Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
NAR Cancer ; 5(3): zcad039, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37519629

RESUMEN

CCNE1 amplification is a common alteration in high-grade serous ovarian cancer and occurs in 15-20% of these tumors. These amplifications are mutually exclusive with homologous recombination deficiency, and, as they have intact homologous recombination, are intrinsically resistant to poly (ADP-ribose) polymerase inhibitors or chemotherapy agents. Understanding the molecular mechanisms that lead to this mutual exclusivity may reveal therapeutic vulnerabilities that could be leveraged in the clinic in this still underserved patient population. Here, we demonstrate that CCNE1-amplified high-grade serous ovarian cancer cells rely on homologous recombination to repair collapsed replication forks. Cyclin-dependent kinase 2, the canonical partner of cyclin E1, uniquely regulates homologous recombination in this genetic context, and as such cyclin-dependent kinase 2 inhibition synergizes with DNA damaging agents in vitro and in vivo. We demonstrate that combining a selective cyclin-dependent kinase 2 inhibitor with a DNA damaging agent could be a powerful tool in the clinic for high-grade serous ovarian cancer.

2.
SLAS Discov ; 25(4): 361-371, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31585521

RESUMEN

A variety of covalent modifications of RNA have been identified and demonstrated to affect RNA processing, stability, and translation. Methylation of adenosine at the N6 position (m6A) in messenger RNA (mRNA) is currently the most well-studied RNA modification and is catalyzed by the RNA methyltransferase complex METTL3/METTL14. Once generated, m6A can modulate mRNA splicing, export, localization, degradation, and translation. Although potent and selective inhibitors exist for several members of the Type I S-adenosylmethionine (SAM)-dependent methyltransferase family, no inhibitors have been reported for METTL3/METTL14 to date. To facilitate drug discovery efforts, a sensitive and robust mass spectrometry-based assay for METTL3/METTL14 using self-assembled monolayer desorption/ionization (SAMDI) technology has been developed. The assay uses an 11-nucleotide single-stranded RNA compared to a previously reported 27-nucleotide substrate. IC50 values of mechanism-based inhibitors S-adenosylhomocysteine (SAH) and sinefungin (SFG) are comparable between the SAMDI and radiometric assays that use the same substrate. This work demonstrates that SAMDI technology is amenable to RNA substrates and can be used for high-throughput screening and compound characterization for RNA-modifying enzymes.


Asunto(s)
Espectrometría de Masas/métodos , Metiltransferasas/genética , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Adenosina/análogos & derivados , Adenosina/genética , Adenosina/farmacología , Descubrimiento de Drogas/tendencias , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Metilación/efectos de los fármacos , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/genética , Procesamiento Postranscripcional del ARN/genética , Estabilidad del ARN/efectos de los fármacos , Estabilidad del ARN/genética , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , S-Adenosilhomocisteína/farmacología
3.
Cancer Cell ; 36(1): 100-114.e25, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31257072

RESUMEN

Type I protein arginine methyltransferases (PRMTs) catalyze asymmetric dimethylation of arginines on proteins. Type I PRMTs and their substrates have been implicated in human cancers, suggesting inhibition of type I PRMTs may offer a therapeutic approach for oncology. The current report describes GSK3368715 (EPZ019997), a potent, reversible type I PRMT inhibitor with anti-tumor effects in human cancer models. Inhibition of PRMT5, the predominant type II PRMT, produces synergistic cancer cell growth inhibition when combined with GSK3368715. Interestingly, deletion of the methylthioadenosine phosphorylase gene (MTAP) results in accumulation of the metabolite 2-methylthioadenosine, an endogenous inhibitor of PRMT5, and correlates with sensitivity to GSK3368715 in cell lines. These data provide rationale to explore MTAP status as a biomarker strategy for patient selection.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Purina-Nucleósido Fosforilasa/deficiencia , Empalme Alternativo , Antineoplásicos/química , Biomarcadores , Línea Celular Tumoral , Sinergismo Farmacológico , Inhibidores Enzimáticos/química , Humanos , Metilación , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Unión Proteica , Proteína-Arginina N-Metiltransferasas/química , Especificidad por Sustrato
4.
PLoS One ; 13(6): e0197372, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29856759

RESUMEN

A key challenge in the development of precision medicine is defining the phenotypic consequences of pharmacological modulation of specific target macromolecules. To address this issue, a variety of genetic, molecular and chemical tools can be used. All of these approaches can produce misleading results if the specificity of the tools is not well understood and the proper controls are not performed. In this paper we illustrate these general themes by providing detailed studies of small molecule inhibitors of the enzymatic activity of two members of the SMYD branch of the protein lysine methyltransferases, SMYD2 and SMYD3. We show that tool compounds as well as CRISPR/Cas9 fail to reproduce many of the cell proliferation findings associated with SMYD2 and SMYD3 inhibition previously obtained with RNAi based approaches and with early stage chemical probes.


Asunto(s)
Adenocarcinoma del Pulmón/tratamiento farmacológico , Carcinogénesis/genética , N-Metiltransferasa de Histona-Lisina/genética , Células A549 , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Sistemas CRISPR-Cas , Carcinogénesis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/química , Humanos , Metilación/efectos de los fármacos , Metiltransferasas/antagonistas & inhibidores , Interferencia de ARN , Bibliotecas de Moléculas Pequeñas/farmacología
5.
Nat Rev Drug Discov ; 17(6): 435-453, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29773918

RESUMEN

All major biological macromolecules (DNA, RNA, proteins and lipids) undergo enzyme-catalysed covalent modifications that impact their structure, function and stability. A variety of covalent modifications of RNA have been identified and demonstrated to affect RNA stability and translation to proteins; these mechanisms of translational control have been termed epitranscriptomics. Emerging data suggest that some epitranscriptomic mechanisms are altered in human cancers as well as other human diseases. In this Review, we examine the current understanding of RNA modifications with a focus on mRNA methylation, highlight their possible roles in specific cancer indications and discuss the emerging potential of RNA-modifying proteins as therapeutic targets.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , ARN/genética , Animales , Humanos , Metilación/efectos de los fármacos , ARN Mensajero/genética
6.
Lancet Oncol ; 19(5): 649-659, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29650362

RESUMEN

BACKGROUND: Activating enhancer of zeste homolog 2 (EZH2) mutations or aberrations of the switch/sucrose non-fermentable (SWI/SNF) complex (eg, mutations or deletions of the subunits INI1 or SMARCA4) can lead to aberrant histone methylation, oncogenic transformation, and a proliferative dependency on EZH2 activity. In this first-in-human study, we aimed to investigate the safety, clinical activity, pharmacokinetics, and pharmacodynamics of tazemetostat, a first-in-class selective inhibitor of EZH2. METHODS: We did an open-label, multicentre, dose-escalation, phase 1 study using a 3 + 3 design with planned cohort expansion at the two highest doses below the maximally tolerated dose. The study was done at two centres in France: Institut Gustave Roussy (Villejuif, Val de Marne) and Institut Bergonié (Bordeaux, Gironde). Eligible patients had relapsed or refractory B-cell non-Hodgkin lymphoma or an advanced solid tumour and were older than 18 years, with Eastern Cooperative Oncology Group performance status of 0 or 1, and adequate end-organ function. Tazemetostat was administered orally from 100 mg twice daily to 1600 mg twice daily in 28-day cycles. The primary endpoint was to establish the maximum tolerated dose or recommended phase 2 dose of tazemetostat, as determined by dose-limiting toxicities, laboratory values, and other safety or pharmacokinetic measures in cycle one according to local investigator assessment. Safety was assessed in patients who received at least one dose of tazemetostat; antitumour activity was assessed in the intention-to-treat population. This study is registered with ClinicalTrials.gov, number NCT01897571. The phase 1 part of the study is complete, and phase 2 is ongoing. FINDINGS: Between June 13, 2013, and Sept 21, 2016, 64 patients (21 with B-cell non-Hodgkin lymphoma, and 43 with advanced solid tumours) received doses of tazemetostat. The most common treatment-related adverse events, regardless of attribution, were asthenia (21 [33%] of 64 treatment-related events), anaemia (nine [14%]), anorexia (four [6%]), muscle spasms (nine [14%]), nausea (13 [20%]), and vomiting (six [9%]), usually grade 1 or 2 in severity. A single dose-limiting toxicity of grade 4 thrombocytopenia was identified at the highest dose of 1600 mg twice daily. No treatment-related deaths occurred; seven (11%) patients had non-treatment-related deaths (one at 200 mg twice daily, four at 400 mg twice daily, and two at 1600 mg twice daily). The recommended phase 2 dose was determined to be 800 mg twice daily. Durable objective responses, including complete responses, were observed in eight (38%) of 21 patients with B-cell non-Hodgkin lymphoma and two (5%) of 43 patients with solid tumours. INTERPRETATION: Tazemetostat showed a favourable safety profile and antitumour activity in patients with refractory B-cell non-Hodgkin lymphoma and advanced solid tumours, including epithelioid sarcoma. Further clinical investigation of tazemetostat monotherapy is ongoing in phase 2 studies in adults and a phase 1 study for children, which are currently enrolling patients who have B-cell non-Hodgkin lymphoma and INI1-negative or SMARCA4-negative tumours. FUNDING: Epizyme and Eisai.


Asunto(s)
Antineoplásicos/administración & dosificación , Benzamidas/administración & dosificación , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Inhibidores Enzimáticos/administración & dosificación , Linfoma de Células B/tratamiento farmacológico , Piridonas/administración & dosificación , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Benzamidas/efectos adversos , Benzamidas/farmacocinética , Compuestos de Bifenilo , Relación Dosis-Respuesta a Droga , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/farmacocinética , Femenino , Francia , Humanos , Linfoma de Células B/enzimología , Linfoma de Células B/patología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Morfolinas , Piridonas/efectos adversos , Piridonas/farmacocinética , Factores de Tiempo , Resultado del Tratamiento
7.
Sci Rep ; 7(1): 17993, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29269946

RESUMEN

CARM1 is an arginine methyltransferase with diverse histone and non-histone substrates implicated in the regulation of cellular processes including transcriptional co-activation and RNA processing. CARM1 overexpression has been reported in multiple cancer types and has been shown to modulate oncogenic pathways in in vitro studies. Detailed understanding of the mechanism of action of CARM1 in oncogenesis has been limited by a lack of selective tool compounds, particularly for in vivo studies. We describe the identification and characterization of, to our knowledge, the first potent and selective inhibitor of CARM1 that exhibits anti-proliferative effects both in vitro and in vivo and, to our knowledge, the first demonstration of a role for CARM1 in multiple myeloma (MM). EZM2302 (GSK3359088) is an inhibitor of CARM1 enzymatic activity in biochemical assays (IC50 = 6 nM) with broad selectivity against other histone methyltransferases. Treatment of MM cell lines with EZM2302 leads to inhibition of PABP1 and SMB methylation and cell stasis with IC50 values in the nanomolar range. Oral dosing of EZM2302 demonstrates dose-dependent in vivo CARM1 inhibition and anti-tumor activity in an MM xenograft model. EZM2302 is a validated chemical probe suitable for further understanding the biological role CARM1 plays in cancer and other diseases.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas Adaptadoras de Señalización CARD/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Guanilato Ciclasa/antagonistas & inhibidores , Isoxazoles/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Pirimidinas/uso terapéutico , Compuestos de Espiro/uso terapéutico , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacocinética , Humanos , Técnicas In Vitro , Isoxazoles/farmacocinética , Masculino , Ratones , Trasplante de Neoplasias , Pirimidinas/farmacocinética , Ratas Sprague-Dawley , Compuestos de Espiro/farmacocinética
8.
Cell Chem Biol ; 24(9): 1120-1147, 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28938089

RESUMEN

Comprehensive whole-exome sequencing, DNA copy-number determination, and transcriptomic analyses of diverse cancers have greatly expanded our understanding of the biology of many tumor types. In addition to mutations in the common cell-of-origin specific driver mutations, these studies have also revealed a large number of loss-of-function and gain-of-function mutations in chromatin-modifying proteins (CMPs). This has revealed that epigenetic dysregulation is a common feature of most pediatric and adult cancers. Many specific and potent inhibitors have been developed for multiple CMP classes, which have assisted in elucidating the role of epigenetics as well as epigenetic vulnerabilities in these cancer types. Clinical trials with numerous CMP inhibitors are also currently in progress to evaluate the therapeutic potential of epigenetic inhibitors. In this review, we aim to provide a summary of genetic mutations in epigenetic genes and a review of CMP inhibitors suitable for preclinical studies or currently in clinical trials. Additionally, we highlight the CMPs for which potent inhibitors have not been developed and additional research focus should be dedicated.


Asunto(s)
Descubrimiento de Drogas , Epigénesis Genética , Neoplasias/patología , Acetilación/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/antagonistas & inhibidores , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Histonas/metabolismo , Humanos , Metilación/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/genética
9.
Mol Cancer Ther ; 16(11): 2586-2597, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28835384

RESUMEN

The EZH2 small-molecule inhibitor tazemetostat (EPZ-6438) is currently being evaluated in phase II clinical trials for the treatment of non-Hodgkin lymphoma (NHL). We have previously shown that EZH2 inhibitors display an antiproliferative effect in multiple preclinical models of NHL, and that models bearing gain-of-function mutations in EZH2 were consistently more sensitive to EZH2 inhibition than lymphomas with wild-type (WT) EZH2 Here, we demonstrate that cell lines bearing EZH2 mutations show a cytotoxic response, while cell lines with WT-EZH2 show a cytostatic response and only tumor growth inhibition without regression in a xenograft model. Previous work has demonstrated that cotreatment with tazemetostat and glucocorticoid receptor agonists lead to a synergistic antiproliferative effect in both mutant and wild-type backgrounds, which may provide clues to the mechanism of action of EZH2 inhibition in WT-EZH2 models. Multiple agents that inhibit the B-cell receptor pathway (e.g., ibrutinib) were found to have synergistic benefit when combined with tazemetostat in both mutant and WT-EZH2 backgrounds of diffuse large B-cell lymphomas (DLBCL). The relationship between B-cell activation and EZH2 inhibition is consistent with the proposed role of EZH2 in B-cell maturation. To further support this, we observe that cell lines treated with tazemetostat show an increase in the B-cell maturation regulator, PRDM1/BLIMP1, and gene signatures corresponding to more advanced stages of maturation. These findings suggest that EZH2 inhibition in both mutant and wild-type backgrounds leads to increased B-cell maturation and a greater dependence on B-cell activation signaling. Mol Cancer Ther; 16(11); 2586-97. ©2017 AACR.


Asunto(s)
Benzamidas/administración & dosificación , Proteína Potenciadora del Homólogo Zeste 2/genética , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Pirazoles/administración & dosificación , Piridonas/administración & dosificación , Pirimidinas/administración & dosificación , Adenina/análogos & derivados , Animales , Linfocitos B/efectos de los fármacos , Compuestos de Bifenilo , Proliferación Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Sinergismo Farmacológico , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Ratones , Morfolinas , Mutación , Piperidinas , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Cancer Ther ; 16(5): 850-860, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28292935

RESUMEN

The SWI/SNF complex is a major regulator of gene expression and is increasingly thought to play an important role in human cancer, as evidenced by the high frequency of subunit mutations across virtually all cancer types. We previously reported that in preclinical models, malignant rhabdoid tumors, which are deficient in the SWI/SNF core component INI1 (SMARCB1), are selectively killed by inhibitors of the H3K27 histone methyltransferase EZH2. Given the demonstrated antagonistic activities of the SWI/SNF complex and the EZH2-containing PRC2 complex, we investigated whether additional cancers with SWI/SNF mutations are sensitive to selective EZH2 inhibition. It has been recently reported that ovarian cancers with dual loss of the redundant SWI/SNF components SMARCA4 and SMARCA2 are characteristic of a rare rhabdoid-like subtype known as small-cell carcinoma of the ovary hypercalcemic type (SCCOHT). Here, we provide evidence that a subset of commonly used ovarian carcinoma cell lines were misdiagnosed and instead were derived from a SCCOHT tumor. We also demonstrate that tazemetostat, a potent and selective EZH2 inhibitor currently in phase II clinical trials, induces potent antiproliferative and antitumor effects in SCCOHT cell lines and xenografts deficient in both SMARCA2 and SMARCA4. These results exemplify an additional class of rhabdoid-like tumors that are dependent on EZH2 activity for survival. Mol Cancer Ther; 16(5); 850-60. ©2017 AACR.


Asunto(s)
Carcinoma de Células Pequeñas/tratamiento farmacológico , ADN Helicasas/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteínas Nucleares/genética , Neoplasias Ováricas/tratamiento farmacológico , Tumor Rabdoide/tratamiento farmacológico , Factores de Transcripción/genética , Animales , Carcinoma de Células Pequeñas/diagnóstico , Carcinoma de Células Pequeñas/genética , Carcinoma de Células Pequeñas/patología , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/genética , Diagnóstico Diferencial , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Hipercalcemia/diagnóstico , Hipercalcemia/tratamiento farmacológico , Hipercalcemia/genética , Hipercalcemia/patología , Ratones , Mutación , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Tumor Rabdoide/diagnóstico , Tumor Rabdoide/genética , Tumor Rabdoide/patología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Cell ; 31(2): 286-299, 2017 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-28196596

RESUMEN

Small cell lung cancer is initially highly responsive to cisplatin and etoposide but in almost every case becomes rapidly chemoresistant, leading to death within 1 year. We modeled acquired chemoresistance in vivo using a series of patient-derived xenografts to generate paired chemosensitive and chemoresistant cancers. Multiple chemoresistant models demonstrated suppression of SLFN11, a factor implicated in DNA-damage repair deficiency. In vivo silencing of SLFN11 was associated with marked deposition of H3K27me3, a histone modification placed by EZH2, within the gene body of SLFN11, inducing local chromatin condensation and gene silencing. Inclusion of an EZH2 inhibitor with standard cytotoxic therapies prevented emergence of acquired resistance and augmented chemotherapeutic efficacy in both chemosensitive and chemoresistant models of small cell lung cancer.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/fisiología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Nucleares/fisiología , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Animales , Resistencia a Antineoplásicos , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Humanos , Ratones , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Proteína 1 Relacionada con Twist/fisiología
13.
PLoS One ; 11(7): e0158888, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27391784

RESUMEN

The catalytic activities of covalent and ATP-dependent chromatin remodeling are central to regulating the conformational state of chromatin and the resultant transcriptional output. The enzymes that catalyze these activities are often contained within multiprotein complexes in nature. Two such multiprotein complexes, the polycomb repressive complex 2 (PRC2) methyltransferase and the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeler have been reported to act in opposition to each other during development and homeostasis. An imbalance in their activities induced by mutations/deletions in complex members (e.g. SMARCB1) has been suggested to be a pathogenic mechanism in certain human cancers. Here we show that preclinical models of synovial sarcoma-a cancer characterized by functional SMARCB1 loss via its displacement from the SWI/SNF complex through the pathognomonic SS18-SSX fusion protein-display sensitivity to pharmacologic inhibition of EZH2, the catalytic subunit of PRC2. Treatment with tazemetostat, a clinical-stage, selective and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity reverses a subset of synovial sarcoma gene expression and results in concentration-dependent cell growth inhibition and cell death specifically in SS18-SSX fusion-positive cells in vitro. Treatment of mice bearing either a cell line or two patient-derived xenograft models of synovial sarcoma leads to dose-dependent tumor growth inhibition with correlative inhibition of trimethylation levels of the EZH2-specific substrate, lysine 27 on histone H3. These data demonstrate a dependency of SS18-SSX-positive, SMARCB1-deficient synovial sarcomas on EZH2 enzymatic activity and suggests the potential utility of EZH2-targeted drugs in these genetically defined cancers.


Asunto(s)
Antineoplásicos/farmacología , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Sarcoma Sinovial/tratamiento farmacológico , Animales , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Proteína SMARCB1/genética , Proteína SMARCB1/metabolismo , Sarcoma Sinovial/genética , Sarcoma Sinovial/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
ACS Chem Biol ; 11(3): 763-71, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26551522

RESUMEN

Coactivator-associated arginine methyltransferase 1 (CARM1) is a protein arginine N-methyltransferase (PRMT) enzyme that has been implicated in a variety of cancers. CARM1 is known to methylate histone H3 and nonhistone substrates. To date, several crystal structures of CARM1 have been solved, including structures with small molecule inhibitors, but no ternary structures with nucleoside and peptide substrates have been reported. Here, the crystal structures of human CARM1 with the S-adenosylmethione (SAM) mimic sinefungin and three different peptide sequences from histone H3 and PABP1 are presented, with both nonmethylated and singly methylated arginine residues exemplified. This is the first example of multiple substrate sequences solved in a single PRMT enzyme and demonstrates how the CARM1 binding site is capable of accommodating a variety of peptide sequences while maintaining a core binding mode for the unmethylated and monomethylated substrates. Comparison of these with other PRMT enzyme-peptide structures shows hydrogen bonding patterns that may be thematic of these binding sites.


Asunto(s)
Proteína-Arginina N-Metiltransferasas/metabolismo , Sitios de Unión , Línea Celular , Cristalografía por Rayos X , Regulación Enzimológica de la Expresión Génica , Humanos , Modelos Moleculares , Conformación Proteica , Proteína-Arginina N-Metiltransferasas/química , Proteína-Arginina N-Metiltransferasas/genética , Especificidad por Sustrato
15.
ACS Med Chem Lett ; 6(6): 655-9, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-26101569

RESUMEN

A novel aryl pyrazole series of arginine methyltransferase inhibitors has been identified. Synthesis of analogues within this series yielded the first potent, selective, small molecule PRMT6 inhibitor tool compound, EPZ020411. PRMT6 overexpression has been reported in several cancer types suggesting that inhibition of PRMT6 activity may have therapeutic utility. Identification of EPZ020411 provides the field with the first small molecule tool compound for target validation studies. EPZ020411 shows good bioavailability following subcutaneous dosing in rats making it a suitable tool for in vivo studies.

16.
Nat Chem Biol ; 11(6): 432-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25915199

RESUMEN

Protein arginine methyltransferase-5 (PRMT5) is reported to have a role in diverse cellular processes, including tumorigenesis, and its overexpression is observed in cell lines and primary patient samples derived from lymphomas, particularly mantle cell lymphoma (MCL). Here we describe the identification and characterization of a potent and selective inhibitor of PRMT5 with antiproliferative effects in both in vitro and in vivo models of MCL. EPZ015666 (GSK3235025) is an orally available inhibitor of PRMT5 enzymatic activity in biochemical assays with a half-maximal inhibitory concentration (IC50) of 22 nM and broad selectivity against a panel of other histone methyltransferases. Treatment of MCL cell lines with EPZ015666 led to inhibition of SmD3 methylation and cell death, with IC50 values in the nanomolar range. Oral dosing with EPZ015666 demonstrated dose-dependent antitumor activity in multiple MCL xenograft models. EPZ015666 represents a validated chemical probe for further study of PRMT5 biology and arginine methylation in cancer and other diseases.


Asunto(s)
Antineoplásicos/farmacología , Isoquinolinas/farmacología , Linfoma de Células del Manto/patología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Pirimidinas/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Isoquinolinas/química , Isoquinolinas/uso terapéutico , Linfoma de Células del Manto/tratamiento farmacológico , Linfoma de Células del Manto/enzimología , Masculino , Metilación , Ratones Endogámicos , Modelos Moleculares , Estructura Molecular , Unión Proteica , Pirimidinas/química , Pirimidinas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Nucleares snRNP/metabolismo
17.
Drug Metab Dispos ; 43(7): 936-43, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25887455

RESUMEN

Pharmacokinetic and metabolite identification studies were conducted to understand the clearance pathways of EPZ011652 [(2-aminoethyl)(methyl)({3-[4-(propan-2-yloxy)phenyl]-1H-pyrazol-4-yl}methyl)amine], a potent protein arginine N-methyltransferase inhibitor. Metabolic clearance was the major pathway of EPZ011652 elimination in rats with structural elucidation of metabolites via liquid chromatography - mass spectrometry (LC-MS(n)) accurate mass measurement revealing the formation of a novel aliphatic N-acetylated metabolite (M1) located on the terminal nitrogen of the ethylene-diamine side chain. EPZ015564, a synthetic standard of the N-acetyl product, was prepared and was also generated by human and rat, but not dog hepatocytes. In rat hepatocytes, on incubation with EPZ011652, the concentration of EPZ015564 initially increased before decreasing with incubation time, suggesting that the metabolite is itself a substrate for other metabolizing enzymes, in agreement with the identification of metabolites M2, M3, and M4 in rat bile, all N-acetylated metabolites, undergoing sequential phase I (demethylation, oxidation) or phase II (sulfation) reactions. Reaction phenotyping with recombinant human N-acetyltransferase (NAT) isoforms revealed that both NAT1 and NAT2 are capable of acetylating EPZ011652, although with different catalytic efficiencies. Kinetic profiles of EPZ015564 formation followed classic Michaelis-Menten behavior with apparent Km values of >1000 µM for NAT1 and 165 ± 14.1 µM for NAT2. The in vitro intrinsic clearance for EPZ011652 by NAT2 (110 µL/min/mg) was 500-fold greater than by NAT1. In summary, we report the unusual N-acetylation of an aliphatic amine and discuss the implications for drug discovery and clinical development.


Asunto(s)
Aminas/metabolismo , Inhibidores Enzimáticos/metabolismo , Etilenodiaminas/metabolismo , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Pirazoles/metabolismo , Animales , Arilamina N-Acetiltransferasa/metabolismo , Bilis/metabolismo , Biotransformación , Perros , Cromatografía de Gases y Espectrometría de Masas , Hepatocitos/metabolismo , Humanos , Isoenzimas/metabolismo , Cinética , Hígado/metabolismo , Masculino , Redes y Vías Metabólicas , Ratas , Ratas Sprague-Dawley
18.
FEBS Lett ; 585(3): 539-44, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21237159

RESUMEN

Exposure of cell lines endogenously expressing the thyroid hormone activating enzyme type 2 deiodinase (D2) to the chemical chaperones tauroursodeoxycholic acid (TUDCA) or 4-phenylbutiric acid (4-PBA) increases D2 expression, activity and T3 production. In brown adipocytes, TUDCA or 4-PBA induced T3-dependent genes and oxygen consumption (∼2-fold), an effect partially lost in D2 knockout cells. In wild type, but not in D2 knockout mice, administration of TUDCA lowered the respiratory quotient, doubled brown adipose tissue D2 activity and normalized the glucose intolerance associated with high fat feeding. Thus, D2 plays a critical role in the metabolic effects of chemical chaperones.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Yoduro Peroxidasa/metabolismo , Fenilbutiratos/farmacología , Ácido Tauroquenodesoxicólico/farmacología , Triyodotironina/metabolismo , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/metabolismo , Animales , Línea Celular , Células Cultivadas , Grasas de la Dieta/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Intolerancia a la Glucosa/prevención & control , Humanos , Yoduro Peroxidasa/genética , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Consumo de Oxígeno/efectos de los fármacos , ARN Mensajero/metabolismo , Yodotironina Deyodinasa Tipo II
19.
Endocrinology ; 151(9): 4573-82, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20660060

RESUMEN

Type 2 deiodinase (D2), which is highly expressed in brown adipose tissue (BAT), is an enzyme that amplifies thyroid hormone signaling in individual cells. Mice with inactivation of the D2 pathway (D2KO) exhibit dramatically impaired thermogenesis in BAT, leading to hypothermia during cold exposure and a greater susceptibility to diet-induced obesity. This was interpreted as a result of defective acute activation of BAT D2. Here we report that the adult D2KO BAT has a permanent thermogenic defect that stems from impaired embryonic BAT development. D2KO embryos have normal serum T3 but due to lack of D2-generated T3 in BAT, this tissue exhibits decreased expression of genes defining BAT identity [i.e. UCP1, PGC-1alpha and Dio2 (nonfunctional)], which results in impaired differentiation and oxidative capacity. Coinciding with a reduction of these T3-responsive genes, there is oxidative stress that in a cell model of brown adipogenesis can be linked to decreased insulin signaling and decreased adipogenesis. This discovery highlights the importance of deiodinase-controlled thyroid hormone signaling in BAT development, where it has important metabolic repercussions for energy homeostasis in adulthood.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Yoduro Peroxidasa/metabolismo , Termogénesis/fisiología , Hormonas Tiroideas/metabolismo , Aclimatación/genética , Aclimatación/fisiología , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/genética , Adipogénesis/fisiología , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/crecimiento & desarrollo , Animales , Western Blotting , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Yoduro Peroxidasa/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Consumo de Oxígeno/genética , Consumo de Oxígeno/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Temperatura , Termogénesis/genética , Hormonas Tiroideas/sangre , Factores de Tiempo , Yodotironina Deyodinasa Tipo II
20.
Genes Dev ; 24(13): 1403-17, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20595232

RESUMEN

The sterol regulatory element-binding protein (SREBP) transcription factor family is a critical regulator of lipid and sterol homeostasis in eukaryotes. In mammals, SREBPs are highly active in the fed state to promote the expression of lipogenic and cholesterogenic genes and facilitate fat storage. During fasting, SREBP-dependent lipid/cholesterol synthesis is rapidly diminished in the mouse liver; however, the mechanism has remained incompletely understood. Moreover, the evolutionary conservation of fasting regulation of SREBP-dependent programs of gene expression and control of lipid homeostasis has been unclear. We demonstrate here a conserved role for orthologs of the NAD(+)-dependent deacetylase SIRT1 in metazoans in down-regulation of SREBP orthologs during fasting, resulting in inhibition of lipid synthesis and fat storage. Our data reveal that SIRT1 can directly deacetylate SREBP, and modulation of SIRT1 activity results in changes in SREBP ubiquitination, protein stability, and target gene expression. In addition, chemical activators of SIRT1 inhibit SREBP target gene expression in vitro and in vivo, correlating with decreased hepatic lipid and cholesterol levels and attenuated liver steatosis in diet-induced and genetically obese mice. We conclude that SIRT1 orthologs play a critical role in controlling SREBP-dependent gene regulation governing lipid/cholesterol homeostasis in metazoans in response to fasting cues. These findings may have important biomedical implications for the treatment of metabolic disorders associated with aberrant lipid/cholesterol homeostasis, including metabolic syndrome and atherosclerosis.


Asunto(s)
Regulación hacia Abajo , Ayuno/fisiología , Sirtuina 1/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Acetilación , Animales , Benzamidas/farmacología , Caenorhabditis elegans , Línea Celular , Colesterol/biosíntesis , Regulación hacia Abajo/efectos de los fármacos , Células HeLa , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Lípidos/biosíntesis , Ratones , Naftoles/farmacología , Niacinamida/farmacología , Estabilidad Proteica/efectos de los fármacos , Sirtuinas/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...