Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
2.
ACS Chem Biol ; 18(1): 34-40, 2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36594833

RESUMEN

WD repeat domain 5 (WDR5) is a member of the WD40-repeat protein family that plays a critical role in multiple processes. It is also a prominent target for pharmacological inhibition in diseases such as cancer, aging, and neurodegenerative disorders. Interactions between WDR5 and various partners are essential for sustaining its function. Most drug discovery efforts center on the WIN (WDR5 interaction motif) site of WDR5 that is responsible for the recruitment of WDR5 to chromatin. Here, we describe the discovery of novel WDR5 inhibitors for the other WBM (WDR5 binding motif) pocket on this scaffold protein, to disrupt WDR5 interaction with its binding partner MYC by high-throughput biochemical screening, subsequent molecule optimization, and biological assessment. These new WDR5 inhibitors provide useful probes for future investigations of WDR5 and an avenue for targeting WDR5 as a therapeutic strategy.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Neoplasias , Humanos , Unión Proteica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cromatina , Descubrimiento de Drogas
3.
Biochim Biophys Acta Mol Basis Dis ; 1869(2): 166600, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36402263

RESUMEN

WD repeat domain 5 (WDR5) is a prominent target for pharmacological inhibition in cancer through its scaffolding role with various oncogenic partners such as MLL and MYC. WDR5-related drug discovery efforts center on blocking these binding interfaces or degradation have been devoted to developing small-molecule inhibitors or degraders of WDR5 for cancer treatment. Nevertheless, the precise role of WDR5 in these cancer cells has not been well elucidated genetically. Here, by using an MLL-AF9 murine leukemia model, we found that genetically deletion of Wdr5 impairs cell growth and colony forming ability of MLL-AF9 leukemia cells in vitro or ex vivo and attenuates the leukemogenesis in vivo as well, which acts through direct regulation of ribosomal genes. Pharmacological inhibition of Wdr5 recapitulates genetic study results in the same model. In conclusion, our current study demonstrated the first genetic evidence for the indispensable role of Wdr5 in MLL-r leukemogenesis in vivo, which supports therapeutically targeting WDR5 in MLL-rearranged leukemia by strengthening its disease linkage genetically and deepening insights into its mechanism of action.


Asunto(s)
Carcinogénesis , Leucemia , Animales , Ratones , Carcinogénesis/genética , Descubrimiento de Drogas , Leucemia/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo
4.
J Med Chem ; 65(7): 5317-5333, 2022 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-35352560

RESUMEN

Polycomb Repressive Complex 2 (PRC2) plays an important role in transcriptional regulation during animal development and in cell differentiation, and alteration of PRC2 activity has been associated with cancer. On a molecular level, PRC2 catalyzes methylation of histone H3 lysine 27 (H3K27), resulting in mono-, di-, or trimethylated forms of H3K27, of which the trimethylated form H3K27me3 leads to transcriptional repression of polycomb target genes. Previously, we have shown that binding of the low-molecular-weight compound EED226 to the H3K27me3 binding pocket of the regulatory subunit EED can effectively inhibit PRC2 activity in cells and reduce tumor growth in mouse xenograft models. Here, we report the stepwise optimization of the tool compound EED226 toward the potent and selective EED inhibitor MAK683 (compound 22) and its subsequent preclinical characterization. Based on a balanced PK/PD profile, efficacy, and mitigated risk of forming reactive metabolites, MAK683 has been selected for clinical development.


Asunto(s)
Histonas , Neoplasias , Animales , Inhibidores Enzimáticos , Histonas/metabolismo , Humanos , Metilación , Ratones , Neoplasias/tratamiento farmacológico , Complejo Represivo Polycomb 2
7.
SLAS Discov ; 23(9): 930-940, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29562800

RESUMEN

Ubiquitin-like with PHD and RING finger domains 1 (UHRF1) is a multidomain protein that plays a critical role in maintaining DNA methylation patterns through concurrent recognition of hemimethylated DNA and histone marks by various domains, and recruitment of DNA methyltransferase 1 (DNMT1). UHRF1 is overexpressed in various cancers, including breast cancer. The tandem tudor domain (TTD) of UHRF1 specifically and tightly binds to histone H3 di- or trimethylated at lysine 9 (H3K9me2 or H3K9me3, respectively), and this binding is essential for UHRF1 function. We developed an H3K9me3 peptide displacement assay, which was used to screen a library of 44,000 compounds for small molecules that disrupt the UHRF1-H3K9me3 interaction. This screen resulted in the identification of NV01, which bound to UHRF1-TTD with a Kd value of 5 µM. The structure of UHRF1-TTD in complex with NV01 confirmed binding to the H3K9me3-binding pocket. Limited structure-based optimization of NV01 led to the discovery of NV03 (Kd of 2.4 µM). These well-characterized small-molecule antagonists of the UHRF1-H3K9me2/3 interaction could be valuable starting chemical matter for developing more potent and cell-active probes toward further characterizing UHRF1 function, with possible applications as anticancer therapeutics.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/química , Descubrimiento de Drogas/métodos , Histonas/química , Unión Proteica/efectos de los fármacos , Dominio Tudor , Sitios de Unión , Bioensayo/métodos , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Histonas/metabolismo , Humanos , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Mutagénesis Sitio-Dirigida , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad , Ubiquitina-Proteína Ligasas
8.
J Med Chem ; 61(3): 1204-1217, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29244490

RESUMEN

PRMT3 catalyzes the asymmetric dimethylation of arginine residues of various proteins. It is crucial for maturation of ribosomes and has been implicated in several diseases. We recently disclosed a highly potent, selective, and cell-active allosteric inhibitor of PRMT3, compound 4. Here, we report comprehensive structure-activity relationship studies that target the allosteric binding site of PRMT3. We conducted design, synthesis, and evaluation of novel compounds in biochemical, selectivity, and cellular assays that culminated in the discovery of 4 and other highly potent (IC50 values: ∼10-36 nM), selective, and cell-active allosteric inhibitors of PRMT3 (compounds 29, 30, 36, and 37). In addition, we generated compounds that are very close analogs of these potent inhibitors but displayed drastically reduced potency as negative controls (compounds 49-51). These inhibitors and negative controls are valuable chemical tools for the biomedical community to further investigate biological functions and disease associations of PRMT3.


Asunto(s)
Diseño de Fármacos , Proteína-Arginina N-Metiltransferasas/metabolismo , Regulación Alostérica/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Células HEK293 , Humanos , Enlace de Hidrógeno , Concentración 50 Inhibidora , Modelos Moleculares , Conformación Proteica , Proteína-Arginina N-Metiltransferasas/química , Relación Estructura-Actividad
9.
Oncotarget ; 8(49): 85085-85101, 2017 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-29156705

RESUMEN

LSD1 (Lysine Specific Demethylase1)/KDM1A (Lysine Demethylase 1A), a flavin adenine dinucleotide (FAD)-dependent histone H3K4/K9 demethylase, sustains oncogenic potential of leukemia stem cells in primary human leukemia cells. However, the pro-differentiation and anti-proliferation effects of LSD1 inhibition in acute myeloid leukemia (AML) are not yet fully understood. Here, we report that small hairpin RNA (shRNA) mediated LSD1 inhibition causes a remarkable transcriptional activation of myeloid lineage marker genes (CD11b/ITGAM and CD86), reduction of cell proliferation and decrease of clonogenic ability of human AML cells. Cell surface expression of CD11b and CD86 is significantly and dynamically increased in human AML cells upon sustained LSD1 inhibition. Chromatin immunoprecipitation and quantitative PCR (ChIP-qPCR) analyses of histone marks revealed that there is a specific increase of H3K4me2 modification and an accompanied increase of H3K4me3 modification at the respective CD11b and CD86 promoter region, whereas the global H3K4me2 level remains constant. Consistently, inhibition of LSD1 in vivo significantly blocks tumor growth and induces a prominent increase of CD11b and CD86. Taken together, our results demonstrate the anti-tumor properties of LSD1 inhibition on human AML cell line and mouse xenograft model. Our findings provide mechanistic insights into the LSD1 functions in controlling both differentiation and proliferation in AML.

10.
Elife ; 62017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28767039

RESUMEN

Tumor suppressor p53 prevents cell transformation by inducing apoptosis and other responses. Homozygous TP53 deletion occurs in various types of human cancers for which no therapeutic strategies have yet been reported. TCGA database analysis shows that the TP53 homozygous deletion locus mostly exhibits co-deletion of the neighboring gene FXR2, which belongs to the Fragile X gene family. Here, we demonstrate that inhibition of the remaining family member FXR1 selectively blocks cell proliferation in human cancer cells containing homozygous deletion of both TP53 and FXR2 in a collateral lethality manner. Mechanistically, in addition to its RNA-binding function, FXR1 recruits transcription factor STAT1 or STAT3 to gene promoters at the chromatin interface and regulates transcription thus, at least partially, mediating cell proliferation. Our study anticipates that inhibition of FXR1 is a potential therapeutic approach to targeting human cancers harboring TP53 homozygous deletion.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Homocigoto , Neoplasias/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Eliminación de Secuencia , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/genética , Secuencia de Bases , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Cromatina , Femenino , Edición Génica , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Inhibidores de las Cinasas Janus/análisis , Ratones , Ratones Endogámicos BALB C , Regiones Promotoras Genéticas , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT3/genética , Factores de Transcripción
11.
Cancer Res ; 77(10): 2594-2606, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28249907

RESUMEN

Histone deacetylase inhibitors (HDACi) may engage host immunity as one basis for their antitumor effects. Herein, we demonstrate an application of this concept using the HDACi panobinostat to augment the antitumor efficacy of trastuzumab (anti-HER2) therapy, through both tumor cell autonomous and nonautonomous mechanisms. In HER2+ tumors that are inherently sensitive to the cytostatic effects of trastuzumab, cotreatment with panobinostat abrogated AKT signaling and triggered tumor regression in mice that lacked innate and/or adaptive immune effector cells. However, the cooperative ability of panobinostat and trastuzumab to harness host anticancer immune defenses was essential for their curative activity in trastuzumab-refractory HER2+ tumors. In trastuzumab-resistant HER2+ AU565pv xenografts and BT474 tumors expressing constitutively active AKT, panobinostat enhanced the antibody-dependent cell-mediated cytotoxicity function of trastuzumab. IFNγ-mediated, CXCR3-dependent increases in tumor-associated NK cells underpinned the combined curative activity of panobinostat and trastuzumab in these tumors. These data highlight the immune-enhancing effects of panobinostat and provide compelling evidence that this HDACi can license trastuzumab to evoke NK-cell-mediated responses capable of eradicating trastuzumab-refractory HER2+ tumors. Cancer Res; 77(10); 2594-606. ©2017 AACR.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Inmunidad Innata/efectos de los fármacos , Indoles/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Trastuzumab/farmacología , Animales , Biomarcadores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Humanos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Recuento de Linfocitos , Ratones , Panobinostat , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Med Chem ; 60(6): 2215-2226, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28092155

RESUMEN

Overexpression and somatic heterozygous mutations of EZH2, the catalytic subunit of polycomb repressive complex 2 (PRC2), are associated with several tumor types. EZH2 inhibitor, EPZ-6438 (tazemetostat), demonstrated clinical efficacy in patients with acceptable safety profile as monotherapy. EED, another subunit of PRC2 complex, is essential for its histone methyltransferase activity through direct binding to trimethylated lysine 27 on histone 3 (H3K27Me3). Herein we disclose the discovery of a first-in-class potent, selective, and orally bioavailable EED inhibitor compound 43 (EED226). Guided by X-ray crystallography, compound 43 was discovered by fragmentation and regrowth of compound 7, a PRC2 HTS hit that directly binds EED. The ensuing scaffold hopping followed by multiparameter optimization led to the discovery of 43. Compound 43 induces robust and sustained tumor regression in EZH2MUT preclinical DLBCL model. For the first time we demonstrate that specific and direct inhibition of EED can be effective as an anticancer strategy.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Sulfonas/química , Sulfonas/farmacología , Triazoles/química , Triazoles/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Perros , Femenino , Haplorrinos , Histonas/metabolismo , Humanos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/metabolismo , Lisina/metabolismo , Masculino , Metilación/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Simulación del Acoplamiento Molecular , Complejo Represivo Polycomb 2/química , Complejo Represivo Polycomb 2/metabolismo , Ratas , Sulfonas/farmacocinética , Sulfonas/uso terapéutico , Triazoles/farmacocinética , Triazoles/uso terapéutico
13.
PLoS One ; 12(1): e0169855, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28072869

RESUMEN

Polycomb repressive complex 2 (PRC2), a histone H3 lysine 27 methyltransferase, plays a key role in gene regulation and is a known epigenetics drug target for cancer therapy. The WD40 domain-containing protein EED is the regulatory subunit of PRC2. It binds to the tri-methylated lysine 27 of the histone H3 (H3K27me3), and through which stimulates the activity of PRC2 allosterically. Recently, we disclosed a novel PRC2 inhibitor EED226 which binds to the K27me3-pocket on EED and showed strong antitumor activity in xenograft mice model. Here, we further report the identification and validation of four other EED binders along with EED162, the parental compound of EED226. The crystal structures for all these five compounds in complex with EED revealed a common deep pocket induced by the binding of this diverse set of compounds. This pocket was created after significant conformational rearrangement of the aromatic cage residues (Y365, Y148 and F97) in the H3K27me3 binding pocket of EED, the width of which was delineated by the side chains of these rearranged residues. In addition, all five compounds interact with the Arg367 at the bottom of the pocket. Each compound also displays unique features in its interaction with EED, suggesting the dynamics of the H3K27me3 pocket in accommodating the binding of different compounds. Our results provide structural insights for rational design of novel EED binder for the inhibition of PRC2 complex activity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Simulación del Acoplamiento Molecular , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Sulfonas/farmacología , Triazoles/farmacología , Animales , Sitios de Unión , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Ensayos Analíticos de Alto Rendimiento , Ratones , Complejo Represivo Polycomb 2/química , Complejo Represivo Polycomb 2/metabolismo , Relación Estructura-Actividad Cuantitativa , Sulfonas/química , Triazoles/química
14.
Nat Chem Biol ; 13(4): 381-388, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28135235

RESUMEN

Polycomb repressive complex 2 (PRC2) consists of three core subunits, EZH2, EED and SUZ12, and plays pivotal roles in transcriptional regulation. The catalytic subunit EZH2 methylates histone H3 lysine 27 (H3K27), and its activity is further enhanced by the binding of EED to trimethylated H3K27 (H3K27me3). Small-molecule inhibitors that compete with the cofactor S-adenosylmethionine (SAM) have been reported. Here we report the discovery of EED226, a potent and selective PRC2 inhibitor that directly binds to the H3K27me3 binding pocket of EED. EED226 induces a conformational change upon binding EED, leading to loss of PRC2 activity. EED226 shows similar activity to SAM-competitive inhibitors in blocking H3K27 methylation of PRC2 target genes and inducing regression of human lymphoma xenograft tumors. Interestingly, EED226 also effectively inhibits PRC2 containing a mutant EZH2 protein resistant to SAM-competitive inhibitors. Together, we show that EED226 inhibits PRC2 activity via an allosteric mechanism and offers an opportunity for treatment of PRC2-dependent cancers.


Asunto(s)
Antineoplásicos/farmacología , Histonas/metabolismo , Lisina/metabolismo , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Sulfonas/química , Sulfonas/farmacología , Triazoles/química , Triazoles/farmacología , Regulación Alostérica/efectos de los fármacos , Animales , Antineoplásicos/química , Sitios de Unión/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Histonas/química , Humanos , Lisina/química , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Moleculares , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Complejo Represivo Polycomb 2/química , Complejo Represivo Polycomb 2/metabolismo , Relación Estructura-Actividad , Sulfonas/metabolismo , Triazoles/metabolismo , Células Tumorales Cultivadas
15.
Cell Chem Biol ; 23(10): 1228-1240, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27642069

RESUMEN

Adipose tissue plays important roles in animals. White fat stores energy in lipids, while brown fat is responsible for nonshivering thermogenesis through UCP1-mediated energy dissipation. Although epigenetic mechanisms modulate differentiation in multiple lineages, the epigenetic regulation of brown adipocyte differentiation is poorly understood. By screening a collection of epigenetic compounds, we found that Lysine-Specific Demethylase 1 (LSD1) inhibitors repress brown adipocyte differentiation. RNAi-mediated Lsd1 knockdown causes a similar effect, which can be rescued by expression of wild-type but not catalytic-inactive LSD1. Mechanistically, LSD1 promotes brown adipogenesis by demethylating H3K4 on promoter regions of Wnt signaling components and repressing the Wnt pathway. Furthermore, deletion of Lsd1 in mice leads to inhibition of brown adipogenesis, validating the pivotal role of LSD1 in brown fat development in vivo. Our work identifies LSD1 as a key epigenetic regulator in brown adipogenesis. The link between LSD1 and the Wnt pathway provides potential opportunities to modulate brown fat differentiation.


Asunto(s)
Adipocitos Marrones/citología , Adipogénesis , Histona Demetilasas/metabolismo , Vía de Señalización Wnt , Adipocitos Marrones/metabolismo , Animales , Células Cultivadas , Epigénesis Genética , Eliminación de Gen , Histona Demetilasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Expert Rev Anti Infect Ther ; 14(3): 345-52, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26822688

RESUMEN

Disequilibrium in the human debilitated immune system favors proliferation of invasive Candida species, a major therapeutic challenge due to development of resistance to several conventional antifungal agents (CAA) worldwide. Multiple mutations observed at specific loci that are targets for CAA are recognized as sources of drug resistance. This has prompted a shift from CAA, to diverse combination therapies, photodynamic and short peptide therapies capable of triggering specific apoptotic reactions within candidal cells. In this review, new designs and combination of short peptide (SP) with CAA as well as current application of photodynamic inactivation (PDI) against Candida species geared at generating reactive species of oxygen (ROS) and nitrogen (RNS) are discussed. It is observed that oxidative and nitrosative stresses provides a superior broad candidacidal effects for eradication of drug-resistant Candida species. The mechanism and limitations in these strategic approaches over CAA is also discussed.


Asunto(s)
Candidiasis/terapia , Farmacorresistencia Fúngica , Fotoquimioterapia , Antifúngicos/uso terapéutico , Candida , Humanos , Péptidos/uso terapéutico , Fotoquimioterapia/tendencias
18.
Nat Commun ; 6: 8651, 2015 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-26471002

RESUMEN

SETDB1 is a histone H3K9 methyltransferase that has a critical role in early development. It is located within a melanoma susceptibility locus and facilitates melanoma formation. However, the mechanism by which SETDB1 regulates tumorigenesis remains unknown. Here we report the molecular interplay between SETDB1 and the well-known hotspot gain-of-function (GOF) TP53 R249S mutation. We show that in hepatocellular carcinoma (HCC) SETDB1 is overexpressed with moderate copy number gain, and GOF TP53 mutations including R249S associate with this overexpression. Inactivation of SETDB1 in HCC cell lines bearing the R249S mutation suppresses cell growth. The TP53 mutation status renders cancer cells dependent on SETDB1. Moreover, SETDB1 forms a complex with p53 and catalyses p53K370 di-methylation. SETDB1 attenuation reduces the p53K370me2 level, which subsequently leads to increased recognition and degradation of p53 by MDM2. Together, we provide both genetic and biochemical evidence for a mechanism by which SETDB1 regulates cancer cell growth via methylation of p53.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Genes p53 , Neoplasias Hepáticas Experimentales/metabolismo , Proteína Metiltransferasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Variaciones en el Número de Copia de ADN , Células HCT116 , N-Metiltransferasa de Histona-Lisina , Humanos , Ratones Desnudos
19.
Genome Res ; 25(9): 1325-35, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26160163

RESUMEN

SETDB1, a histone methyltransferase responsible for methylation of histone H3 lysine 9 (H3K9), is involved in maintenance of embryonic stem (ES) cells and early embryonic development of the mouse. However, how SETDB1 regulates gene expression during development is largely unknown. Here, we characterized genome-wide SETDB1 binding and H3K9 trimethylation (H3K9me3) profiles in mouse ES cells and uncovered two distinct classes of SETDB1 binding sites, termed solo and ensemble peaks. The solo peaks were devoid of H3K9me3 and enriched near developmental regulators while the ensemble peaks were associated with H3K9me3. A subset of the SETDB1 solo peaks, particularly those near neural development-related genes, was found to be associated with Polycomb Repressive Complex 2 (PRC2) as well as PRC2-interacting proteins JARID2 and MTF2. Genetic deletion of Setdb1 reduced EZH2 binding as well as histone 3 lysine 27 (H3K27) trimethylation level at SETDB1 solo peaks and facilitated neural differentiation. Furthermore, we found that H3K27me3 inhibits SETDB1 methyltransferase activity. The currently identified reciprocal action between SETDB1 and PRC2 reveals a novel mechanism underlying ES cell pluripotency and differentiation regulation.


Asunto(s)
Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Animales , Sitios de Unión , Metilación , Ratones , Regiones Promotoras Genéticas , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos
20.
Angew Chem Int Ed Engl ; 54(17): 5166-70, 2015 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-25728001

RESUMEN

PRMT3 catalyzes the asymmetric dimethylation of arginine residues of various proteins. It is essential for maturation of ribosomes, may have a role in lipogenesis, and is implicated in several diseases. A potent, selective, and cell-active PRMT3 inhibitor would be a valuable tool for further investigating PRMT3 biology. Here we report the discovery of the first PRMT3 chemical probe, SGC707, by structure-based optimization of the allosteric PRMT3 inhibitors we reported previously, and thorough characterization of this probe in biochemical, biophysical, and cellular assays. SGC707 is a potent PRMT3 inhibitor (IC50 =31±2 nM, KD =53±2 nM) with outstanding selectivity (selective against 31 other methyltransferases and more than 250 non-epigenetic targets). The mechanism of action studies and crystal structure of the PRMT3-SGC707 complex confirm the allosteric inhibition mode. Importantly, SGC707 engages PRMT3 and potently inhibits its methyltransferase activity in cells. It is also bioavailable and suitable for animal studies. This well-characterized chemical probe is an excellent tool to further study the role of PRMT3 in health and disease.


Asunto(s)
Inhibidores Enzimáticos/química , Isoquinolinas/química , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Regulación Alostérica , Sitios de Unión , Calorimetría , Línea Celular Tumoral , Inhibidores Enzimáticos/metabolismo , Células HEK293 , Histonas , Humanos , Isoquinolinas/metabolismo , Metilación , Simulación de Dinámica Molecular , Mutagénesis , Unión Proteica , Estructura Terciaria de Proteína , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA