Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Nucleic Acids Res ; 47(15): 7734-7752, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31350900

RESUMEN

DAXX displays complex biological functions. Remarkably, DAXX overexpression is a common feature in diverse cancers, which correlates with tumorigenesis, disease progression and treatment resistance. Structurally, DAXX is modular with an N-terminal helical bundle, a docking site for many DAXX interactors (e.g. p53 and ATRX). DAXX's central region folds with the H3.3/H4 dimer, providing a H3.3-specific chaperoning function. DAXX has two functionally critical SUMO-interacting motifs. These modules are connected by disordered regions. DAXX's structural features provide a framework for deciphering how DAXX mechanistically imparts its functions and how its activity is regulated. DAXX modulates transcription through binding to transcription factors, epigenetic modifiers, and chromatin remodelers. DAXX's localization in the PML nuclear bodies also plays roles in transcriptional regulation. DAXX-regulated genes are likely important effectors of its biological functions. Deposition of H3.3 and its interactions with epigenetic modifiers are likely key events for DAXX to regulate transcription, DNA repair, and viral infection. Interactions between DAXX and its partners directly impact apoptosis and cell signaling. DAXX's activity is regulated by posttranslational modifications and ubiquitin-dependent degradation. Notably, the tumor suppressor SPOP promotes DAXX degradation in phase-separated droplets. We summarize here our current understanding of DAXX's complex functions with a focus on how it promotes oncogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/genética , Neoplasias/genética , Proteínas Nucleares/genética , Procesamiento Proteico-Postraduccional , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular Tumoral , Cromatina/química , Proteínas Co-Represoras , Humanos , Chaperonas Moleculares , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fosforilación , Unión Proteica , Mapeo de Interacción de Proteínas , Transducción de Señal , Sumoilación , Transcripción Genética , Ubiquitinación
2.
Prostate ; 78(15): 1140-1156, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30009471

RESUMEN

BACKGROUND: Mutations or truncation of the ligand-binding domain (LBD) of androgen receptor (AR) underlie treatment resistance for prostate cancer (PCa). Thus, targeting the AR N-terminal domain (NTD) could overcome such resistance. METHODS: Luciferase reporter assays after transient transfection of various DNA constructs were used to assess effects of E1A proteins on AR-mediated transcription. Immunofluorescence microscopy and subcellular fractionation were applied to assess intracellular protein localization. Immunoprecipitation and mammalian two-hybrid assays were used to detect protein-protein interactions. qRT-PCR was employed to determine RNA levels. Western blotting was used to detect protein expression in cells. Effects of adenoviruses on prostate cancer cell survival were evaluated with CellTiter-Glo assays. RESULTS: Adenovirus 12 E1A (E1A12) binds specifically to the AR. Interestingly, the full-length E1A12 (266 aa) preferentially binds to full-length AR, while the small E1A12 variant (235 aa) interacts more strongly with AR-V7. E1A12 promotes AR nuclear translocation, likely through mediating intramolecular AR NTD-LBD interactions. In the nucleus, AR and E1A12 co-expression in AR-null PCa cells results in E1A12 redistribution from nuclear foci containing CBX4 (also known as Pc2), suggesting a preferential AR-E1A12 interaction over other E1A12 interactors. E1A12 represses AR-mediated transcription in reporter gene assays and endogenous AR target genes such as ATAD2 and MYC in AR-expressing PCa cells. AR-expressing PCa cells are more sensitive to death induced by a recombinant adenovirus expressing E1A12 (Ad-E1A12) than AR-deficient PCa cells, which could be attributed to the increased viral replication promoted by androgen stimulation. Targeting the AR by E1A12 promotes apoptosis in PCa cells that express the full-length AR or C-terminally truncated AR variants. Importantly, inhibition of mTOR signaling that blocks the expression of anti-apoptotic proteins markedly augments Ad-E1A12-induced apoptosis of AR-expressing cells. Mechanistically, Ad-E1A12 infection triggers apoptotic response while activating the PI3K-AKT-mTOR signaling axis; thus, mTOR inhibition enhances apoptosis in AR-expressing PCa cells infected by Ad-E1A12. CONCLUSION: Ad12 E1A inhibits AR-mediated transcription and suppresses PCa cell survival, suggesting that targeting the AR by E1A12 might have therapeutic potential for treating advanced PCa with heightened AR signaling.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Neoplasias de la Próstata/terapia , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Proteínas E1A de Adenovirus/genética , Adenovirus Humanos/genética , Adenovirus Humanos/metabolismo , Línea Celular Tumoral , Supervivencia Celular/fisiología , Células HEK293 , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/virología , Dominios Proteicos , Activación Transcripcional , Transfección
3.
FASEB J ; 28(10): 4265-79, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24948597

RESUMEN

Histone deacetylases (HDACs) that deacetylate histone and nonhistone proteins play crucial roles in a variety of cellular processes. The overexpression of HDACs is reported in many cancer types and is directly linked to accelerated cell proliferation and survival. However, little is known about how HDAC expression is regulated in cancer cells. In this study, we found that HDAC1 and HDAC2 promoters are regulated through collaborative binding of transcription factors Sp1/Sp3 and epigenetic modulators, including histone H3K4 methyltransferase SET1 and histone acetyltransferase p300, whose levels are also elevated in colon cancer cell lines and patient samples. Interestingly, Sp1 and Sp3 differentially regulate HDAC1 and HDAC2 promoter activity. In addition, Sp1/Sp3 recruits SET1 and p300 to the promoters. SET1 knockdown (KD) results in a loss of the H3K4 trimethylation mark at the promoters, as well as destabilizes p300 at the promoters. Conversely, p300 also influences SET1 recruitment and H3K4me3 level, indicating a crosstalk between p300 and SET1. Further, SET1 KD reduces Sp1 binding to the HDAC1 promoter through the increase of Sp1 acetylation. These results indicate that interactions among transcription factors and epigenetic modulators orchestrate the activation of HDAC1 and HDAC2 promoter activity in colon cancer cells.


Asunto(s)
Neoplasias del Colon/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Células HCT116 , Células HT29 , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Regiones Promotoras Genéticas , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
4.
Drug Discov Today Technol ; 18: 24-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26723889

RESUMEN

Histone deacetylases (HDACs) are promising drug targets for treating cancer, neurologic, inflammatory and metabolic diseases. Four small molecule inhibitors of HDACs have gained regulatory approval for treating lymphomas and multiple myelomas. Highly sensitive in vitro and cell-based profiling technologies have been developed to discover HDAC inhibitors (HDACi) and characterize their inhibitory potency, target-binding specificity and kinetics. In particular, proteomic profiling can define the specificity of an inhibitor at a single residue resolution. Chemoproteomic profiling can determine the potency, specificity and binding kinetics of an inhibitor on a specific HDAC complex in cell extracts. As inhibitors with new chemical scaffolds are of particular interest to improve HDAC isoform-specificity and pharmaceutical properties, effective profiling technologies will continue to have important utility. Here we briefly review recent developments of HDAC inhibitor profiling technologies and discuss distinct features of various technologies.


Asunto(s)
Descubrimiento de Drogas/métodos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Proteómica/métodos , Bibliotecas de Moléculas Pequeñas/farmacología , Tecnología Farmacéutica/métodos , Animales , Ensayos Analíticos de Alto Rendimiento , Inhibidores de Histona Desacetilasas/química , Humanos , Isomerismo , Unión Proteica , Bibliotecas de Moléculas Pequeñas/química
5.
bioRxiv ; 2024 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-39253438

RESUMEN

Proteolysis targeting chimeras (PROTACs) are an emerging targeted cancer therapy approach, but wide-spread clinical use of PROTAC is limited due to poor cell targeting and penetration, and instability in vivo. To overcome such issues and enhance the in vivo efficacy of PROTAC drugs, microfluidic droplet-based electroporation (µDES) was developed as a novel extracellular vesicle (EVs) transfection system, which enables the high-efficient PROTAC loading and effective delivery in vivo. Our previously developed YX968 PROTAC drug had shown the selectively degradation of HDAC3 and 8, which effectively suppresses the growth of breast tumor cell lines, including MDA-MB-231 triple negative breast cancer (TNBC) line, via dual degradation without provoking a global histone hyperacetylation. In this study, we demonstrated that µDES-based PROTAC loading in EVs significantly enhanced therapeutic function of PROTAC drug in vivo in the TNBC breast tumor mouse model. NSG mice with pre-established MDA-MB-231 tumors and treated with intraperitoneal injection of EVs for tumor inhibition study, which showed significantly higher HDAC 3 and 8 degradation efficiency and tumor inhibition than PROTAC only group. The liver, spleen, kidney, lung, heart, and brain were collected for safety testing, which exhibited improved toxicity. The EV delivery of PROTAC drug enhances drug stability and bioavailability in vivo, transportability, and drug targeting ability, which fills an important gap in current development of PROTAC therapeutic functionality in vivo and clinical translation. This novel EV-based drug transfection and delivery strategy could be applicable to various therapeutics for enhancing in vivo delivery, efficacy, and safety.

6.
J Med Chem ; 67(15): 12784-12806, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38949959

RESUMEN

HDAC8 plays crucial roles in biological processes, from gene regulation to cell motility, making it a highly desirable target for therapeutic intervention. HDAC8 also has deacetylase-independent activity which cannot be blocked by a conventional inhibitor. In this study, we report the discovery of YX862, a highly potent and selective hydrazide-based HDAC8-proteolysis targeting chimera (PROTAC) degrader. The selectivity is achieved through rational design of the warhead to spare HDAC3 activity from the previous HDAC3/8 dual degrader YX968. We demonstrate that the degradation of HDAC8 by YX862 increases acetylation levels of its nonhistone substrates such as SMC3 without significantly triggering histone PTM, supporting HDAC8's major role in nonhistone PTM regulation. YX862 exhibits promising on-target antiproliferative activity against DLBCL cells with higher potency than the HDAC8 selective inhibitor PCI-34051. As a selective HDAC8 degrader that avoids pan-HDAC inhibition, YX862 represents a valuable tool for exploring the biological and therapeutic potential of HDAC8.


Asunto(s)
Inhibidores de Histona Desacetilasas , Histona Desacetilasas , Proteínas Represoras , Humanos , Histona Desacetilasas/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/síntesis química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Estructura-Actividad , Descubrimiento de Drogas , Proteolisis/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Acetilación
7.
Cell Chem Biol ; 30(11): 1421-1435.e12, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37572669

RESUMEN

HDAC3 and HDAC8 have critical biological functions and represent highly sought-after therapeutic targets. Because histone deacetylases (HDACs) have a very conserved catalytic domain, developing isozyme-selective inhibitors remains challenging. HDAC3/8 also have deacetylase-independent activity, which cannot be blocked by conventional enzymatic inhibitors. Proteolysis-targeting chimeras (PROTACs) can selectively degrade a target enzyme, abolishing both enzymatic and scaffolding function. Here, we report a novel HDAC3/8 dual degrader YX968 that induces highly potent, rapid, and selective degradation of both HDAC3/8 without triggering pan-HDAC inhibitory effects. Unbiased quantitative proteomic experiments confirmed its high selectivity. HDAC3/8 degradation by YX968 does not induce histone hyperacetylation and broad transcriptomic perturbation. Thus, histone hyperacetylation may be a major factor for altering transcription. YX968 promotes apoptosis and kills cancer cells with a high potency in vitro. YX968 thus represents a new probe for dissecting the complex biological functions of HDAC3/8.


Asunto(s)
Inhibidores de Histona Desacetilasas , Histonas , Histonas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Acetilación , Proteómica , Procesamiento Proteico-Postraduccional
8.
Nat Commun ; 14(1): 1927, 2023 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-37045819

RESUMEN

Cancer cells exhibit elevated lipid synthesis. In breast and other cancer types, genes involved in lipid production are highly upregulated, but the mechanisms that control their expression remain poorly understood. Using integrated transcriptomic, lipidomic, and molecular studies, here we report that DAXX is a regulator of oncogenic lipogenesis. DAXX depletion attenuates, while its overexpression enhances, lipogenic gene expression, lipogenesis, and tumor growth. Mechanistically, DAXX interacts with SREBP1 and SREBP2 and activates SREBP-mediated transcription. DAXX associates with lipogenic gene promoters through SREBPs. Underscoring the critical roles for the DAXX-SREBP interaction for lipogenesis, SREBP2 knockdown attenuates tumor growth in cells with DAXX overexpression, and DAXX mutants unable to bind SREBP1/2 have weakened activity in promoting lipogenesis and tumor growth. Remarkably, a DAXX mutant deficient of SUMO-binding fails to activate SREBP1/2 and lipogenesis due to impaired SREBP binding and chromatin recruitment and is defective of stimulating tumorigenesis. Hence, DAXX's SUMO-binding activity is critical to oncogenic lipogenesis. Notably, a peptide corresponding to DAXX's C-terminal SUMO-interacting motif (SIM2) is cell-membrane permeable, disrupts the DAXX-SREBP1/2 interactions, and inhibits lipogenesis and tumor growth. These results establish DAXX as a regulator of lipogenesis and a potential therapeutic target for cancer therapy.


Asunto(s)
Lipogénesis , Neoplasias , Carcinogénesis/genética , Transformación Celular Neoplásica , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Lípidos , Lipogénesis/genética , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Ratones
9.
J Virol ; 85(16): 7976-88, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21680522

RESUMEN

Adenovirus E1B-55K represses p53-mediated transcription. However, the phenotypic consequence of p53 inhibition by E1B-55K for cell cycle regulation and drug sensitivity in tumor cells has not been examined. In HCT116 cells with constitutive E1B-55K expression, the activation of p53 target genes such as the p21, Mdm2, and Puma genes was attenuated, despite markedly elevated p53 protein levels. HCT116 cells with E1B-55K expression displayed a cell cycle profile similar to that of the isogenic HCT116p53(-/-) cells, including unhindered S-phase entry despite DNA damage. Surprisingly, E1B-55K-expressing cells were more sensitive to drug treatment than parental cells. Compared to HCT116 cells, HCT116p53(-/-) cells were more susceptible to both doxorubicin and etoposide, and E1B-55K expression had no effects on drug treatment. E1B-55K expression increased the rate of cell proliferation in HCT116 but not in HCT116p53(-/-) cells. Thus, deregulation of p53-mediated cell cycle control by E1B-55K probably underlies sensitization of HCT116 cells to anticancer drugs. Consistently, E1B-55K expression in A549, A172, and HepG2 cells, all containing wild-type (wt) p53, also enhanced etoposide-induced cytotoxicity, whereas in p53-null H1299 cells, E1B-55K had no effects. We generated several E1B-55K mutants with mutations at positions occupied by the conserved Phe/Trp/His residues. Most of these mutants showed no or reduced binding to p53, although some of them could still stabilize p53, suggesting that binding might not be essential for E1B-55K-induced p53 stabilization. Despite heightened p53 protein levels in cells expressing certain E1B-55K mutants, p53 activity was largely suppressed. Furthermore, most of these E1B-55K mutants could sensitize HCT116 cells to etoposide and doxorubicin. These results indicate that E1B-55K might have utility for enhancing chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Ciclo Celular , Genes p53 , Proteína p53 Supresora de Tumor/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Proteínas E1B de Adenovirus/genética , Proteínas E1B de Adenovirus/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Etopósido/farmacología , Expresión Génica , Células Hep G2 , Humanos , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Proteínas Virales/metabolismo
10.
Cancers (Basel) ; 13(12)2021 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-34201346

RESUMEN

The CREB-binding protein (CBP) and p300 are two paralogous lysine acetyltransferases (KATs) that were discovered in the 1980s-1990s. Since their discovery, CBP/p300 have emerged as important regulatory proteins due to their ability to acetylate histone and non-histone proteins to modulate transcription. Work in the last 20 years has firmly established CBP/p300 as critical regulators for nuclear hormone signaling pathways, which drive tumor growth in several cancer types. Indeed, CBP/p300 are critical co-activators for the androgen receptor (AR) and estrogen receptor (ER) signaling in prostate and breast cancer, respectively. The AR and ER are stimulated by sex hormones and function as transcription factors to regulate genes involved in cell cycle progression, metabolism, and other cellular functions that contribute to oncogenesis. Recent structural studies of the AR/p300 and ER/p300 complexes have provided critical insights into the mechanism by which p300 interacts with and activates AR- and ER-mediated transcription. Breast and prostate cancer rank the first and forth respectively in cancer diagnoses worldwide and effective treatments are urgently needed. Recent efforts have identified specific and potent CBP/p300 inhibitors that target the acetyltransferase activity and the acetytllysine-binding bromodomain (BD) of CBP/p300. These compounds inhibit AR signaling and tumor growth in prostate cancer. CBP/p300 inhibitors may also be applicable for treating breast and other hormone-dependent cancers. Here we provide an in-depth account of the critical roles of CBP/p300 in regulating the AR and ER signaling pathways and discuss the potential of CBP/p300 inhibitors for treating prostate and breast cancer.

11.
Cancers (Basel) ; 13(11)2021 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-34199844

RESUMEN

Estrogen receptor alpha (ER) is the oncogenic driver for ER+ breast cancer (BC). ER antagonists are the standard-of-care treatment for ER+ BC; however, primary and acquired resistance to these agents is common. CBP and p300 are critical ER co-activators and their acetyltransferase (KAT) domain and acetyl-lysine binding bromodomain (BD) represent tractable drug targets, but whether CBP/p300 inhibitors can effectively suppress ER signaling remains unclear. We report that the CBP/p300 KAT inhibitor A-485 and the BD inhibitor GNE-049 downregulate ER, attenuate estrogen-induced c-Myc and Cyclin D1 expression, and inhibit growth of ER+ BC cells through inducing senescence. Microarray and RNA-seq analysis demonstrates that A-485 or EP300 (encoding p300) knockdown globally inhibits expression of estrogen-regulated genes, confirming that ER inhibition is an on-target effect of A-485. Using ChIP-seq, we report that A-485 suppresses H3K27 acetylation in the enhancers of ER target genes (including MYC and CCND1) and this correlates with their decreased expression, providing a mechanism underlying how CBP/p300 inhibition downregulates ER gene network. Together, our results provide a preclinical proof-of-concept that CBP/p300 represent promising therapeutic targets in ER+ BC for inhibiting ER signaling.

12.
J Vis Exp ; (162)2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32831305

RESUMEN

Lysine acetyltransferases (KATs) catalyze acetylation of lysine residues on histones and other proteins to regulate chromatin dynamics and gene expression. KATs, such as CBP/p300, are under intense investigation as therapeutic targets due to their critical role in tumorigenesis of diverse cancers. The development of novel small molecule inhibitors targeting the histone acetyltransferase (HAT) function of KATs is challenging and requires robust assays that can validate the specificity and potency of potential inhibitors. This article outlines a pipeline of three methods that provide rigorous in vitro validation for novel HAT inhibitors (HATi). These methods include a test tube HAT assay, Chromatin Hyperacetylation Inhibition (ChHAI) assay, and Chromatin Immunoprecipitation-quantitative PCR (ChIP-qPCR). In the HAT assay, recombinant HATs are incubated with histones in a test tube reaction, allowing for acetylation of specific lysine residues on the histone tails. This reaction can be blocked by a HATi and the relative levels of site-specific histone acetylation can be measured via immunoblotting. Inhibitors identified in the HAT assay need to be confirmed in the cellular environment. The ChHAI assay uses immunoblotting to screen for novel HATi that attenuate the robust hyperacetylation of histones induced by a histone deacetylase inhibitor (HDACi). The addition of an HDACi is helpful because basal levels of histone acetylation can be difficult to detect via immunoblotting. The HAT and ChHAI assays measure global changes in histone acetylation, but do not provide information regarding acetylation at specific genomic regions. Therefore, ChIP-qPCR is used to investigate the effects of HATi on histone acetylation levels at gene regulatory elements. This is accomplished through selective immunoprecipitation of histone-DNA complexes and analysis of the purified DNA through qPCR. Together, these three assays allow for the careful validation of the specificity, potency, and mechanism of action of novel HATi.


Asunto(s)
Bioensayo/métodos , Histona Acetiltransferasas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Acetilación/efectos de los fármacos , Ácidos Anacárdicos/farmacología , Ciclina D1/genética , Ciclina D1/metabolismo , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Proteína p300 Asociada a E1A/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Histonas/metabolismo , Humanos , Lisina/metabolismo , Células MCF-7 , Regiones Promotoras Genéticas/genética , Reproducibilidad de los Resultados , Sonicación
13.
Chem Commun (Camb) ; 56(68): 9866-9869, 2020 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-32840532

RESUMEN

Histone deacetylases (HDACs) are validated drug targets for cancer treatment. Increased HDAC isozyme selectivity and novel strategies to inhibit HDAC activity could lead to safer and more effective drug candidates. Nonetheless, it is quite challenging to develop isozyme-specific HDACi due to the highly conserved catalytic domain. We discovered XZ9002, a first-in-class HDAC3-specific PROTAC that potently degraded HDAC3. Importantly, XZ9002 is more effective to inhibit cancer cell proliferation than its proteolysis-inactive counterpart, suggesting HDAC3 degradation is a novel and promising anticancer approach.


Asunto(s)
Histona Desacetilasas/metabolismo , Proteolisis , Benzamidas/química , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/química , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
14.
Virology ; 521: 118-128, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29906705

RESUMEN

Adenovirus E1B 55-kilodalton (E1B-55K) mediated DAXX degradation represents a potential mechanism by which E1B-55K sensitizes cancer cells to chemotherapy. Here we report the effects of E1B-55K-mediated DAXX degradation in chemoresistant ovarian cancer cells on response to chemotherapy. Cells with E1B-55K expression were more sensitive to cisplatin than cells without E1B-55K expression. In vivo C13* xenograft studies showed that the combination of cisplatin and E1B-55K was markedly more effective to slow tumor growth and to confer prolonged survival of tumor-bearing mice than either cisplatin or E1B-55K alone. Our studies show that DAXX plays an important role in cisplatin resistance in ovarian cancer, and strategies that promote DAXX degradation such as E1B-55K expression in combination with cisplatin can overcome drug resistance and improve responses to standard chemotherapy. These results also indicate that E1B-55K might be a novel agent for enhancing treatment responses for cisplatin-resistant ovarian cancer.


Asunto(s)
Proteínas E1B de Adenovirus/genética , Antineoplásicos/farmacología , Proteínas Portadoras/metabolismo , Cisplatino/farmacología , Resistencia a Antineoplásicos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Adenovirus Humanos/genética , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Proteínas Co-Represoras , Femenino , Xenoinjertos , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Desnudos , Chaperonas Moleculares , Proteínas Nucleares/genética , Neoplasias Ováricas/genética , Adhesión en Parafina
15.
Sci Rep ; 8(1): 4879, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29559707

RESUMEN

Renal Cell Carcinoma (RCC) is one of the most lethal urological cancers worldwide. The disease does not present early clinical symptoms and is commonly diagnosed at an advanced stage. Limited molecular drivers have been identified for RCC, resulting in the lack of effective treatment for patients with progressive disease. Ubiquitous ßArrestin2 (ßArr2) is well established for its function in the desensitization and trafficking of G protein-coupled receptors. More recently, ßArr2 has been implicated in the regulation of fundamental cellular functions, including proliferation and invasion. We used bioinformatic and genetic approaches to determine role of ßArr2 in RCC tumor growth. Analysis of published human datasets shows that ARRB2 (gene encoding ßArr2) expression is increased in RCC tumor compared to normal tissue and that high levels of ARRB2 correlate with worse patient survival. Experimentally, we show that knockout of ARRB2 decreases rate of RCC cell proliferation and migration in vitro and xenograft tumor growth in animals. Mechanistically, ßArr2 regulates c-Src activity, Cyclin A expression and cell cycle progression that are involved in tumor growth. These results show that ßArr2 is a critical regulator of RCC tumor growth and suggest its utility as a potential marker and drug target to treat advanced disease.


Asunto(s)
Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Arrestina beta 2/fisiología , Animales , Proteína Tirosina Quinasa CSK , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Biología Computacional/métodos , Xenoinjertos , Humanos , Neoplasias Renales/patología , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , ARN Interferente Pequeño/uso terapéutico , Transducción de Señal , Arrestina beta 2/genética , Arrestina beta 2/metabolismo , Familia-src Quinasas/uso terapéutico
16.
Cell Chem Biol ; 24(2): 128-129, 2017 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-28132895

RESUMEN

Bromodomains are protein domains that serve as "readers" of acetylated lysine marks and mediate DNA-templated processes. In this issue of Cell Chemical Biology, Olzscha et al. (2017) report that the CBP/p300 bromodomains mediate the formation of amyloid-like aggregates and that inhibitors specific to these bromodomains reduce the degree of protein aggregation and mitigate HDAC inhibitor-induced cytotoxicity.


Asunto(s)
Lisina , Agregado de Proteínas , Acetilación , Modelos Moleculares , Dominios Proteicos
17.
Sci Rep ; 6: 37213, 2016 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-27849011

RESUMEN

Cancer cells of epithelial and mesenchymal phenotypes exhibit different sensitivities to apoptosis stimuli, but the mechanisms underlying this phenomenon remain partly understood. We constructed a novel recombinant adenovirus expressing Ad12 E1A (Ad-E1A12) that can strongly induce apoptosis. Ad-E1A12 infection of epithelial cancer cells displayed dramatic detachment and apoptosis, whereas cancer cells of mesenchymal phenotypes with metastatic propensity were markedly more resistant to this virus. Notably, forced detachment of epithelial cells did not further sensitize them to Ad-E1A12-induced apoptosis, suggesting that cell detachment is a consequence rather than the cause of Ad-E1A12-induced apoptosis. Ad-E1A12 increased phosphorylation of AKT1 and ribosomal protein S6 through independent mechanisms in different cell types. Ad-E1A12-induced AKT1 phosphorylation was PI3K-dependent in epithelial cancer cells, and mTOR-dependent in mesenchymal cancer cells. Epithelial cancer cells upon Ad-E1A12-induced detachment could not sustain AKT activation due to AKT1 degradation, but AKT1 activation was maintained in mesenchymal cancer cells. Expression of epithelial cell-restricted miR-200 family in mesenchymal cells limited mTOR signaling and sensitized them to Ad-E1A12-induced cell killing. Thus, epithelial cancer cells rely on the canonical PI3K-AKT signaling pathway for survival, while mesenchymal cancer cells deploy the PI3K-independent mTORC2-AKT axis in response to strong death stimuli.


Asunto(s)
Adenoviridae/fisiología , Apoptosis/fisiología , Neoplasias/virología , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto , Células A549 , Adenoviridae/genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Células HCT116 , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo
18.
Oncogene ; 22(51): 8316-29, 2003 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-14614455

RESUMEN

The tumor suppressor p53-related p73 shares significant amino-acid sequence identity with p53. Like p53, p73 recognizes canonical p53 DNA-binding sites and activates p53-responsive target genes and induces apoptosis. Moreover, transcription coactivator p300/CBP binds to and coactivates with both p53 and p73 in stimulating the expression of their target genes. Here, we report that coactivator PCAF binds to p73. The N-terminal transactivation domain (TAD) and the conserved oligomerization domain (OD) of p73 are both required for its interaction with PCAF. Conversely, PCAF's HAT-domain is required for and both the N-terminal region and Bromo domain enhance binding of PCAF to p73. Significantly, PCAF stimulates p73-mediated transactivation, and binding of PCAF to p73 is necessary for p73's transactivation activity. PCAF-specific siRNA dramatically reduces p73-mediated transactivation. Stimulation of p73-mediated transactivation by PCAF requires the HAT domain of PCAF and the p53-binding site within the p21 promoter. In vivo, coexpression of wild-type, but not HAT-deficient PCAF with p73beta markedly increases p21 expression. Furthermore, cotransfection of PCAF and p73 leads to increased apoptosis and reduced colony formation. Collectively, these data suggest that p73 recruit PCAF to specific promoters to activate the transcription of p73 target genes.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas Nucleares/fisiología , Transactivadores/fisiología , Activación Transcripcional/fisiología , Apoptosis , Secuencia de Bases , Línea Celular Tumoral , Genes Supresores de Tumor , Humanos , ARN , Proteína Tumoral p73 , Proteínas Supresoras de Tumor , Técnicas del Sistema de Dos Híbridos
19.
Methods Mol Biol ; 1238: 539-48, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25421679

RESUMEN

Lysine acetyltransferases (KATs) acetylate various proteins including histones, transcription factors, metabolic enzymes, and other cellular substrates. Protein acetylation significantly impacts protein stability and function. Certain KATs such as p300 (KAT3B) are overexpressed in cancer cells and are linked to tumor progression and drug resistance. Thus, pharmacologic inhibition of KATs represents a new strategy for cancer therapy. Quantitative biochemical assays of KAT enzymatic activity have been developed and adapted for high-throughput screens of small-molecule compounds to discover specific KAT inhibitors. Such compounds are useful probes for understanding the cellular functions of these critical enzymes and importantly, they may be further developed as anticancer therapeutics. Here we describe a fluorescence-based KAT activity assay and cell-based validation of KAT inhibition by small-molecule compounds.


Asunto(s)
Descubrimiento de Drogas/métodos , Inhibidores Enzimáticos/farmacología , Lisina/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Secuencia de Aminoácidos , Pruebas de Enzimas , Células HCT116 , Humanos , Datos de Secuencia Molecular , Espectrometría de Fluorescencia , Factores de Transcripción p300-CBP/metabolismo
20.
Genom Data ; 5: 257-259, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26217556

RESUMEN

Histone deacetylase (HDAC) inhibitors (HDIs) have therapeutic potentials for treating cancer and other diseases. Modulation of gene expression by HDIs is a major mechanism underlying their therapeutic effects. A novel class of HDIs with a previously undescribed benzoylhydrazide scaffold has been discovered through a high throughput screening campaign. Using microarray profiling of gene expression, we have previously demonstrated that treatment of breast cancer cells with a lead benzoylhydrazide HDI UF010 results in cell cycle arrest and apoptosis, likely through activation of tumor suppression pathways with concurrent inhibition of oncogenic pathways. In this brief report, we show methodological and analytical details and discuss additional pathways such as immune signaling that are affected by UF010. Raw and processed data from the microarray were deposited in NCBI's Gene Expression Omnibus (GEO) database under the accession number: GSE56823.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA