEBPD-regulated LC3B and ATG3 gene transcription under metformin treatment. We also found that AMPK is involved in metformin-induced CEBPD expression. Combined treatment with metformin and rapamycin can enhance autophagic cell death through the AMPK-dependent and AMPK-independent pathway, respectively. Taken together, we provide a new insight and therapeutic approach by targeting autophagy in the treatment of HCC.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Metformina/farmacología , Animales , Antineoplásicos/farmacología , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Hipoglucemiantes/farmacología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa
20.
FASEB J ; 17(12): 1694-6, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12958179

RESUMEN

Phospholipid hydroperoxide glutathione peroxidase (PHGPx), a selenium-dependent glutathione peroxidase, can interact with lipophilic substrates, including the phospholipid hydroperoxides, fatty-acid hydroperoxides, and cholesteryl ester hydroperoxides, and reduce them to hydroxide compounds. We studied the functional role of endogenous PHGPx in regulation of 12(S)-lipoxygenase and cyclooxygenase 1 activities in human epidermoid carcinoma A431 cells by using a cell system overexpressing anti-PHGPx mRNA. A retroviral expression vector designated as L1-3, wherein cDNA of PHGPx was reversely inserted into pFB-ERV in antisense orientation, was constructed. A number of stable transfectants of A431 cells with PHGPx depletion were generated from virions containing plasmid L1-3. In an intact cell assay system, the metabolism of arachidonic acid to prostaglandin E2 and 12(S)-hydroxyeicosatetraenoic acid was significantly enhanced in stable L1-3 transfectants compared with that in vector-control cells. Flow cytometric analysis revealed a significant elevated level of intracellular hydroperoxides in stable L1-3 transfectants. Treatment of stable L1-3 transfectants with 50 microM arsenite induced more significant formation of intracellular hydroperoxides than that of vector-control cells. Taken together, these results support the notion that the endogenous PHGPx plays a pivotal role in the regulation of 12(S)-lipoxygenase and cyclooxygenase 1 activities by reducing the level of intracellular lipid hydroperoxides in arachidonate metabolism in A431 cells.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Ácido Araquidónico/metabolismo , Carcinoma de Células Escamosas/enzimología , Glutatión Peroxidasa/fisiología , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Ciclooxigenasa 1 , Glutatión Peroxidasa/genética , Humanos , Peróxidos Lipídicos/biosíntesis , Proteínas de la Membrana , Modelos Biológicos , Fosfolípido Hidroperóxido Glutatión Peroxidasa , ARN sin Sentido/genética , Transfección , Células Tumorales Cultivadas , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA