Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 69(6): 923-937.e8, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29547721

RESUMEN

Dietary supplements such as vitamins and minerals are widely used in the hope of improving health but may have unidentified risks and side effects. In particular, a pathogenic link between dietary supplements and specific oncogenes remains unknown. Here we report that chondroitin-4-sulfate (CHSA), a natural glycosaminoglycan approved as a dietary supplement used for osteoarthritis, selectively promotes the tumor growth potential of BRAF V600E-expressing human melanoma cells in patient- and cell line-derived xenograft mice and confers resistance to BRAF inhibitors. Mechanistically, chondroitin sulfate glucuronyltransferase (CSGlcA-T) signals through its product CHSA to enhance casein kinase 2 (CK2)-PTEN binding and consequent phosphorylation and inhibition of PTEN, which requires CHSA chains and is essential to sustain AKT activation in BRAF V600E-expressing melanoma cells. However, this CHSA-dependent PTEN inhibition is dispensable in cancer cells expressing mutant NRAS or PI3KCA, which directly activate the PI3K-AKT pathway. These results suggest that dietary supplements may exhibit oncogene-dependent pro-tumor effects.


Asunto(s)
Carcinógenos/toxicidad , Transformación Celular Neoplásica/genética , Sulfatos de Condroitina/toxicidad , Suplementos Dietéticos/toxicidad , Melanoma/inducido químicamente , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/inducido químicamente , Animales , Antinematodos/farmacología , Quinasa de la Caseína II/metabolismo , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , GTP Fosfohidrolasas/genética , Células HEK293 , Células HT29 , Humanos , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Melanoma/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones Transgénicos , Células 3T3 NIH , Proteínas Nucleares/genética , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/genética , Factores de Transcripción/genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Mol Cell ; 59(3): 345-358, 2015 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-26145173

RESUMEN

Many human cancers share similar metabolic alterations, including the Warburg effect. However, it remains unclear whether oncogene-specific metabolic alterations are required for tumor development. Here we demonstrate a "synthetic lethal" interaction between oncogenic BRAF V600E and a ketogenic enzyme 3-hydroxy-3-methylglutaryl-CoA lyase (HMGCL). HMGCL expression is upregulated in BRAF V600E-expressing human primary melanoma and hairy cell leukemia cells. Suppression of HMGCL specifically attenuates proliferation and tumor growth potential of human melanoma cells expressing BRAF V600E. Mechanistically, active BRAF upregulates HMGCL through an octamer transcription factor Oct-1, leading to increased intracellular levels of HMGCL product, acetoacetate, which selectively enhances binding of BRAF V600E but not BRAF wild-type to MEK1 in V600E-positive cancer cells to promote activation of MEK-ERK signaling. These findings reveal a mutation-specific mechanism by which oncogenic BRAF V600E "rewires" metabolic and cell signaling networks and signals through the Oct-1-HMGCL-acetoacetate axis to selectively promote BRAF V600E-dependent tumor development.


Asunto(s)
Leucemia de Células Pilosas/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Melanoma/metabolismo , Factor 1 de Transcripción de Unión a Octámeros/metabolismo , Oxo-Ácido-Liasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal , Acetoacetatos/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Regulación hacia Arriba
3.
Mol Cell ; 53(4): 534-48, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24486017

RESUMEN

Mitochondrial pyruvate dehydrogenase complex (PDC) is crucial for glucose homeostasis in mammalian cells. The current understanding of PDC regulation involves inhibitory serine phosphorylation of pyruvate dehydrogenase (PDH) by PDH kinase (PDK), whereas dephosphorylation of PDH by PDH phosphatase (PDP) activates PDC. Here, we report that lysine acetylation of PDHA1 and PDP1 is common in epidermal growth factor (EGF)-stimulated cells and diverse human cancer cells. K321 acetylation inhibits PDHA1 by recruiting PDK1, and K202 acetylation inhibits PDP1 by dissociating its substrate PDHA1, both of which are important in promoting glycolysis in cancer cells and consequent tumor growth. Moreover, we identified mitochondrial ACAT1 and SIRT3 as the upstream acetyltransferase and deacetylase, respectively, of PDHA1 and PDP1, while knockdown of ACAT1 attenuates tumor growth. Furthermore, Y381 phosphorylation of PDP1 dissociates SIRT3 and recruits ACAT1 to PDC. Together, hierarchical, distinct posttranslational modifications act in concert to control molecular composition of PDC and contribute to the Warburg effect.


Asunto(s)
Acetil-CoA C-Acetiltransferasa/metabolismo , Piruvato Deshidrogenasa (Lipoamida)-Fosfatasa/metabolismo , Piruvato Deshidrogenasa (Lipoamida)/metabolismo , Sirtuina 3/metabolismo , Tirosina/química , Animales , Línea Celular Tumoral , Proliferación Celular , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , Lisina/química , Masculino , Ratones , Ratones Desnudos , Mitocondrias/metabolismo , Trasplante de Neoplasias , Neoplasias/metabolismo , Fosforilación
4.
Mol Cell ; 55(4): 552-65, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25042803

RESUMEN

Although the oxidative pentose phosphate pathway is important for tumor growth, how 6-phosphogluconate dehydrogenase (6PGD) in this pathway is upregulated in human cancers is unknown. We found that 6PGD is commonly activated in EGF-stimulated cells and human cancer cells by lysine acetylation. Acetylation at K76 and K294 of 6PGD promotes NADP(+) binding to 6PGD and formation of active 6PGD dimers, respectively. Moreover, we identified DLAT and ACAT2 as upstream acetyltransferases of K76 and K294, respectively, and HDAC4 as the deacetylase of both sites. Expressing acetyl-deficient mutants of 6PGD in cancer cells significantly attenuated cell proliferation and tumor growth. This is due in part to reduced levels of 6PGD products ribulose-5-phosphate and NADPH, which led to reduced RNA and lipid biosynthesis as well as elevated ROS. Furthermore, 6PGD activity is upregulated with increased lysine acetylation in primary leukemia cells from human patients, providing mechanistic insights into 6PGD upregulation in cancer cells.


Asunto(s)
Acetil-CoA C-Acetiltransferasa/metabolismo , Acetiltransferasa de Residuos Dihidrolipoil-Lisina/metabolismo , Histona Desacetilasas/metabolismo , Leucemia/patología , Neoplasias Pulmonares/patología , Lisina/metabolismo , Fosfogluconato Deshidrogenasa/metabolismo , Acetilación , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Leucemia/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , NADP/metabolismo , Neoplasias Experimentales , Unión Proteica/fisiología , Multimerización de Proteína
5.
J Biol Chem ; 292(24): 10142-10152, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28468827

RESUMEN

Contributions of metabolic changes to cancer development and maintenance have received increasing attention in recent years. Although many human cancers share similar metabolic alterations, it remains unclear whether oncogene-specific metabolic alterations are required for tumor development. Using an RNAi-based screen targeting the majority of the known metabolic proteins, we recently found that oncogenic BRAFV600E up-regulates HMG-CoA lyase (HMGCL), which converts HMG-CoA to acetyl-CoA and a ketone body, acetoacetate, that selectively enhances BRAFV600E-dependent MEK1 activation in human cancer. Here, we identified HMG-CoA synthase 1 (HMGCS1), the upstream ketogenic enzyme of HMGCL, as an additional "synthetic lethal" partner of BRAFV600E Although HMGCS1 expression did not correlate with BRAFV600E mutation in human melanoma cells, HMGCS1 was selectively important for proliferation of BRAFV600E-positive melanoma and colon cancer cells but not control cells harboring active N/KRAS mutants, and stable knockdown of HMGCS1 only attenuated colony formation and tumor growth potential of BRAFV600E melanoma cells. Moreover, cytosolic HMGCS1 that co-localized with HMGCL and BRAFV600E was more important than the mitochondrial HMGCS2 isoform in BRAFV600E-expressing cancer cells in terms of acetoacetate production. Interestingly, HMGCL knockdown did not affect HMGCS1 expression levels, whereas HMGCS1 knockdown caused a compensating increase in HMGCL protein level because of attenuated protein degradation. However, this increase did not reverse the reduced ketogenesis in HMGCS1 knockdown cells. Mechanistically, HMGCS1 inhibition decreased intracellular acetoacetate levels, leading to reduced BRAFV600E-MEK1 binding and consequent MEK1 activation. We conclude that the ketogenic HMGCS1-HMGCL-acetoacetate axis may represent a promising therapeutic target for managing BRAFV600E-positive human cancers.


Asunto(s)
Neoplasias del Colon/enzimología , Hidroximetilglutaril-CoA Sintasa/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Melanoma/enzimología , Proteínas de Neoplasias/metabolismo , Oxo-Ácido-Liasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Acetoacetatos/metabolismo , Sustitución de Aminoácidos , Animales , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Citosol/enzimología , Citosol/metabolismo , Activación Enzimática , Estabilidad de Enzimas , Femenino , Humanos , Hidroximetilglutaril-CoA Sintasa/antagonistas & inhibidores , Hidroximetilglutaril-CoA Sintasa/genética , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/metabolismo , MAP Quinasa Quinasa 1/química , Melanoma/metabolismo , Melanoma/patología , Ratones Desnudos , Mutación , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Oxo-Ácido-Liasas/antagonistas & inhibidores , Oxo-Ácido-Liasas/química , Oxo-Ácido-Liasas/genética , Proteolisis , Proteínas Proto-Oncogénicas B-raf/genética , Interferencia de ARN , Carga Tumoral
6.
BMC Cancer ; 18(1): 605, 2018 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-29843645

RESUMEN

BACKGROUND: Aberrant hyperactivation of epithelial proliferation, AKT signaling, and association with unopposed estrogen (E2) exposure is the most common endometrial cancer dysfunction. In the normal uterus, progesterone (P4) inhibits proliferation by coordinating stromal-epithelial cross-talk, which we previously showed is mediated by the function of Mitogen-inducible gene 6 (Mig-6). Despite their attractive characteristics, non-surgical conservative therapies based on progesterone alone have not been universally successful. One barrier to this success has been the lack of understanding of the P4 effect on endometrial cells. METHOD: To further understand the role of Mig-6 and P4 in controlling uterine proliferation, we developed a Sprr2f-cre driven mouse model where Mig-6 is specifically ablated only in the epithelial cells of the uterus (Sprr2f cre+ Mig-6 f/f ). We examined P4 effect and regulation of AKT signaling in the endometrium of mutant mice. RESULTS: Sprr2f cre+ Mig-6 f/f mice developed endometrial hyperplasia. P4 treatment abated the development of endometrial hyperplasia and restored morphological and histological characteristics of the uterus. P4 treatment reduced cell proliferation which was accompanied by decreased AKT signaling and the restoration of stromal PGR and ESR1 expression. Furthermore, our in vitro studies revealed an inhibitory effect of MIG-6 on AKT phosphorylation as well as MIG-6 and AKT protein interactions. CONCLUSIONS: These data suggest that endometrial epithelial cell proliferation is regulated by P4 mediated Mig-6 inhibition of AKT phosphorylation, uncovering new mechanisms of P4 action. This information may help guide more effective non-surgical interventions in the future.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Endometriales/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Progesterona/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proliferación Celular , Proteínas Ricas en Prolina del Estrato Córneo/genética , Endometrio/citología , Endometrio/metabolismo , Endometrio/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Modelos Animales , Fosforilación , Receptores de Progesterona/metabolismo , Transducción de Señal
7.
Hum Mol Genet ; 24(4): 1127-41, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25305081

RESUMEN

Deciphering the molecular basis of neuronal cell death is a central issue in the etiology of neurodegenerative diseases, such as Parkinson's and Alzheimer's. Dysregulation of p53 levels has been implicated in neuronal apoptosis. The role of histone deacetylase 3 (HDAC3) in suppressing p53-dependent apoptosis has been recently emphasized; however, the molecular basis of modulation of p53 function by HDAC3 remains unclear. Here we show that PTEN-induced putative kinase 1 (PINK1), which is linked to autosomal recessive early-onset familial Parkinson's disease, phosphorylates HDAC3 at Ser-424 to enhance its HDAC activity in a neural cell-specific manner. PINK1 prevents H2O2-induced C-terminal cleavage of HDAC3 via phosphorylation of HDAC3 at Ser-424, which is reversed by protein phosphatase 4c. PINK1-mediated phosphorylation of HDAC3 enhances its direct association with p53 and causes subsequent hypoacetylation of p53. Genetic deletion of PINK1 partly impaired the suppressive role of HDAC3 in regulating p53 acetylation and transcriptional activity. However, depletion of HDAC3 fully abolished the PINK1-mediated p53 inhibitory loop. Finally, ectopic expression of phosphomometic-HDAC3(S424E) substantially overcomes the defective action of PINK1 against oxidative stress in dopaminergic neuronal cells. Together, our results uncovered a mechanism by which PINK1-HDAC3 network mediates p53 inhibitory loop in response to oxidative stress-induced damage.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Quinasas/metabolismo , Acetilación/efectos de los fármacos , Animales , Caspasa 7/metabolismo , Muerte Celular/genética , Línea Celular , Citoplasma/metabolismo , Neuronas Dopaminérgicas/patología , Activación Enzimática , Histona Desacetilasas/genética , Humanos , Peróxido de Hidrógeno/farmacología , Ratones , Especificidad de Órganos , Fosforilación , Proteínas Quinasas/genética , Proteolisis , Proteína p53 Supresora de Tumor/metabolismo
8.
J Biol Chem ; 289(38): 26533-26541, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25104357

RESUMEN

The mitochondrial pyruvate dehydrogenase complex (PDC) plays a crucial role in regulation of glucose homoeostasis in mammalian cells. PDC flux depends on catalytic activity of the most important enzyme component pyruvate dehydrogenase (PDH). PDH kinase inactivates PDC by phosphorylating PDH at specific serine residues, including Ser-293, whereas dephosphorylation of PDH by PDH phosphatase restores PDC activity. The current understanding suggests that Ser-293 phosphorylation of PDH impedes active site accessibility to its substrate pyruvate. Here, we report that phosphorylation of a tyrosine residue Tyr-301 also inhibits PDH α 1 (PDHA1) by blocking pyruvate binding through a novel mechanism in addition to Ser-293 phosphorylation. In addition, we found that multiple oncogenic tyrosine kinases directly phosphorylate PDHA1 at Tyr-301, and Tyr-301 phosphorylation of PDHA1 is common in EGF-stimulated cells as well as diverse human cancer cells and primary leukemia cells from human patients. Moreover, expression of a phosphorylation-deficient PDHA1 Y301F mutant in cancer cells resulted in increased oxidative phosphorylation, decreased cell proliferation under hypoxia, and reduced tumor growth in mice. Together, our findings suggest that phosphorylation at distinct serine and tyrosine residues inhibits PDHA1 through distinct mechanisms to impact active site accessibility, which act in concert to regulate PDC activity and promote the Warburg effect.


Asunto(s)
Procesamiento Proteico-Postraduccional , Piruvato Deshidrogenasa (Lipoamida)/metabolismo , Células 3T3 , Sustitución de Aminoácidos , Animales , Metabolismo de los Hidratos de Carbono , Dominio Catalítico , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Factor de Crecimiento Epidérmico/fisiología , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fosforilación Oxidativa , Fosforilación , Unión Proteica , Piruvato Deshidrogenasa (Lipoamida)/química , Piruvato Deshidrogenasa (Lipoamida)/genética , Ácido Pirúvico/química , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Carga Tumoral , Tirosina/metabolismo
9.
J Biol Chem ; 289(31): 21413-22, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24962578

RESUMEN

Many cancer cells rely more on aerobic glycolysis (the Warburg effect) than mitochondrial oxidative phosphorylation and catabolize glucose at a high rate. Such a metabolic switch is suggested to be due in part to functional attenuation of mitochondria in cancer cells. However, how oncogenic signals attenuate mitochondrial function and promote the switch to glycolysis remains unclear. We previously reported that tyrosine phosphorylation activates and inhibits mitochondrial pyruvate dehydrogenase kinase (PDK) and phosphatase (PDP), respectively, leading to enhanced inhibitory serine phosphorylation of pyruvate dehydrogenase (PDH) and consequently inhibition of pyruvate dehydrogenase complex (PDC) in cancer cells. In particular, Tyr-381 phosphorylation of PDP1 dissociates deacetylase SIRT3 and recruits acetyltransferase ACAT1 to PDC, resulting in increased inhibitory lysine acetylation of PDHA1 and PDP1. Here we report that phosphorylation at another tyrosine residue, Tyr-94, inhibits PDP1 by reducing the binding ability of PDP1 to lipoic acid, which is covalently attached to the L2 domain of dihydrolipoyl acetyltransferase (E2) to recruit PDP1 to PDC. We found that multiple oncogenic tyrosine kinases directly phosphorylated PDP1 at Tyr-94, and Tyr-94 phosphorylation of PDP1 was common in diverse human cancer cells and primary leukemia cells from patients. Moreover, expression of a phosphorylation-deficient PDP1 Y94F mutant in cancer cells resulted in increased oxidative phosphorylation, decreased cell proliferation under hypoxia, and reduced tumor growth in mice. Together, our findings suggest that phosphorylation at different tyrosine residues inhibits PDP1 through independent mechanisms, which act in concert to regulate PDC activity and promote the Warburg effect.


Asunto(s)
División Celular , Neoplasias/patología , Piruvato Deshidrogenasa (Lipoamida)-Fosfatasa/antagonistas & inhibidores , Tirosina/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Humanos , Ácido Láctico/metabolismo , Datos de Secuencia Molecular , Neoplasias/enzimología , Consumo de Oxígeno , Fosforilación , Piruvato Deshidrogenasa (Lipoamida)-Fosfatasa/química , Piruvato Deshidrogenasa (Lipoamida)-Fosfatasa/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Homología de Secuencia de Aminoácido
10.
Sci Rep ; 13(1): 22111, 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38092801

RESUMEN

The multilevel current states of synaptic devices in artificial neural networks enable next-generation computing to perform cognitive functions in an energy-efficient manner. Moreover, considering large-scale synaptic arrays, multiple states programmed in a low-current regime may be required to achieve low energy consumption, as demonstrated by simple numerical calculations. Thus, we propose a three-terminal Cu-ion-actuated CuOx/HfOx/WO3 synaptic transistor array that exhibits analogously modulated channel current states in the range of tens of nanoamperes, enabled by WO3 channel engineering. The introduction of an amorphous stoichiometric WO3 channel formed by reactive sputtering with O gas significantly lowered the channel current but left it almost unchanged with respect to consecutive gate voltage pulses. An additional annealing process at 450 °C crystallized the WO3, allowing analog switching in the range of tens of nanoamperes. The incorporation of N gas during annealing induced a highly conductive channel, making the channel current modulation negligible as a function of the gate pulse. Using this optimized gate stack, Poole-Frenkel conduction was identified as a major transport characteristic in a temperature-dependent study. In addition, we found that the channel current modulation is a function of the gate current response, which is related to the degree of progressive movement of the Cu ions. Finally, the synaptic characteristics were updated using fully parallel programming and demonstrated in a 7 × 7 array. Using the CuOx/HfOx/WO3 synaptic transistors as weight elements in multilayer neural networks, we achieved a 90% recognition accuracy on the Fashion-MNIST dataset.

11.
Micromachines (Basel) ; 13(3)2022 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-35334745

RESUMEN

To enhance the computing efficiency in a neuromorphic architecture, it is important to develop suitable memory devices that can emulate the role of biological synapses. More specifically, not only are multiple conductance states needed to be achieved in the memory but each state is also analogously adjusted by consecutive identical pulses. Recently, electrochemical random-access memory (ECRAM) has been dedicatedly designed to realize the desired synaptic characteristics. Electric-field-driven ion motion through various electrolytes enables the conductance of the ECRAM to be analogously modulated, resulting in a linear and symmetric response. Therefore, the aim of this study is to review recent advances in ECRAM technology from the material and device engineering perspectives. Since controllable mobile ions play an important role in achieving synaptic behavior, the prospect and challenges of ECRAM devices classified according to mobile ion species are discussed.

12.
Cell Mol Life Sci ; 67(20): 3499-510, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20473547

RESUMEN

We identified CREB3 as a novel HDAC3-interacting protein in a yeast two-hybrid screen for HDAC3-interacting proteins. Among all class I HDACs, CREB3 specifically interacts with HDAC3, in vitro and in vivo. HDAC3 efficiently inhibited CREB3-enhanced NF-κB activation, whereas the other class I HDACs did not alter NF-κB-dependent promoter activities or the expression of NF-κB target genes. Importantly, both knock-down of CREB3 and overexpression of HDAC3 suppressed the transcriptional activation of the novel CREB3-regulated gene, CXCR4. Furthermore, CREB3 was shown to bind to the CRE element in the CXCR4 promoter and to activate the transcription of the CXCR4 gene by causing dissociation of HDAC3 and subsequently increasing histone acetylation. Importantly, both the depletion of HDAC3 and the overexpression of CREB3 substantially increased the migration of MDA-MB-231 metastatic breast cancer cells. Taken together, these findings suggest that HDAC3 selectively represses CREB3-mediated transcriptional activation and chemotactic signalling in human metastatic breast cancer cells.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Movimiento Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/química , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasas/química , Humanos , FN-kappa B/metabolismo , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
13.
Cell Death Dis ; 12(3): 250, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674559

RESUMEN

Tumors are composed of subpopulations of cancer cells with functionally distinct features. Intratumoral heterogeneity limits the therapeutic effectiveness of cancer drugs. To address this issue, it is important to understand the regulatory mechanisms driving a subclonal variety within a therapy-resistant tumor. We identified tumor subclones of HN9 head and neck cancer cells showing distinct responses to radiation with different levels of p62 expression. Genetically identical grounds but epigenetic heterogeneity of the p62 promoter regions revealed that radioresistant HN9-R clones displayed low p62 expression via the creation of repressive chromatin architecture, in which cooperation between DNMT1 (DNA methyltransferases 1) and HDAC1 (histone deacetylases 1) resulted in DNA methylation and repressive H3K9me3 and H3K27me3 marks in the p62 promoter. Combined inhibition of DNMT1 and HDAC1 by genetic depletion or inhibitors enhanced the suppressive effects on proliferative capacity and in vivo tumorigenesis following irradiation. Importantly, ectopically p62-overexpressed HN9-R clones increased the induction of senescence along with p62-dependent autophagy activation. These results demonstrate the heterogeneous expression of p62 as the key component of clonal variation within a tumor against irradiation. Understanding the epigenetic diversity of p62 heterogeneity among subclones allows for improved identification of the functional state of subclones and provides a novel treatment option to resolve resistance to current therapies.


Asunto(s)
Autofagia/efectos de los fármacos , Senescencia Celular/efectos de la radiación , Epigénesis Genética , Neoplasias de Cabeza y Cuello/radioterapia , Tolerancia a Radiación , Proteína Sequestosoma-1/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/radioterapia , Acetilación , Animales , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Islas de CpG , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Heterogeneidad Genética , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Histona Desacetilasa 1/metabolismo , Humanos , Masculino , Ratones Desnudos , Regiones Promotoras Genéticas , Tolerancia a Radiación/genética , Proteína Sequestosoma-1/genética , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Sci Rep ; 10(1): 7620, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32376896

RESUMEN

Epithelial-mesenchymal transition (EMT) is a major cellular process in which epithelial cells lose cell polarity and cell-cell adhesion and become motility and invasiveness by transforming into mesenchymal cells. Catechol is one of the natural compounds present in fruits and vegetables and has various pharmacological and physiological activities including anti-carcinogenic effects. However, the effects of catechol on EMT has not been reported. Epidermal growth factor (EGF) is one of the growth factors and is known to play a role in inducing EMT. The present study showed that catechol suppressed not only the morphological changes to the mesenchymal phenotype of epithelial HCC cells, but also the reduction of E-cadherin and the increment of Vimentin, which are typical hallmark of EMT. In addition, catechol suppressed EMT-related steps such as migration, invasion, anoikis resistance acquisition, and stem cell-like characterization through the EGFR-AKT-ERK signaling pathway during liver cancer metastasis. Therefore, these results suggest that catechol may be able to regulate the early metastasis of liver cancer in vitro.


Asunto(s)
Carcinoma Hepatocelular/patología , Catecoles/farmacología , Factor de Crecimiento Epidérmico/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias Hepáticas/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción de la Familia Snail/metabolismo
15.
Mol Endocrinol ; 22(5): 1093-104, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18202150

RESUMEN

Lis-homology (LisH) motifs are involved in protein dimerization, and the discovery of the conserved N-terminal LisH domain in transducin beta-like protein 1 and its receptor (TBL1 and TBLR1) led us to examine the role of this domain in transcriptional repression. Here we show that multiple beta-transducin (WD-40) repeat-containing proteins interact to form oligomers in solution and that oligomerization depends on the presence of the LisH domain in each protein. Repression of transcription, as assayed using Gal4 fusion proteins, also depended on the presence of the LisH domain, suggesting that oligomerization is a prerequisite for efficient transcriptional repression. Furthermore, we show that the LisH domain is responsible for the binding to the hypoacetylated histone H4 tail and for stable chromatin targeting by the nuclear receptor corepressor complex. Mutations in conserved residues in the LisH motif of TBL1 and TBLR1 block histone binding, oligomerization, and transcriptional repression, supporting the functional importance of the LisH motif in transcriptional repression. Our results indicate that another WD-40 protein, TBL3, also preferentially binds to the N-terminal domain of TBL1 and TBLR1, and forms oligomers with other WD-40 proteins. Finally, we observed that the WD-40 proteins RbAp46 and RbAp48 of the sin3A corepressor complex failed to dimerize. We also found the specific interaction UbcH/E2 with TBL1, but not RbAp46/48. Altogether, our results thus indicate that the presence of multiple LisH/WD-40 repeat containing proteins is exclusive to nuclear receptor corepressor/ silencing mediator for retinoic and thyroid receptor complexes compared with other class 1 histone deacetylase-containing corepessor complexes.


Asunto(s)
Proteínas Nucleares/metabolismo , Receptores de Ácido Retinoico/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Sitios de Unión/genética , Western Blotting , Línea Celular , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Análisis Mutacional de ADN , Células HeLa , Histonas/metabolismo , Humanos , Inmunoprecipitación , Proteínas Nucleares/genética , Co-Represor 1 de Receptor Nuclear , Unión Proteica , Receptores de Ácido Retinoico/genética , Receptores de Hormona Tiroidea/genética , Secuencias Repetitivas de Aminoácido/genética , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido
16.
Biochem J ; 411(1): 19-26, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18052923

RESUMEN

A central issue in mediating repression by nuclear hormone receptors is the distinct or redundant function between co-repressors N-CoR (nuclear receptor co-repressor) and SMRT (silencing mediator of retinoid and thyroid hormone receptor). To address the functional relationship between SMRT and N-CoR in TR (thyroid hormone receptor)-mediated repression, we have identified multiple TR target genes, including BCL3 (B-cell lymphoma 3-encoded protein), Spot14 (thyroid hormone-inducible hepatic protein), FAS (fatty acid synthase), and ADRB2 (beta-adrenergic receptor 2). We demonstrated that siRNA (small interfering RNA) treatment against either N-CoR or SMRT is sufficient for the de-repression of multiple TR target genes. By the combination of sequence mining and physical association as determined by ChIP (chromatin immunoprecipitation) assays, we mapped the putative TREs (thyroid hormone response elements) in BCL3, Spot14, FAS and ADRB2 genes. Our data clearly show that SMRT and N-CoR are independently recruited to various TR target genes. We also present evidence that overexpression of N-CoR can restore repression of endogenous genes after knocking down SMRT. Finally, unliganded, co-repressor-free TR is defective in repression and interacts with a co-activator, p300. Collectively, these results suggest that both SMRT and N-CoR are limited in cells and that knocking down either of them results in co-repressor-free TR and consequently de-repression of TR target genes.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas Nucleares/fisiología , Proteínas Represoras/fisiología , Receptores alfa de Hormona Tiroidea/genética , Transcripción Genética , Proteínas del Linfoma 3 de Células B , Ácido Graso Sintasas/genética , Células HeLa , Humanos , Inmunoprecipitación , Proteínas Nucleares/genética , Co-Represor 1 de Receptor Nuclear , Co-Represor 2 de Receptor Nuclear , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Elementos de Respuesta , Factores de Transcripción/genética , Transfección
17.
J Clin Invest ; 129(6): 2431-2445, 2019 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-31081803

RESUMEN

How altered metabolism contributes to chemotherapy resistance in cancer cells remains unclear. Through a metabolism-related kinome RNAi screen, we identified inositol-trisphosphate 3-kinase B (ITPKB) as a critical enzyme that contributes to cisplatin-resistant tumor growth. We demonstrated that inositol 1,3,4,5-tetrakisphosphate (IP4), the product of ITPKB, plays a critical role in redox homeostasis upon cisplatin exposure by reducing cisplatin-induced ROS through inhibition of a ROS-generating enzyme, NADPH oxidase 4 (NOX4), which promotes cisplatin-resistant tumor growth. Mechanistically, we identified that IP4 competes with the NOX4 cofactor NADPH for binding and consequently inhibits NOX4. Targeting ITPKB with shRNA or its small-molecule inhibitor resulted in attenuation of NOX4 activity, imbalanced redox status, and sensitized cancer cells to cisplatin treatment in patient-derived xenografts. Our findings provide insight into the crosstalk between kinase-mediated metabolic regulation and platinum-based chemotherapy resistance in human cancers. Our study also suggests a distinctive signaling function of IP4 that regulates NOX4. Furthermore, pharmaceutical inhibition of ITPKB displayed synergistic attenuation of tumor growth with cisplatin, suggesting ITPKB as a promising synthetic lethal target for cancer therapeutic intervention to overcome cisplatin resistance.


Asunto(s)
Cisplatino/farmacología , Resistencia a Antineoplásicos , NADPH Oxidasa 4/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Transducción de Señal , Células A549 , Animales , Femenino , Humanos , Ratones , Ratones Desnudos , NADPH Oxidasa 4/genética , Proteínas de Neoplasias/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Oxidación-Reducción/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Med Food ; 21(8): 793-800, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30048215

RESUMEN

Obesity is the most common metabolic disease in developed countries and has become a global epidemic in recent years. Obesity is associated with various metabolic abnormalities, including glucose intolerance, insulin resistance, type 2 diabetes, dyslipidemia, and hypertension. Leaves from the plant Dendropanax morbiferus are beneficial to health as they contain high levels of vitamin C and tannin. There have been seminal studies on the anticancer, antimicrobial, antidiabetes, and antihyperglycemic effects of treatments with D. morbiferus trees. Herein, we investigated the toxicity of D. morbiferus water (DLW) extracts in vitro, and demonstrated no toxicity at 5-500 µg/mL in 24-72-h experiments with 3T3-L1 cells. The DLW increased cell viability at 48 h and inhibited adipogenesis in 3T3-L1 cells by reducing intracellular triglyceride levels and glucose uptake. In addition, mRNA and protein expression levels of adipogenesis-related genes were lowered by DLW, suggesting antiobesity effects in mouse 3T3-L1 cells. Because few studies have demonstrated cholesterol-lowering effects of D. morbiferus, we investigated the activities of adipogenic transcriptional factors following treatments of 3T3-L1 cells with D. morbiferus and observed increased CEBPα, CEBPß, PPARγ, and SREBP1 activities in the cells, indicating that DLW extracts inhibit adipogenesis.


Asunto(s)
Células 3T3-L1/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Araliaceae , Obesidad/tratamiento farmacológico , Extractos Vegetales/farmacología , Células 3T3-L1/metabolismo , Animales , Fármacos Antiobesidad/uso terapéutico , Colesterol/metabolismo , Ratones , Fitoterapia , Extractos Vegetales/uso terapéutico , Triglicéridos/metabolismo
19.
Cancer Cell ; 34(2): 315-330.e7, 2018 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-30033091

RESUMEN

Platinum-based chemotherapeutics represent a mainstay of cancer therapy, but resistance limits their curative potential. Through a kinome RNAi screen, we identified microtubule-associated serine/threonine kinase 1 (MAST1) as a main driver of cisplatin resistance in human cancers. Mechanistically, cisplatin but no other DNA-damaging agents inhibit the MAPK pathway by dissociating cRaf from MEK1, while MAST1 replaces cRaf to reactivate the MAPK pathway in a cRaf-independent manner. We show clinical evidence that expression of MAST1, both initial and cisplatin-induced, contributes to platinum resistance and worse clinical outcome. Targeting MAST1 with lestaurtinib, a recently identified MAST1 inhibitor, restores cisplatin sensitivity, leading to the synergistic attenuation of cancer cell proliferation and tumor growth in human cancer cells and patient-derived xenograft models.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , MAP Quinasa Quinasa 1/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos , Activación Enzimática , Femenino , Humanos , Ratones
20.
Exp Mol Med ; 39(4): 544-55, 2007 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-17934343

RESUMEN

We have investigated the function and mechanisms of the CARM1-SNF5 complex in T3-dependent transcriptional activation. Using specific small interfering RNAs (siRNA) to knock down coactivators in HeLa alpha2 cells, we found that coactivator associated arginine methyltransferase 1 (CARM1) and SWI/SNF complex component 5 (SNF5) are important for T3-dependent transcriptional activation. The CARM1- SWI/SNF chromatin remodeling complex serves as a mechanism for the rapid reversal of H3-K9 methylation. Importantly, siRNA treatment against CARM1 and/or SNF5 increased the recruitment of HMTase G9a to the type 1 deiodinase (D1) promoter even with T3. Knocking-down either CARM1 or SNF5 also inhibited the down-regulation of histone macroH2A, which is correlated with transcriptional activation. Finally, knocking down CARM1 and SNF5 by siRNA impaired the association of these coactivators to the D1 promoter, suggesting functional importance of CARM1- SNF5 complex in T3-dependent transcriptional activation.


Asunto(s)
Proteínas Cromosómicas no Histona/fisiología , Proteínas de Unión al ADN/fisiología , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína-Arginina N-Metiltransferasas/fisiología , Receptores de Hormona Tiroidea/fisiología , Factores de Transcripción/fisiología , Activación Transcripcional , Células HeLa , Histona Metiltransferasas , Histonas/metabolismo , Humanos , Yoduro Peroxidasa/metabolismo , Metilación , Regiones Promotoras Genéticas , Proteína Metiltransferasas , Proteína SMARCB1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA