Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Theranostics ; 11(17): 8172-8184, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34373735

RESUMO

Chromosome translocations involving mixed lineage leukemia (MLL) gene cause acute leukemia with a poor prognosis. MLL is frequently fused with transcription cofactors AF4 (~35%), AF9 (25%) or its paralog ENL (10%). The AHD domain of AF9/ENL binds to AF4, its paralog AFF4, or histone-H3 lysine-79 (H3K79) methyltransferase DOT1L. Formation of AF9/ENL/AF4/AFF4-containing super elongation complexes (SEC) and the catalytic activity of DOT1L are essential for MLL-rearranged leukemia. Protein-protein interactions (PPI) between AF9/ENL and DOT1L/AF4/AFF4 are therefore a potential drug target. Methods: Compound screening followed by medicinal chemistry was used to find inhibitors of such PPIs, which were examined for their biological activities against MLL-rearranged leukemia and other cancer cells. Results: Compound-1 was identified to be a novel small-molecule inhibitor of the AF9/ENL-DOT1L/AF4/AFF4 interaction with IC50s of 0.9-3.5 µM. Pharmacological inhibition of the PPIs significantly reduced SEC and DOT1L-mediated H3K79 methylation in the leukemia cells. Gene profiling shows compound-1 significantly suppressed the gene signatures related to onco-MLL, DOT1L, HoxA9 and Myc. It selectively inhibited proliferation of onco-MLL- or Myc-driven cancer cells and induced cell differentiation and apoptosis. Compound-1 exhibited strong antitumor activity in a mouse model of MLL-rearranged leukemia. Conclusions: The AF9/ENL-DOT1L/AF4/AFF4 interactions are validated to be an anticancer target and compound-1 is a useful in vivo probe for biological studies as well as a pharmacological lead for further drug development.


Assuntos
Antineoplásicos/farmacologia , Leucemia Mieloide Aguda , Proteínas de Fusão Oncogênica , Animais , Expressão Gênica/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/genética , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Camundongos , Proteínas de Fusão Oncogênica/química , Proteínas de Fusão Oncogênica/efeitos dos fármacos , Proteínas de Fusão Oncogênica/genética , Oncogenes/efeitos dos fármacos , Domínios e Motivos de Interação entre Proteínas , Fatores de Elongação da Transcrição/química , Fatores de Elongação da Transcrição/efeitos dos fármacos , Fatores de Elongação da Transcrição/genética
2.
Br J Haematol ; 195(2): 289-292, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34254287
3.
J Med Chem ; 64(3): 1584-1592, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33522809

RESUMO

Increased activity of the lysine methyltransferase NSD2 driven by translocation and activating mutations is associated with multiple myeloma and acute lymphoblastic leukemia, but no NSD2-targeting chemical probe has been reported to date. Here, we present the first antagonists that block the protein-protein interaction between the N-terminal PWWP domain of NSD2 and H3K36me2. Using virtual screening and experimental validation, we identified the small-molecule antagonist 3f, which binds to the NSD2-PWWP1 domain with a Kd of 3.4 µM and abrogates histone H3K36me2 binding to the PWWP1 domain in cells. This study establishes an alternative approach to targeting NSD2 and provides a small-molecule antagonist that can be further optimized into a chemical probe to better understand the cellular function of this protein.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Proteínas Repressoras/antagonistas & inibidores , Simulação por Computador , Cristalografia por Raios X , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Humanos , Ligantes , Modelos Moleculares , Simulação de Acoplamento Molecular , Domínios Proteicos , Proteínas Repressoras/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas , Relação Estrutura-Atividade
4.
Pediatr Blood Cancer ; 67(7): e28284, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32333633

RESUMO

BACKGROUND: VTP-50469 is a potent inhibitor of the menin-MLL1 interaction and is implicated in signaling downstream of EWSR1-FLI1. PROCEDURE: VTP-50469 was evaluated against seven Ewing sarcoma (EwS) xenograft models and in vitro against EwS cell lines. RESULTS: VTP-50469 showed limited antitumor activity, statistically significantly slowing tumor progression in four tumor models but with no evidence of tumor regression. In vitro, the IC50 concentration was 10 nM for the mixed lineage leukemia (MLL)-rearranged leukemia cell line MV4;11, but > 3 µM for EwS cell lines. CONCLUSIONS: In contrast to its high level of activity against MLL1-rearranged leukemia xenografts, VTP-50469 shows little activity against EwS models.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Proteína de Leucina Linfoide-Mieloide/efeitos dos fármacos , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Sarcoma de Ewing/tratamento farmacológico , Animais , Antineoplásicos/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Feminino , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Camundongos , Proteína de Leucina Linfoide-Mieloide/metabolismo , Pediatria , Proteínas Proto-Oncogênicas/metabolismo , Sarcoma de Ewing/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Bioorg Med Chem Lett ; 29(4): 638-645, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30626558

RESUMO

The protein-protein interaction between WDR5 (WD40 repeat protein 5) and MLL1 (mixed-lineage leukemia 1) is important for maintaining optimal H3K4 methyltransferase activity of MLL1. Dysregulation of MLL1 catalytic function is relevant to mixed-lineage leukemia, and targeting WDR5-MLL1 interaction could be a promising therapeutic strategy for leukemia harboring MLL1 fusion proteins. To date, several peptidomimetic and non-peptidomimetic small-molecule inhibitors targeting WDR5-MLL1 interaction have been reported, yet the discovery walk of new drugs inhibiting MLL1 methytransferase activity is still in its infancy. It's urgent to find other small-molecule WDR5-MLL1 inhibitors with novel scaffolds. In this study, through fluorescence polarization (FP)-based high throughput screening, several small-molecule inhibitors with potent inhibitory activities in vitro against WDR5-MLL1 interaction were discovered. Nuclear Magnetic Resonance (NMR) assays were carried out to confirm the direct binding between hit compounds and WDR5. Subsequent similarity-based analog searching of the 4 hits led to several inhibitors with better activity, among them, DC_M5_2 displayed highest inhibitory activity with IC50 values of 9.63 ±â€¯1.46 µM. Furthermore, a molecular docking study was performed and disclosed the binding modes and interaction mechanisms between two most potent inhibitors and WDR5.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Proteína de Leucina Linfoide-Mieloide/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Polarização de Fluorescência , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Ligação Proteica
6.
Int J Neuropsychopharmacol ; 22(4): 292-302, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30590608

RESUMO

BACKGROUND: Tolerance to ethanol-induced anxiolysis promotes alcohol intake, thus contributing to alcohol use disorder development. Recent studies implicate histone deacetylase-mediated histone H3K9 deacetylation in regulating neuropeptide Y expression during rapid ethanol tolerance to the anxiolytic effects of ethanol. Furthermore, the histone methyltransferase, G9a, and G9a-mediated H3K9 dimethylation (H3K9me2) have recently emerged as regulators of addiction and anxiety; however, their role in rapid ethanol tolerance is unknown. Therefore, we investigated the role of G9a-mediated H3K9me2 in neuropeptide Y expression during rapid ethanol tolerance. METHODS: Adult male rats were administered one injection of n-saline followed by single acute ethanol injection (1 g/kg) 24 hours later (ethanol group) or 2 injections (24 hours apart) of either n-saline (saline group) or ethanol (tolerance group). Anxiety-like behaviors and global and Npy-specific G9a and H3K9me2 levels in the amygdala were measured. Effects of G9a inhibitor (UNC0642) treatment on behavioral and epigenetic measures were also examined. RESULTS: Acute ethanol produced anxiolysis and decreased global H3K9me2 and G9a protein levels in the central and medial nucleus of the amygdala as well as decreased occupancy levels of H3K9me2 and G9a near a putative binding site for cAMP-response element binding protein on the Npy gene. Two identical doses of ethanol produced no behavioral or epigenetic changes relative to controls, indicating development of rapid ethanol tolerance. Interestingly, treatment with UNC0642, before the second ethanol dose reversed rapid ethanol tolerance, decreased global H3K9me2 and increased neuropeptide Y levels in the central and medial nucleus of the amygdala. CONCLUSIONS: These results implicate amygdaloid G9a-mediated H3K9me2 mechanisms in regulating rapid tolerance to the anxiolytic effects of ethanol via neuropeptide Y expression regulation.


Assuntos
Tonsila do Cerebelo , Ansiolíticos/farmacologia , Ansiedade/tratamento farmacológico , Tolerância a Medicamentos , Etanol/farmacologia , Histona-Lisina N-Metiltransferase , Neuropeptídeo Y , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Ansiolíticos/administração & dosagem , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Epigênese Genética/efeitos dos fármacos , Etanol/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/metabolismo , Masculino , Neuropeptídeo Y/efeitos dos fármacos , Neuropeptídeo Y/metabolismo , Quinazolinas/farmacologia , Ratos , Ratos Sprague-Dawley
7.
Nat Commun ; 9(1): 4559, 2018 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-30455465

RESUMO

Epigenetic regulators are attractive anticancer targets, but the promise of therapeutic strategies inhibiting some of these factors has not been proven in vivo or taken into account tumor cell heterogeneity. Here we show that the histone methyltransferase G9a, reported to be a therapeutic target in many cancers, is a suppressor of aggressive lung tumor-propagating cells (TPCs). Inhibition of G9a drives lung adenocarcinoma cells towards the TPC phenotype by de-repressing genes which regulate the extracellular matrix. Depletion of G9a during tumorigenesis enriches tumors in TPCs and accelerates disease progression metastasis. Depleting histone demethylases represses G9a-regulated genes and TPC phenotypes. Demethylase inhibition impairs lung adenocarcinoma progression in vivo. Therefore, inhibition of G9a is dangerous in certain cancer contexts, and targeting the histone demethylases is a more suitable approach for lung cancer treatment. Understanding cellular context and specific tumor populations is critical when targeting epigenetic regulators in cancer for future therapeutic development.


Assuntos
Progressão da Doença , Histona Desmetilases/metabolismo , Histona Metiltransferases/metabolismo , Neoplasias Pulmonares/metabolismo , Adenocarcinoma de Pulmão/metabolismo , Animais , Carcinogênese , Linhagem Celular Tumoral/efeitos dos fármacos , Sobrevivência Celular , Modelos Animais de Doenças , Matriz Extracelular/genética , Histona Desmetilases/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Organoides/anatomia & histologia , Fenótipo , Proteínas Proto-Oncogênicas p21(ras)/genética
8.
Endocrinology ; 159(5): 2216-2228, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29718165

RESUMO

Ample evidence suggests that environmental and occupational exposure to bisphenol A (BPA) and phthalate, two chemicals widely used in the plastics industry, disturbs homeostasis of innate immunity and causes inflammatory diseases. However, the underlying molecular mechanisms of these toxicants in the regulation of macrophage inflammatory functions remain poorly understood. In this study, we addressed the effect of chronic exposure to BPA or phthalate at levels relevant to human exposure, either in vitro or in vivo, on the inflammatory reprograming of peritoneal macrophages. Our studies revealed that BPA and phthalates adversely affected expression levels of the proinflammatory cytokines and mediators in response to lipopolysaccharide stimulation. Exposure to these toxicants also affected gene expression of scavenger receptors and phagocytic capacity of peritoneal macrophages. Our studies revealed that the epigenetic inhibitors differentially modulated target gene expression in these cells. Further analysis revealed that certain histone modification enzymes were aberrantly expressed in response to BPA or phthalate exposure, leading to alteration in the levels of H3K36 acetylation and dimethylation, two chromatin modifications that are critical for transcriptional efficacy and accuracy. Our results further revealed that silencing of H3K36-specific methyltransferase Smyd2 expression or inhibition of SMYD2 enzymatic activity attenuated H3K36 dimethylation and enhanced interleukin-6 and tumor necrosis factor-α expression but dampened the phagocytic capacity of peritoneal macrophages. In summary, our results indicate that peritoneal macrophages are vulnerable to BPA or phthalate at levels relevant to human exposure. These environmental toxicants affect phenotypic programming of macrophages via epigenetic mechanisms involving SMYD2-mediated H3K36 modification.


Assuntos
Compostos Benzidrílicos/farmacologia , Estrogênios não Esteroides/farmacologia , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Histonas/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Fenóis/farmacologia , Ácidos Ftálicos/farmacologia , Animais , Epigênese Genética , Feminino , Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Interleucina-6/genética , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/metabolismo , Camundongos , Fagocitose/efeitos dos fármacos , Fagocitose/genética , Fator de Necrose Tumoral alfa/efeitos dos fármacos
9.
Oncol Rep ; 38(5): 2796-2802, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28901481

RESUMO

NSD3 is a histone lysine methyltransferase that methylates histone H3 at lysine 36. NSD3 is located at chromosome 8p11.23, the locus that exhibits strong cancer relevance. Thus, NSD3 is likely involved in multiple human cancers. Nevertheless, its roles in human carcinogenesis remain unknown. In the present study, we demonstrated that silencing of NSD3 in osteosarcoma, the most common primary bone cancer in children and adolescents, results in a marked decrease in the number of viable cancer cells, accompanied by increases in the cell population at the G2/M phase and the number of apoptotic cells. In addition, 549 NSD3­regulated genes were identified and a set of selected candidate genes were validated. Bioinformatic analysis revealed that NSD3 negatively regulates a number of genes that are involved in the process of negative regulation of signal transduction as well as negative regulation of signaling and cell communication. Our results indicate the oncogenic roles of NSD3 in the development and progression of human osteosarcoma, and implicate NSD3 as a potential molecular target for selective therapy for human osteosarcoma.


Assuntos
Neoplasias Ósseas/genética , Redes Reguladoras de Genes , Histona-Lisina N-Metiltransferase/genética , Proteínas Nucleares/genética , Osteossarcoma/genética , RNA Interferente Pequeno/farmacologia , Análise de Sequência de RNA/métodos , Adolescente , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Criança , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Humanos , Proteínas Nucleares/efeitos dos fármacos
10.
Mol Pain ; 13: 1744806917729305, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28814147

RESUMO

Background: Previous studies disclosed the pivotal role of methyltransferase complex G9a/Glp in the pathogenesis of neuropathic hypersensitivity induced by peripheral nerve injury. We observed that higher dose of G9a inhibitor improved nociceptive behavior, but the lower dose worsened pain. The aim of this study is to extensively observe the differential effect of various dosages of G9a/Glp inhibitors on nerve injury-induced allodynia. Materials and methods: After approval by the institutional ethical committee on pain research in conscious animals, C57BL/6 mice were used for measuring nociceptive behavior evoked with von Frey filaments after spared nerve injury. G9a/Glp inhibitor BIX01294 or UNC0638 was injected through the pre-buried intrathecal catheter. The dose­response curves of behavioral changes were depicted when inhibitors were administered once in bolus at the 14th day post spared nerve injury. Withdrawal behaviors were compared during the 49 days' observation window after spared nerve injury with various dosages of inhibitors injected intrathecally for 14 days. Results: Dose­behavior curves of a single bolus of both BIX01294 and UNC0638 displayed a "V"-shaped responses of allodynia withdrawal from lower through higher dose when measured at the 14th day post spared nerve injury. A threshold dose of 10.0 µg for BIX01294 and 80.0 µg for UNC0638 significantly worsened allodynia. However, daily bolus intrathecal injection for 14 days of both inhibitors lower or higher than these threshold doses prominently improved nociceptive behavior, producing contrasting results. On the same animal, threshold dose followed by a lower or higher dose with a 14 days' interval also showed contrast effect on nociceptive behavior, and a lower or higher dose to threshold dose sequence of inhibitor administration was vice versa. Conclusions: Methyltransferase complex G9a/Glp has a threshold role in mediating peripheral nerve injury-induced hypersensitivity at its low level versus high level through inhibiting and facilitating the nociceptive behavior, respectively.


Assuntos
Hiperalgesia/tratamento farmacológico , Hipersensibilidade/complicações , Traumatismos dos Nervos Periféricos/tratamento farmacológico , Quinazolinas/farmacologia , Animais , Modelos Animais de Doenças , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Hipersensibilidade/etiologia , Injeções Espinhais/métodos , Masculino , Camundongos Endogâmicos C57BL , Dor/tratamento farmacológico , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Traumatismos dos Nervos Periféricos/complicações
11.
PLoS One ; 11(9): e0083032, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27622288

RESUMO

BACKGROUND: The dynamic and differential regulation and expression of genes is majorly governed by the complex interactions of a subset of biomolecules in the cell operating at multiple levels starting from genome organisation to protein post-translational regulation. The regulatory layer contributed by the epigenetic layer has been one of the favourite areas of interest recently. This layer of regulation as we know today largely comprises of DNA modifications, histone modifications and noncoding RNA regulation and the interplay between each of these major components. Epigenetic regulation has been recently shown to be central to development of a number of disease processes. The availability of datasets of high-throughput screens for molecules for biological properties offer a new opportunity to develop computational methodologies which would enable in-silico screening of large molecular libraries. METHODS: In the present study, we have used data from high throughput screens for the inhibitors of epigenetic modifiers. Computational predictive models were constructed based on the molecular descriptors. Machine learning algorithms for supervised training, Naive Bayes and Random Forest, were used to generate predictive models for the small molecule inhibitors of histone methyl-transferases and demethylases. Random forest, with the accuracy of 80%, was identified as the most accurate classifier. Further we complemented the study with substructure search approach filtering out the probable pharmacophores from the active molecules leading to drug molecules. RESULTS: We show that effective use of appropriate computational algorithms could be used to learn molecular and structural correlates of biological activities of small molecules. The computational models developed could be potentially used to screen and identify potential new biological activities of molecules from large molecular libraries and prioritise them for in-depth biological assays. To the best of our knowledge, this is the first and most comprehensive computational analysis towards understanding activities of small molecules inhibitors of epigenetic modifiers.


Assuntos
Biologia Computacional/métodos , Descoberta de Drogas/métodos , Epigênese Genética/efeitos dos fármacos , Algoritmos , Animais , Ensaios de Triagem em Larga Escala , Histona Desmetilases/efeitos dos fármacos , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Humanos , Modelos Teóricos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Bibliotecas de Moléculas Pequenas , Relação Estrutura-Atividade
12.
Psychiatry Clin Neurosci ; 70(12): 536-550, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27485392

RESUMO

The mammalian brain dynamically activates or silences gene programs in response to environmental input and developmental cues. This neuroplasticity is controlled by signaling pathways that modify the activity, localization, and/or expression of transcriptional-regulatory enzymes in combination with alterations in chromatin structure in the nucleus. Consistent with this key neurobiological role, disruptions in the fine-tuning of epigenetic and transcriptional regulation have emerged as a recurrent theme in studies of the genetics of neurodevelopmental and neuropsychiatric disorders. Furthermore, environmental factors have been implicated in the increased risk of heterogeneous, multifactorial, neuropsychiatric disorders via epigenetic mechanisms. Aberrant epigenetic regulation of gene expression thus provides an attractive unifying model for understanding the complex risk architecture of mental illness. Here, we review emerging genetic evidence implicating dysregulation of histone lysine methylation in neuropsychiatric disease and outline advancements in small-molecule probes targeting this chromatin modification. The emerging field of neuroepigenetic research is poised to provide insight into the biochemical basis of genetic risk for diverse neuropsychiatric disorders and to develop the highly selective chemical tools and imaging agents necessary to dissect dynamic transcriptional-regulatory mechanisms in the nervous system. On the basis of these findings, continued advances may lead to the validation of novel, disease-modifying therapeutic targets for a range of disorders with aberrant chromatin-mediated neuroplasticity.


Assuntos
Descoberta de Drogas , Epigênese Genética/fisiologia , Histona Desmetilases/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Transtornos Mentais/enzimologia , Doenças do Sistema Nervoso/enzimologia , Plasticidade Neuronal/fisiologia , Animais , Epigênese Genética/efeitos dos fármacos , Histona Desmetilases/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Humanos , Transtornos Mentais/tratamento farmacológico , Doenças do Sistema Nervoso/tratamento farmacológico , Plasticidade Neuronal/efeitos dos fármacos
13.
J Med Chem ; 59(6): 2478-96, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-26958703

RESUMO

WD repeat-containing protein 5 (WDR5) is an important component of the multiprotein complex essential for activating mixed-lineage leukemia 1 (MLL1). Rearrangement of the MLL1 gene is associated with onset and progression of acute myeloid and lymphoblastic leukemias, and targeting the WDR5-MLL1 interaction may result in new cancer therapeutics. Our previous work showed that binding of small molecule ligands to WDR5 can modulate its interaction with MLL1, suppressing MLL1 methyltransferase activity. Initial structure-activity relationship studies identified N-(2-(4-methylpiperazin-1-yl)-5-substituted-phenyl) benzamides as potent and selective antagonists of this protein-protein interaction. Guided by crystal structure data and supported by in silico library design, we optimized the scaffold by varying the C-1 benzamide and C-5 substituents. This allowed us to develop the first highly potent (Kdisp < 100 nM) small molecule antagonists of the WDR5-MLL1 interaction and demonstrate that N-(4-(4-methylpiperazin-1-yl)-3'-(morpholinomethyl)-[1,1'-biphenyl]-3-yl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyridine-3-carboxamide 16d (OICR-9429) is a potent and selective chemical probe suitable to help dissect the biological role of WDR5.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Compostos de Bifenilo/síntese química , Compostos de Bifenilo/farmacologia , Di-Hidropiridinas/síntese química , Di-Hidropiridinas/farmacologia , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Leucemia/tratamento farmacológico , Proteína de Leucina Linfoide-Mieloide/antagonistas & inibidores , Animais , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Desenho de Fármacos , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos SCID , Modelos Moleculares , Simulação de Acoplamento Molecular , Bibliotecas de Moléculas Pequenas , Relação Estrutura-Atividade , Difração de Raios X
14.
Oncol Rep ; 33(5): 2384-92, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25738706

RESUMO

The aim of the present study was to demonstrate that ribavirin, a known inhibitor of eIF4E and inosine 5'-phosphate dehydrogenase (IMPDH), also inhibits histone methyltransferase zeste homolog 2 (EZH2). A computational searching revealed that ribavirin has a high structural similarity to 3-deazaneplanocin A (DZNep). The growth inhibitory effects of ribavirin as well as its effects upon epigenetic enzymes were evaluated in various cancer cell lines. siRNA assays were used to downregulate eIF4E, EZH2 and IMPDH to determine the contribution of these targets to the growth inhibitory effects of ribavirin. Ribavirin decreased EZH2 expression, inhibited histone methyltransferase activity and decreased H3K27 trimethylation. Ribavirin induced variable growth inhibition in a number of cell lines and downregulation of the targets, EZH2, eIF4E and IMPDH1 and 2 by siRNA led to comparable growth inhibition while no significant further reduction in viability was observed when siRNA transfected cells were treated with ribavirin. The results showed that ribavirin inhibits these cancer targets and should thus be studied for cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Antivirais/farmacologia , Proliferação de Células/efeitos dos fármacos , Fator de Iniciação 4E em Eucariotos/efeitos dos fármacos , IMP Desidrogenase/efeitos dos fármacos , Neoplasias/genética , Complexo Repressor Polycomb 2/efeitos dos fármacos , Ribavirina/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Simulação por Computador , Reposicionamento de Medicamentos , Proteína Potenciadora do Homólogo 2 de Zeste , Fator de Iniciação 4E em Eucariotos/genética , Células HeLa , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Humanos , IMP Desidrogenase/genética , Células MCF-7 , Neoplasias/metabolismo , Complexo Repressor Polycomb 2/genética , RNA Interferente Pequeno
15.
J Am Chem Soc ; 135(2): 669-82, 2013 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-23210835

RESUMO

Mixed lineage leukemia 1 (MLL1) is a histone H3 lysine 4 (H3K4) methyltransferase, and targeting the MLL1 enzymatic activity has been proposed as a novel therapeutic strategy for the treatment of acute leukemia harboring MLL1 fusion proteins. The MLL1/WDR5 protein-protein interaction is essential for MLL1 enzymatic activity. In the present study, we designed a large number of peptidomimetics to target the MLL1/WDR5 interaction based upon -CO-ARA-NH-, the minimum binding motif derived from MLL1. Our study led to the design of high-affinity peptidomimetics, which bind to WDR5 with K(i) < 1 nM and function as potent antagonists of MLL1 activity in a fully reconstituted in vitro H3K4 methyltransferase assay. Determination of co-crystal structures of two potent peptidomimetics in complex with WDR5 establishes their structural basis for high-affinity binding to WDR5. Evaluation of one such peptidomimetic, MM-102, in bone marrow cells transduced with MLL1-AF9 fusion construct shows that the compound effectively decreases the expression of HoxA9 and Meis-1, two critical MLL1 target genes in MLL1 fusion protein mediated leukemogenesis. MM-102 also specifically inhibits cell growth and induces apoptosis in leukemia cells harboring MLL1 fusion proteins. Our study provides the first proof-of-concept for the design of small-molecule inhibitors of the WDR5/MLL1 protein-protein interaction as a novel therapeutic approach for acute leukemia harboring MLL1 fusion proteins.


Assuntos
Sistemas de Liberação de Medicamentos , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Proteína de Leucina Linfoide-Mieloide/antagonistas & inibidores , Peptidomiméticos , Bibliotecas de Moléculas Pequenas , Ligação Competitiva , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Modelos Biológicos , Modelos Moleculares , Proteína de Leucina Linfoide-Mieloide/efeitos dos fármacos , Proteína de Leucina Linfoide-Mieloide/metabolismo , Peptidomiméticos/química , Ligação Proteica/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
17.
Nat Cell Biol ; 9(11): 1273-85, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17952062

RESUMO

Histone modifications induced by activated signalling cascades are crucial to cell-lineage decisions. Osteoblast and adipocyte differentiation from common mesenchymal stem cells is under transcriptional control by numerous factors. Although PPAR-gamma (peroxisome proliferator activated receptor-gamma) has been established as a prime inducer of adipogenesis, cellular signalling factors that determine cell lineage in bone marrow remain generally unknown. Here, we show that the non-canonical Wnt pathway through CaMKII-TAK1-TAB2-NLK transcriptionally represses PPAR-gamma transactivation and induces Runx2 expression, promoting osteoblastogenesis in preference to adipogenesis in bone marrow mesenchymal progenitors. Wnt-5a activates NLK (Nemo-like kinase), which in turn phosphorylates a histone methyltransferase, SETDB1 (SET domain bifurcated 1), leading to the formation of a co-repressor complex that inactivates PPAR-gamma function through histone H3-K9 methylation. These findings suggest that the non-canonical Wnt signalling pathway suppresses PPAR-gamma function through chromatin inactivation triggered by recruitment of a repressing histone methyltransferase, thus leading to an osteoblastic cell lineage from mesenchymal stem cells.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , PPAR gama/metabolismo , Transdução de Sinais/fisiologia , Ativação Transcricional/fisiologia , Proteínas Wnt/fisiologia , Adipogenia , Animais , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Regulação para Baixo , Vetores Genéticos , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Mutação , Osteogênese , PPAR gama/efeitos dos fármacos , PPAR gama/genética , Fosforilação , Plasmídeos , Transdução de Sinais/efeitos dos fármacos , Proteínas Wnt/genética , Proteínas Wnt/farmacologia , Proteína Wnt-5a
18.
Expert Opin Emerg Drugs ; 9(1): 135-54, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15155140

RESUMO

In eukaryotes, genomic DNA is packaged with histone proteins into the cell nucleus as chromatin, condensing the DNA > 10,000-fold. Chromatin is highly dynamic and exerts profound control on gene expression. Localised chromatin decondensation facilitates access of nuclear machinery. Chromatin displays epigenetic inheritance, in that changes in its structure can pass to the next generation independently of the DNA sequence itself. It is now clear that the post-translational modification of histones, for example, acetylation, methylation and phosphorylation, plays a crucial role in the regulation of nuclear function through the 'histone code'. There has been significant progress in identifying and understanding the enzymes that control these complex processes, in particular histone acetyltransferases and histone deacetylases. The exciting discovery that compounds inhibiting histone deacetylase activity also have antitumour properties has focused attention on their use as anticancer drugs. As a consequence, there is ongoing evaluation of several histone deacetylase inhibitor compounds in Phase I and II clinical trials with promising early results. It is likely that many of the enzymes involved in the control of histone modification will provide therapeutic opportunities for the treatment of cancer, including histone methyltransferases and Aurora kinases.


Assuntos
Acetiltransferases/efeitos dos fármacos , Antineoplásicos/farmacologia , Histona Desacetilases/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Histonas/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Protamina Quinase/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Acetilação/efeitos dos fármacos , Antineoplásicos/classificação , Antineoplásicos/uso terapêutico , Cromatina/efeitos dos fármacos , Cromatina/metabolismo , Ensaios Clínicos como Assunto , Desenho de Fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Histona Acetiltransferases , Histona Metiltransferases , Histonas/química , Metilação/efeitos dos fármacos , Estudos Multicêntricos como Assunto , Neoplasias/enzimologia , Neoplasias/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Metiltransferases , Estrutura Terciária de Proteína , Proteínas Tirosina Fosfatases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...