Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 55(1): 31-46, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24882211

RESUMEN

MutS protein homolog 2 (MSH2) is a key DNA mismatch repair protein. It forms the MSH2-MSH6 (MutSα) and MSH2-MSH3 (MutSß) heterodimers, which help to ensure genomic integrity. MutSα not only recognizes and repairs mismatched nucleotides but also recognizes DNA adducts induced by DNA-damaging agents, and triggers cell-cycle arrest and apoptosis. Loss or depletion of MutSα from cells leads to microsatellite instability (MSI) and resistance to DNA damage. Although the level of MutSα can be reduced by the ubiquitin-proteasome pathway, the detailed mechanisms of this regulation remain elusive. Here we report that histone deacetylase 6 (HDAC6) sequentially deacetylates and ubiquitinates MSH2, leading to MSH2 degradation. In addition, HDAC6 significantly reduces cellular sensitivity to DNA-damaging agents and decreases cellular DNA mismatch repair activities by downregulation of MSH2. Overall, these findings reveal a mechanism by which proper levels of MutSα are maintained.


Asunto(s)
Histona Desacetilasas/fisiología , Proteína 2 Homóloga a MutS/metabolismo , Acetilación , Animales , Células Cultivadas , Células HEK293 , Células HeLa , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Ratones , Estabilidad Proteica , Ubiquitinación
2.
Angew Chem Int Ed Engl ; 60(30): 16404-16408, 2021 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-33979017

RESUMEN

Li-CO2 batteries are regarded as next-generation high-energy-density electrochemical devices. However, the greatest challenge arises from the formation of the discharge product, Li2 CO3 , which would accumulate and deactivate heterogenous catalysts to cause huge polarization. Herein, Ru(bpy)3 Cl2 was employed as a solution-phase catalyst for Li-CO2 batteries and proved to be the most effective one screened so far. Spectroscopy and electrochemical analyses elucidate that the RuII center could interact with both CO2 and amorphous Li2 C2 O4 intermediate, thus promoting electroreduction process and delaying carbonate transformation. As a result, the charge potential is reduced to 3.86 V and over 60 discharge/charge cycles are achieved with a fixed capacity of 1000 mAh g-1 at a current density of 300 mA g-1 . Our work provides a new avenue to improve the electrochemical performance of Li-CO2 batteries with efficient mobile catalysts.

3.
J Biol Chem ; 294(15): 5813-5826, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30770470

RESUMEN

MutL homolog 1 (MLH1) is a key DNA mismatch repair protein, which plays an important role in maintenance of genomic stability and the DNA damage response. Here, we report that MLH1 is a novel substrate of histone deacetylase 6 (HDAC6). HDAC6 interacts with and deacetylates MLH1 both in vitro and in vivo Interestingly, deacetylation of MLH1 blocks the assembly of the MutSα-MutLα complex. Moreover, we have identified four novel acetylation sites in MLH1 by MS analysis. The deacetylation mimetic mutant, but not the WT and the acetylation mimetic mutant, of MLH1 confers resistance to 6-thioguanine. Overall, our findings suggest that the MutSα-MutLα complex serves as a sensor for DNA damage response and that HDAC6 disrupts the MutSα-MutLα complex by deacetylation of MLH1, leading to the tolerance of DNA damage.


Asunto(s)
Daño del ADN , Histona Desacetilasa 6/metabolismo , Homólogo 1 de la Proteína MutL/metabolismo , Acetilación/efectos de los fármacos , Línea Celular , Histona Desacetilasa 6/genética , Humanos , Homólogo 1 de la Proteína MutL/genética , Proteínas MutL/genética , Proteínas MutL/metabolismo , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/genética , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/metabolismo , Mutación , Tioguanina/farmacología
4.
Am J Gastroenterol ; 115(7): 1036-1044, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32618654

RESUMEN

INTRODUCTION: Data on the associations between esophageal histological lesions and risk of esophageal squamous cell carcinoma (ESCC) in general populations are limited. We aimed to investigate these associations in a large Chinese general population to inform future Chinese ESCC screening guidelines. METHODS: We performed endoscopic screening of 21,111 participants aged 40-69 years from 3 high-risk areas of China in 2005-2009, and followed the cohort through 2016. Cumulative incidence and mortality rates of ESCC were calculated by baseline histological diagnosis, and hazard ratios of ESCC, overall and by age and sex, were assessed using the Cox proportional hazards models. RESULTS: We identified 143 new ESCC cases (0.68%) and 62 ESCC deaths (0.29%) during a median follow-up of 8.5 years. Increasing grades of squamous dysplasia were associated with the increasing risk of ESCC incidence and mortality. The cumulative ESCC incidence rates for severe dysplasia/carcinoma in situ, moderate dysplasia (MD), and mild dysplasia were 15.5%, 4.5%, and 1.4%, respectively. Older individuals (50-69 years) had 3.1 times higher ESCC incidence than younger individuals (40-49 years), and men had 2.4 times higher ESCC incidence than women. DISCUSSION: This study confirmed that increasing grades of squamous dysplasia are associated with increasing risk of ESCC and that severe dysplasia and carcinoma in situ require clinical treatment. This study suggests that in high-risk areas of China, patients with endoscopically worrisome MD should also receive therapy, the first screening can be postponed to 50 years, and endoscopic surveillance intervals for unremarkable MD and mild dysplasia can be lengthened to 3 and 5 years, respectively.


Asunto(s)
Neoplasias Esofágicas/epidemiología , Neoplasias Esofágicas/patología , Lesiones Precancerosas/epidemiología , Lesiones Precancerosas/patología , Adulto , Anciano , Biopsia , China/epidemiología , Esofagoscopía , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Riesgo , Encuestas y Cuestionarios
5.
J Biol Chem ; 293(6): 1976-1993, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29259132

RESUMEN

Histone deacetylase 6 (HDAC6), a class IIb HDAC, plays an important role in many biological and pathological processes. Previously, we found that ERK1, a downstream kinase in the mitogen-activated protein kinase signaling pathway, phosphorylates HDAC6, thereby increasing HDAC6-mediated deacetylation of α-tubulin. However, whether HDAC6 reciprocally modulates ERK1 activity is unknown. Here, we report that both ERK1 and -2 are acetylated and that HDAC6 promotes ERK1 activity via deacetylation. Briefly, we found that both ERK1 and -2 physically interact with HDAC6. Endogenous ERK1/2 acetylation levels increased upon treatment with a pan-HDAC inhibitor, an HDAC6-specific inhibitor, or depletion of HDAC6, suggesting that HDAC6 deacetylates ERK1/2. We also noted that the acetyltransferases CREB-binding protein and p300 both can acetylate ERK1/2. Acetylated ERK1 exhibits reduced enzymatic activity toward the transcription factor ELK1, a well-known ERK1 substrate. Furthermore, mass spectrometry analysis indicated Lys-72 as an acetylation site in the ERK1 N terminus, adjacent to Lys-71, which binds to ATP, suggesting that acetylation status of Lys-72 may affect ERK1 ATP binding. Interestingly, an acetylation-mimicking ERK1 mutant (K72Q) exhibited less phosphorylation than the WT enzyme and a deacetylation-mimicking mutant (K72R). Of note, the K72Q mutant displayed decreased enzymatic activity in an in vitro kinase assay and in a cellular luciferase assay compared with the WT and K72R mutant. Taken together, our findings suggest that HDAC6 stimulates ERK1 activity. Along with our previous report that ERK1 promotes HDAC6 activity, we propose that HDAC6 and ERK1 may form a positive feed-forward loop, which might play a role in cancer.


Asunto(s)
Histona Desacetilasa 6/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Acetilación , Secuencias de Aminoácidos , Animales , Cristalografía por Rayos X , Histona Desacetilasa 6/química , Histona Desacetilasa 6/genética , Humanos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/química , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/química , Proteína Quinasa 3 Activada por Mitógenos/genética , Mutación , Fosforilación , Unión Proteica , Proteína Elk-1 con Dominio ets/genética , Proteína Elk-1 con Dominio ets/metabolismo
6.
J Biol Chem ; 291(20): 10783-91, 2016 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-26975374

RESUMEN

MSH2 is a key DNA mismatch repair protein, which plays an important role in genomic stability. In addition to its DNA repair function, MSH2 serves as a sensor for DNA base analogs-provoked DNA replication errors and binds to various DNA damage-induced adducts to trigger cell cycle arrest or apoptosis. Loss or depletion of MSH2 from cells renders resistance to certain DNA-damaging agents. Therefore, the level of MSH2 determines DNA damage response. Previous studies showed that the level of MSH2 protein is modulated by the ubiquitin-proteasome pathway, and histone deacetylase 6 (HDAC6) serves as an ubiquitin E3 ligase. However, the deubiquitinating enzymes, which regulate MSH2 remain unknown. Here we report that ubiquitin-specific peptidase 10 (USP10) interacts with and stabilizes MSH2. USP10 deubiquitinates MSH2 in vitro and in vivo Moreover, the protein level of MSH2 is positively correlated with the USP10 protein level in a panel of lung cancer cell lines. Knockdown of USP10 in lung cancer cells exhibits increased cell survival and decreased apoptosis upon the treatment of DNA-methylating agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) and antimetabolite 6-thioguanine (6-TG). The above phenotypes can be rescued by ectopic expression of MSH2. In addition, knockdown of MSH2 decreases the cellular mismatch repair activity. Overall, our results suggest a novel USP10-MSH2 pathway regulating DNA damage response and DNA mismatch repair.


Asunto(s)
Daño del ADN , Proteína 2 Homóloga a MutS/genética , Reparación del ADN , Proteínas de Unión al ADN/genética , Humanos , Péptido Hidrolasas/genética , Ubiquitinas/genética
7.
J Biol Chem ; 289(18): 12217-31, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24619425

RESUMEN

Fe65 is a multidomain adaptor with established functions in neuronal cells and neurodegeneration diseases. It binds to the C terminus of the Aß amyloid precursor protein and is involved in regulating gene transcription. The present studies show that Fe65 is expressed in breast cancer (BCa) cells and acts as an ERα transcriptional coregulator that is recruited by 17ß-estradiol to the promoters of estrogen target genes. Deletion analyses mapped the ERα binding domain to the phosphotyrosine binding domain 2 (PTB2). Ectopic Fe65 increased the transcriptional activity of the ERα in a PTB2-dependent manner in reporter assays. Fe65 knockdown decreased, whereas its stable expression increased the transcriptional activity of endogenous ERα in BCa cells and the ability of estrogens to stimulate target gene expression, ERα, and coactivator recruitment to target gene promoters and cell growth. Furthermore, Fe65 expression decreased the antagonistic activity of tamoxifen (TAM), suggesting a role for Fe65 in TAM resistance. Overall, the studies define a novel role for the neuronal adaptor in estrogen actions in BCa cells.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Sitios de Unión/genética , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Células Epiteliales/metabolismo , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Inmunohistoquímica , Células MCF-7 , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/metabolismo , Mutación , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Unión Proteica/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tamoxifeno/farmacología
8.
J Biol Chem ; 288(46): 33156-70, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24089523

RESUMEN

Histone deacetylase 6 (HDAC6) is well known for its ability to promote cell migration through deacetylation of its cytoplasmic substrates such as α-tubulin. However, how HDAC6 itself is regulated to control cell motility remains elusive. Previous studies have shown that one third of extracellular signal-regulated kinase (ERK) is associated with the microtubule cytoskeleton in cells. Yet, no connection between HDAC6 and ERK has been discovered. Here, for the first time, we reveal that ERK binds to and phosphorylates HDAC6 to promote cell migration via deacetylation of α-tubulin. We have identified two novel ERK-mediated phosphorylation sites: threonine 1031 and serine 1035 in HDAC6. Both sites were phosphorylated by ERK1 in vitro, whereas Ser-1035 was phosphorylated in response to the activation of EGFR-Ras-Raf-MEK-ERK signaling pathway in vivo. HDAC6-null mouse embryonic fibroblasts rescued by the nonphosphorylation mimicking mutant displayed significantly reduced cell migration compared with those rescued by the wild type. Consistently, the nonphosphorylation mimicking mutant exerted lower tubulin deacetylase activity in vivo compared with the wild type. These data indicate that ERK/HDAC6-mediated cell motility is through deacetylation of α-tubulin. Overall, our results suggest that HDAC6-mediated cell migration could be governed by EGFR-Ras-Raf-MEK-ERK signaling.


Asunto(s)
Movimiento Celular/fisiología , Histona Desacetilasas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Tubulina (Proteína)/metabolismo , Acetilación , Animales , Células CHO , Cricetinae , Cricetulus , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Humanos , Ratones , Ratones Mutantes , Proteína Quinasa 3 Activada por Mitógenos/genética , Tubulina (Proteína)/genética
9.
Nat Cell Biol ; 9(11): 1253-62, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17934453

RESUMEN

SIRT1 is the closest mammalian homologue of yeast SIR2, an important ageing regulator that prolongs lifespan in response to caloric restriction. Despite its importance, the mechanisms that regulate SIRT1 activity are unclear. Our study identifies a novel post-translational modification of SIRT1, namely sumoylation at Lys 734. In vitro sumoylation of SIRT1 increased its deacetylase activity. Conversely, mutation of SIRT1 at Lys 734 or desumoylation by SENP1, a nuclear desumoylase, reduced its deacetylase activity. Stress-inducing agents promoted the association of SIRT1 with SENP1 and cells depleted of SENP1 (but not of SENP1 and SIRT1) were more resistant to stress-induced apoptosis than control cells. We suggest that stress-inducing agents counteract the anti-apoptotic activity of SIRT1 by recruiting SENP1 to SIRT1, which results in the desumoylation and inactivation of SIRT1 and the consequent acetylation and activation of apoptotic proteins.


Asunto(s)
Acetilesterasa/efectos de los fármacos , Daño del ADN , Procesamiento Proteico-Postraduccional , Sirtuinas/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Acetilación , Animales , Apoptosis , Línea Celular , Cisteína Endopeptidasas , Endopeptidasas/metabolismo , Humanos , Proteínas/metabolismo , Sirtuina 1 , Sirtuinas/genética , Sirtuinas/farmacología , Proteína p53 Supresora de Tumor/fisiología
10.
Sci Total Environ ; 934: 173137, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38740207

RESUMEN

Non-conventional water recovery, recycling, and reuse have been considered imperative approaches to addressing water scarcity in China. The objective of this study was to evaluate the technical and economic feasibility of Water Reclamation Plants (WRP) based on an anaerobic-anoxic-oxic membrane bioreactor (A2O-MBR) system for unconventional water resource treatment and reuse in towns (domestic sewage and rainwater). Rainwater is collected and stored in the rainwater reservoir through the rainwater pipe network, and then transported to the WRP for treatment and reuse through the rainwater reuse pumping station during the peak water demand period. During a year of operation and evaluation process, a total of 610,000 cubic meters of rainwater were reused, accounting for 10.4 % of the treated wastewater. In the A2O-MBR operation, the average effluent concentrations for COD (chemical oxygen demand), NH4+-N (ammonium), TN (total nitrogen), and TP (total phosphorus) were 14.23 ± 4.07 mg/L, 0.22 ± 0.26 mg/L, 11.97 ± 1.54 mg/L, and 0.13 ± 0.09 mg/L, respectively. The effluent quality met standards suitable for reuse in industrial cooling water or for direct discharge. The WRP demonstrates a positive financial outlook, with total capital and operating costs totaling 0.16 $/m3. A comprehensive cost-benefit analysis indicates a positive net present value for the WRP, and the estimated annualized net profit is 0.024 $/m3. This research has achieved near-zero discharge of wastewater and effective allocation of rainwater resources across time and space.

11.
J Biol Chem ; 287(49): 41297-309, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23055531

RESUMEN

Telomerase is an essential enzyme that counteracts the telomere attrition accompanying DNA replication during cell division. Regulation of the promoter activity of the gene encoding its catalytic subunit, the telomerase reverse transcriptase, is established as the dominant mechanism conferring the high telomerase activity in proliferating cells, such as embryonic stem and cancer cells. This study reveals a new mechanism of telomerase regulation through non-coding small RNA by showing that microRNA-498 (miR-498) induced by 1,25-dihydroxyvitamin D3 (1,25(OH)(2)D(3)) decreases the mRNA expression of the human telomerase reverse transcriptase. MiR-498 was first identified in a microarray analysis as the most induced microRNA by 1,25(OH)(2)D(3) in ovarian cancer cells and subsequently validated by quantitative polymerase chain reaction assays in multiple human cancer types. A functional vitamin D response element was defined in the 5-prime regulatory region of the miR-498 genome, which is occupied by the vitamin D receptor and its coactivators. Further studies showed that miR-498 targeted the 3-prime untranslated region of human telomerase reverse transcriptase mRNA and decreased its expression. The levels of miR-498 expression were decreased in malignant human ovarian tumors as well as human ovarian cancer cell lines. The ability of 1,25(OH)(2)D(3) to decrease human telomerase reverse transcriptase mRNA and to suppress ovarian cancer growth was compromised when miR-498 was depleted using the sponges in cell lines and mouse tumor models. Taken together, our studies define a novel mechanism of telomerase regulation by small non-coding RNAs and identify miR-498 as an important mediator for the anti-tumor activity of 1,25(OH)(2)D(3).


Asunto(s)
Calcitriol/farmacología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , MicroARNs/biosíntesis , Neoplasias/metabolismo , Telomerasa/antagonistas & inhibidores , Telomerasa/biosíntesis , Animales , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Femenino , Genoma , Humanos , Ratones , Ratones Desnudos , MicroARNs/fisiología , Mutagénesis , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN no Traducido/metabolismo
12.
J Biochem ; 174(2): 183-191, 2023 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-37094360

RESUMEN

The DNA mismatch repair (MMR) proteins recognize and repair DNA base pair mismatches and insertions/deletions of DNA that have occurred during DNA replication. Additionally, they are involved in regulation of the DNA damage response, including cell cycle checkpoints and apoptosis. Therefore, regulation of these proteins is essential for maintaining genomic integrity. It has been recognized that post-translational modifications, such as phosphorylation, ubiquitination, and acetylation, are being used as an important means to regulate the functions and stability of MMR proteins. Here, we report that a histone acetyltransferase CREB binding protein (CBP) interacts with and acetylates MLH1, a component of the MutLα complex (MLH1-PMS2). Moreover, CBP stabilizes MLH1 by preventing it from degradation via the ubiquitin-proteasome degradation pathway. Consistently, acetylation induced by a pan-histone deacetylase inhibitor, Trichostatin A, promotes the assembly between the MutSα (MSH2-MSH6) and MutLα complexes. Furthermore, overexpression of CBP enhances MMR activities in cells. Overall, our results suggest a novel role of CBP in prolonging MLH1 stability and enhancing MutSα-MutLα complex formation, leading to increased cellular MMR activity.


Asunto(s)
Proteína de Unión a CREB , Reparación de la Incompatibilidad de ADN , Acetilación , Reparación del ADN , ADN , Procesamiento Proteico-Postraduccional
13.
FEBS J ; 288(14): 4364-4381, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33492759

RESUMEN

Inhibitor of DNA-binding 1 (ID1) protein has been studied intensively for its functions in tumorigenesis and maintenance of stem cell-like properties, but its roles in virus infection are less understood. In the present study, we have clearly shown that the foot-and-mouth disease virus (FMDV) promotes ID1 degradation via Cdh1-mediated ubiquitination to facilitate its replication. Mechanistic investigations reveal Forkhead Box O1 (FOXO1) as an ID1 partner, which suppresses interferon regulatory factors 3 expression and interferon (IFN) production. Further investigation identified that ID1 suppresses FOXO1 transcription activity through HDAC4-mediated deacetylation, promoting IFN production and antiviral immune response. These studies establish a prominent role for ID1 in suppressing FDMV replication, which may be extended to other viruses.


Asunto(s)
Virus de la Fiebre Aftosa/aislamiento & purificación , Fiebre Aftosa/prevención & control , Interacciones Huésped-Patógeno , Proteína 1 Inhibidora de la Diferenciación/fisiología , Replicación Viral , Acetilación , Animales , Femenino , Fiebre Aftosa/virología , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferones/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
14.
Mol Cell Biol ; 27(4): 1321-33, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17130235

RESUMEN

The C-terminal fragment of the beta-amyloid precursor protein produced after cleavage by gamma-secretase, namely, APPct or AICD, has been shown to form a multimeric complex with the adaptor protein Fe65 and to regulate transcription through the recruitment of the histone acetyltransferase Tip60. The present study shows that 17beta-estradiol inhibits the transcriptional and apoptotic activities of the APPct complex by a process involving the interaction of estrogen receptor alpha (ERalpha) with Fe65. ERalpha-Fe65 complexes were detected both in vitro and in the mouse brain, and recruitment of ERalpha to the promoter of an APPct target gene (KAI1) was demonstrated. Our studies reveal a novel mechanism of estrogen action, which may explain the well-known neuroprotective functions of estrogens as well as the complex role of this female hormone in the pathogenesis of neuronal degeneration diseases.


Asunto(s)
Precursor de Proteína beta-Amiloide/antagonistas & inhibidores , Núcleo Celular/efectos de los fármacos , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal/efectos de los fármacos , Precursor de Proteína beta-Amiloide/genética , Animales , Apoptosis/efectos de los fármacos , Encéfalo/citología , Encéfalo/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Células HeLa , Humanos , Proteína Kangai-1/metabolismo , Ratones , Neuronas/citología , Neuronas/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Transcripción Genética/efectos de los fármacos
15.
Mol Endocrinol ; 23(2): 213-25, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19074551

RESUMEN

FoxO (mammalian forkhead subclass O) proteins are transcription factors acting downstream of the PTEN (phosphatase and tensin homolog deleted on chromosome 10) tumor suppressor. Their activity is negatively regulated by AKT-mediated phosphorylation. Our previous studies showed that the transcriptional activity of the androgen receptor (AR) was inhibited by PTEN in an AKT-sensitive manner. Here, we report the repression of the activity of the full-length AR and its N-terminal domain by FoxO1 and the participation of FoxO1 in AR inhibition by PTEN. Ectopic expression of active FoxO1 decreased the transcriptional activity of AR as well as androgen-induced cell proliferation and production of prostate-specific antigen. FoxO1 knock down by RNA interference increased the transcriptional activity of the AR in PTEN-intact cells and relieved its inhibition by ectopic PTEN in PTEN-null cells. Mutational analysis revealed that FoxO1 fragment 150-655, which contains the forkhead box and C-terminal activation domain, was required for AR inhibition. Mammalian two-hybrid and glutathione-S-transferase pull-down assays demonstrated that the inhibition of AR activity by PTEN through FoxO1 involved the interference of androgen-induced interaction of the N- and C-termini of the AR and the recruitment of the p160 coactivators to its N terminus and to the androgen response elements of natural AR target genes. These studies reveal new mechanisms for the inhibition of AR activity by PTEN-FoxO axis and establish FoxO proteins as important nuclear factors that mediate the mutual antagonism between AR and PTEN tumor suppressor in prostate cancer cells.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Fosfohidrolasa PTEN/metabolismo , Receptores Androgénicos/metabolismo , Animales , Línea Celular , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Humanos , Fosfohidrolasa PTEN/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Interferencia de ARN , Receptores Androgénicos/química , Receptores Androgénicos/genética , Transcripción Genética , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
16.
Chem Commun (Camb) ; 56(83): 12566-12569, 2020 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-32940264

RESUMEN

A stable artificial solid electrolyte interphase (ASEI) containing phosphazene and perfluoroalkoxy groups was designed to protect Li anodes. The ASEI with high ionic conductivity and mechanical robustness successfully suppressed the growth of Li dendrites, significantly enhancing the electrochemical performance of the Li-O2 batteries.

17.
Cells ; 9(10)2020 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-33020410

RESUMEN

We have previously discovered that HDAC6 regulates the DNA damage response (DDR) via modulating the homeostasis of a DNA mismatch repair protein, MSH2, through HDAC6's ubiquitin E3 ligase activity. Here, we have reported HDAC6's second potential E3 ligase substrate, a critical cell cycle checkpoint protein, Chk1. We have found that HDAC6 and Chk1 directly interact, and that HDAC6 ubiquitinates Chk1 in vivo and in vitro. Specifically, HDAC6 interacts with Chk1 via the DAC1 domain, which contains its ubiquitin E3 ligase activity. During the cell cycle, Chk1 protein levels fluctuate, peaking at the G2 phase, subsequently resolving via the ubiquitin-proteasome pathway, and thereby allowing cells to progress to the M phase. However, in HDAC6 knockdown non-small cell lung cancer (NSCLC) cells, Chk1 is constitutively active and fails to resolve post-ionizing radiation (IR), and this enhanced Chk1 activity leads to preferential G2 arrest in HDAC6 knockdown cells accompanied by a reduction in colony formation capacity and viability. Depletion or pharmacological inhibition of Chk1 in HDAC6 knockdown cells reverses this radiosensitive phenotype, suggesting that the radiosensitivity of HDAC6 knockdown cells is dependent on increased Chk1 kinase activity. Overall, our results highlight a novel mechanism of Chk1 regulation at the post-translational level, and a possible strategy for sensitizing NSCLC to radiation via inhibiting HDAC6's E3 ligase activity.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Histona Desacetilasa 6/metabolismo , Neoplasias Pulmonares/genética , Tolerancia a Radiación/fisiología , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Noqueados , Transfección
18.
Cell Death Dis ; 11(5): 328, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32382008

RESUMEN

Ubiquitin-specific peptidase 10 (USP10) stabilizes both tumor suppressors and oncogenes in a context-dependent manner. However, the nature of USP10's role in non-small cell lung cancer (NSCLC) remains unclear. By analyzing The Cancer Genome Atlas (TCGA) database, we have shown that high levels of USP10 are associated with poor overall survival in NSCLC with mutant p53, but not with wild-type p53. Consistently, genetic depletion or pharmacological inhibition of USP10 dramatically reduces the growth of lung cancer xenografts lacking wild-type p53 and sensitizes them to cisplatin. Mechanistically, USP10 interacts with, deubiquitinates, and stabilizes oncogenic protein histone deacetylase 6 (HDAC6). Furthermore, reintroducing either USP10 or HDAC6 into a USP10-knockdown NSCLC H1299 cell line with null-p53 renders cisplatin resistance. This result suggests the existence of a "USP10-HDAC6-cisplatin resistance" axis. Clinically, we have found a positive correlation between USP10 and HDAC6 expression in a cohort of NSCLC patient samples. Moreover, we have shown that high levels of USP10 mRNA correlate with poor overall survival in a cohort of advanced NSCLC patients who received platinum-based chemotherapy. Overall, our studies suggest that USP10 could be a potential biomarker for predicting patient response to platinum, and that targeting USP10 could sensitize lung cancer patients lacking wild-type p53 to platinum-based therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos , Histona Desacetilasa 6/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Proteína p53 Supresora de Tumor/deficiencia , Ubiquitina Tiolesterasa/metabolismo , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones SCID , Mutación/genética , Neoplasias Ováricas/patología , Platino (Metal)/farmacología , Unión Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Immunol Sci ; 2(3): 59-68, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30854521

RESUMEN

The oncogene HDAC6 controls numerous cell processes that are related to tumorigenesis and metastasis, and has recently arisen as a target to treat malignancies. The ERK cascade is a classic pathway driving oncogenesis, and the components of this pathway are either highly mutated in cancers or are vital in cancer's pathological activity. The interactions between these important components of tumor proliferation have been examined, and our research has demonstrated that they regulate each other as evidenced by different posttranslational modifications. Preclinical evidence also supports clinical trials cotargeting these two pathways, which may provide better efficacy than single treatment. Furthermore, HDAC6 and ERK both participate in the regulation of T cell maturation and may have implications on the functions of immune cells. This leads to the possibility of connecting HDAC6 and ERK to immunotherapy. In this review, we summarize the published studies about the interaction of HDAC6 and ERK cascade and their relationship to cancers. We also include the association of HDAC6 and ERK to immune system and discuss the plausibility of linking these to immunotherapy.

20.
Mol Cell Biol ; 22(16): 5835-45, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12138194

RESUMEN

Estrogen receptors are phosphoproteins which can be activated by ligands, kinase activators, or phosphatase inhibitors. Our previous study showed that p38 mitogen-activated protein kinase was involved in estrogen receptor activation by estrogens and MEKK1. Here, we report estrogen receptor-dependent p38 activation by estrogens in endometrial adenocarcinoma cells and in vitro and in vivo phosphorylation of the estrogen receptor alpha mediated through p38. The phosphorylation site was identified as threonine-311 (Thr(311)), located in helix 1 of the hormone-binding domain. The mutation of threonine-311 to alanine did not affect estrogen binding of the receptor but compromised its interaction with coactivators. Suppression of p38 activity or mutation of the site inhibited the estrogen-induced receptor nuclear localization as well as its transcriptional activation by estrogens and MEKK1. The inhibition of the p38 signal pathway by a specific chemical inhibitor blocked the biological activities of estrogens in regulating endogenous gene expression as well as endometrial cancer cell growth. Our studies demonstrate the role of estrogen receptor phosphorylation induced by the natural ligand in estrogen receptor's cellular distribution and its significant contribution to the growth-stimulating activity of estrogens in endometrial cancer cells.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Estradiol/farmacología , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores de Estrógenos/metabolismo , Tamoxifeno/análogos & derivados , Transporte Activo de Núcleo Celular/fisiología , Adenocarcinoma/fisiopatología , Secuencia de Aminoácidos , Animales , Neoplasias Endometriales/fisiopatología , Activación Enzimática , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno , Ácidos Grasos Insaturados/farmacología , Femenino , Humanos , Ligandos , Datos de Secuencia Molecular , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Señales de Clasificación de Proteína , Receptores de Estrógenos/genética , Transducción de Señal/fisiología , Tamoxifeno/farmacología , Treonina/metabolismo , Transcripción Genética , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA