Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS One ; 19(4): e0299198, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38635661

RESUMO

Herpesviruses have two distinct life cycle stages, latency and lytic replication. Epstein-Barr virus (EBV), a gamma-herpesvirus, establishes latency in vivo and in cultured cells. Cell lines harboring latent EBV can be induced into the lytic cycle by treatment with chemical inducing agents. In the Burkitt lymphoma cell line HH514-16 the viral lytic cycle is triggered by butyrate, a histone deacetylase (HDAC) inhibitor. Butyrate also alters expression of thousands of cellular genes. However, valproic acid (VPA), another HDAC inhibitor with global effects on cellular gene expression blocks EBV lytic gene expression in Burkitt lymphoma cell lines. Valpromide (VPM), an amide derivative of VPA, is not an HDAC inhibitor, but like VPA blocks induction of the EBV lytic cycle. VPA and VPM are the first examples of inhibitors of initial stages of lytic reactivation. We compared the effects of VPA and VPM, alone and in combination with butyrate, on host cellular gene expression using whole transcriptome analysis (RNA-seq). Gene expression was analyzed 6 h after addition of the compounds, a time before the first EBV lytic transcripts are detected. The results address two alternative, yet possibly complementary, mechanisms for regulation of EBV lytic reactivation. First, cellular genes that were up- or down-regulated by butyrate, but no longer altered in the presence of VPA or VPM, represent genes that correlated with EBV lytic reactivation. Second, genes regulated similarly by VPA and VPM in the absence and presence of butyrate are candidates for suppressors of EBV reactivation. Two genes upregulated by the lytic cycle inhibitors, CHAC1 and SLC7A11, are related to redox status and the iron-dependent cell death pathway ferroptosis. This study generates new hypotheses for control of the latency to lytic cycle switch of EBV and provides the first description of effects of the anti-convulsant drug VPM on global human cellular gene expression.


Assuntos
Linfoma de Burkitt , Infecções por Vírus Epstein-Barr , Ácido Valproico/análogos & derivados , Humanos , Linfoma de Burkitt/tratamento farmacológico , Linfoma de Burkitt/genética , Herpesvirus Humano 4/fisiologia , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/metabolismo , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Ativação Viral , Perfilação da Expressão Gênica , Butiratos/farmacologia
2.
Vascul Pharmacol ; 146: 107096, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35952961

RESUMO

Recent studies have shown that short-chain fatty acids (SCFAs), primarily acetate, propionate and butyrate, play a crucial role in the pathogenesis of cardiovascular disease. Whether SCFAs regulate vascular calcification, a common pathological change in cardiovascular tissues, remains unclear. This study aimed to investigate the potential role of SCFAs in vascular calcification. Using cellular and animal models of vascular calcification, we showed that butyrate significantly enhanced high phosphate (Pi)-induced calcification and osteogenic transition of vascular smooth muscle cells (VSMC) in vitro, whereas acetate and propionate had no effects. Subsequent studies confirmed that butyrate significantly promoted high Pi-induced aortic ring calcification ex vivo and high dose vitamin D3 (vD3)-induced mouse vascular calcification in vivo. Mechanistically, butyrate significantly inhibited histone deacetylase (HDAC) expression in VSMCs, and a pan HDAC inhibitor Trichostatin A showed similar inductive effects on calcification and osteogenic transition of VSMCs to butyrate. In addition, the SCFA sensing receptors Gpr41 and Gpr109a were primarily expressed by VSMCs, and butyrate induced the rapid activation of NF-κB, Wnt and Akt signaling in VSMCs. Intriguingly, the NF-κB inhibitor SC75741 significantly attenuated butyrate-induced calcification and the osteogenic gene Msx2 expression in VSMCs. We showed that knockdown of Gpr41 but not Gpr109a attenuated butyrate-induced VSMC calcification. This study reveals that butyrate accelerates vascular calcification via its dual effects on HDAC inhibition and NF-κB activation. Our data provide novel insights into the role of microbe-host interaction in vascular calcification, and may have implications for the development of potential therapy for vascular calcification.


Assuntos
NF-kappa B , Calcificação Vascular , Animais , Butiratos/metabolismo , Butiratos/farmacologia , Células Cultivadas , Inibidores de Histona Desacetilases/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Camundongos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , NF-kappa B/metabolismo , Fosfatos , Propionatos/metabolismo , Propionatos/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Calcificação Vascular/patologia , Vitamina D
3.
J Med Chem ; 64(8): 4709-4729, 2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33797924

RESUMO

We describe the discovery of histone deacetylase (HDACs) 1, 2, and 3 inhibitors with ethyl ketone as the zinc-binding group. These HDACs 1, 2, and 3 inhibitors have good enzymatic and cellular activity. Their serum shift in cellular potency has been minimized, and selectivity against hERG has been improved. They are also highly selective over HDACs 6 and 8. These inhibitors contain a variety of substituted heterocycles on the imidazole or oxazole scaffold. Compounds 31 and 48 stand out due to their good potency, high selectivity over HDACs 6 and 8, reduced hERG activity, optimized serum shift in cellular potency, and good rat and dog PK profiles.


Assuntos
Canal de Potássio ERG1/metabolismo , HIV-1/fisiologia , Inibidores de Histona Desacetilases/química , Histona Desacetilases/metabolismo , Cetonas/química , Animais , Cães , Avaliação Pré-Clínica de Medicamentos , Meia-Vida , Histona Desacetilase 1/antagonistas & inibidores , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/antagonistas & inibidores , Histona Desacetilase 2/metabolismo , Inibidores de Histona Desacetilases/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/química , Humanos , Imidazóis/química , Oxazóis/química , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Ratos , Relação Estrutura-Atividade , Ativação Viral/efeitos dos fármacos
4.
J Med Chem ; 64(7): 3794-3812, 2021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33769811

RESUMO

The structures of melatonin and ferulic acid were merged into tertiary amide-based histone deacetylase 6 (HDAC6) inhibitors to develop multi-target-directed inhibitors for neurodegenerative diseases to incorporate antioxidant effects without losing affinity and selectivity at HDAC6. Structure-activity relationships led to compound 10b as a hybrid molecule showing pronounced and selective inhibition of HDAC6 (IC50 = 30.7 nM, > 25-fold selectivity over other subtypes). This compound shows comparable DPPH radical scavenging ability to ferulic acid, comparable ORAC value to melatonin and comparable Cu2+ chelating ability to EDTA. It also lacks neurotoxicity on HT-22 cells, exhibits a pronounced immunomodulatory effect, and is active in vivo showing significantly higher efficacy in an AD mouse model to prevent both Aß25-35-induced spatial working and long-term memory dysfunction at lower dose (0.3 mg/kg) compared to positive control HDAC6 inhibitor ACY1215 and an equimolar mixture of the three entities ACY1215, melatonin and ferulic acid, suggesting potentially disease-modifying properties.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Ácidos Cumáricos/uso terapêutico , Desacetilase 6 de Histona/antagonistas & inibidores , Fatores Imunológicos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Triptaminas/uso terapêutico , Doença de Alzheimer/enzimologia , Doença de Alzheimer/metabolismo , Animais , Domínio Catalítico , Linhagem Celular Transformada , Ácidos Cumáricos/síntese química , Ácidos Cumáricos/metabolismo , Desacetilase 6 de Histona/química , Desacetilase 6 de Histona/metabolismo , Inibidores de Histona Desacetilases/síntese química , Inibidores de Histona Desacetilases/metabolismo , Inibidores de Histona Desacetilases/uso terapêutico , Fatores Imunológicos/síntese química , Fatores Imunológicos/metabolismo , Masculino , Melatonina/análogos & derivados , Melatonina/metabolismo , Melatonina/uso terapêutico , Camundongos , Simulação de Acoplamento Molecular , Fármacos Neuroprotetores/síntese química , Fármacos Neuroprotetores/metabolismo , Relação Estrutura-Atividade , Triptaminas/síntese química , Triptaminas/metabolismo
5.
Biomolecules ; 11(2)2021 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-33498722

RESUMO

The HPC-1/syntaxin 1A (Stx1a) gene, which is involved in synaptic transmission and neurodevelopmental disorders, is a TATA-less gene with several transcription start sites. It is activated by the binding of Sp1 and acetylated histone H3 to the -204 to +2 core promoter region (CPR) in neuronal cell/tissue. Furthermore, it is depressed by the association of class 1 histone deacetylases (HDACs) to Stx1a-CPR in non-neuronal cell/tissue. To further clarify the factors characterizing Stx1a gene silencing in non-neuronal cell/tissue not expressing Stx1a, we attempted to identify the promoter region forming DNA-protein complex only in non-neuronal cells. Electrophoresis mobility shift assays (EMSA) demonstrated that the -183 to -137 OL2 promoter region forms DNA-protein complex only in non-neuronal fetal rat skin keratinocyte (FRSK) cells which do not express Stx1a. Furthermore, the Yin-Yang 1 (YY1) transcription factor binds to the -183 to -137 promoter region of Stx1a in FRSK cells, as shown by competitive EMSA and supershift assay. Chromatin immunoprecipitation assay revealed that YY1 in vivo associates to Stx1a-CPR in cell/tissue not expressing Stx1a and that trichostatin A treatment in FRSK cells decreases the high-level association of YY1 to Stx1a-CPR in default. Reporter assay indicated that YY1 negatively regulates Stx1a transcription. Finally, mass spectrometry analysis showed that gene silencing factors, including HDAC1, associate onto the -183 to -137 promoter region together with YY1. The current study is the first to report that Stx1a transcription is negatively regulated in a cell/tissue-specific manner by YY1 transcription factor, which binds to the -183 to -137 promoter region together with gene silencing factors, including HDAC.


Assuntos
Regulação da Expressão Gênica , Inativação Gênica , Histona Desacetilases/genética , Regiões Promotoras Genéticas , Sintaxina 1/biossíntese , Fator de Transcrição YY1/biossíntese , Animais , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Inibidores de Histona Desacetilases/metabolismo , Ácidos Hidroxâmicos/farmacologia , Espectrometria de Massas , Ratos , Proteínas Repressoras/metabolismo
6.
Brain Res ; 1751: 147191, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33152341

RESUMO

The objective of this study was to examine the effect of epigenetic treatment using an histone deacetylases (HDAC) inhibitor in addition to aerobic exercise on the epigenetic markers and neurotrophic gene expressions in the motor cortex, to find a more enriched brain pre-conditioning for motor learning in neurorehabilitation. ICR mice were divided into four groups based on two factors: HDAC inhibition and exercise. Intraperitoneal administration of an HDAC inhibitor (1.2 g/kg sodium butyrate, NaB) and treadmill exercise (approximately at 10 m/min for 60 min) were conducted five days a week for four weeks. NaB administration inhibited total HDAC activity and enhanced acetylation level of histones specifically in histone H4, accompanying the increase of transcription levels of immediate-early genes (IEGs) (c-fos and Arc) and neurotrophins (BDNF and NT-4) crucial for neuroplasticity in the motor cortex. However, exercise enhanced HDAC activity and acetylation level of histone H4 and H3 without the modification of transcription levels. In addition, there were no synergic effects between HDAC inhibition and the exercise regime on the gene expressions. This study showed that HDAC inhibition could present more enriched condition for neuroplasticity to the motor cortex. However, exercise-induced neurotrophic gene expressions could depend on exercise regimen based on the intensity, the term etc. Therefore, this study has a novelty suggesting that pharmacological HDAC inhibition could be an alternative potent approach to present a neuronal platform with enriched neuroplasticity for motor learning and motor recovery, however, an appropriate exercise regimen is expected in this approach.


Assuntos
Ácido Butírico/farmacologia , Plasticidade Neuronal/genética , Condicionamento Físico Animal/fisiologia , Acetilação/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Ácido Butírico/metabolismo , Cognição/fisiologia , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/fisiologia , Feminino , Expressão Gênica/genética , Expressão Gênica/fisiologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Hipocampo/metabolismo , Inibidores de Histona Desacetilases/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Histonas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Córtex Motor/metabolismo , Córtex Motor/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo
7.
Bioorg Chem ; 100: 103934, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32446120

RESUMO

Epigenetics plays a fundamental role in cancer progression, and developing agents that regulate epigenetics is crucial for cancer management. Among Class I and Class II HDACs, HDAC8 is one of the essential epigenetic players in cancer progression. Therefore, we designed, synthesized, purified, and structurally characterized novel compounds containing N-substituted TZD (P1-P25). Cell viability assay of all compounds on leukemic cell lines (CEM, K562, and KCL22) showed the cytotoxic potential of P8, P9, P10, P12, P19, and P25. In-vitro screening of different HDACs isoforms revealed that P19 was the most potent and selective inhibitor for HDAC8 (IC50 - 9.3 µM). Thermal shift analysis (TSA) confirmed the binding of P19 to HDAC8. In-vitro screening of all compounds on the transport activity of GLUT1, GLUT4, and GLUT5 indicated that P19 inhibited GLUT1 (IC50 - 28.2 µM). P10 and P19 induced apoptotic cell death in CEM cells (55.19% and 60.97% respectively) and P19 was less cytotoxic on normal WBCs (CC50 - 104.2 µM) and human fibroblasts (HS27) (CC50 - 105.0 µM). Thus, among this novel series of TZD derivatives, compound P19 was most promising HDAC8 inhibitor and cytotoxic on leukemic cells. Thus, P19 could serve as a lead for further development of optimized molecules with enhanced selectivity and potency.


Assuntos
Inibidores de Histona Desacetilases/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Tiazolidinedionas/química , Apoptose/efeitos dos fármacos , Sítios de Ligação , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Transportador de Glucose Tipo 1/antagonistas & inibidores , Transportador de Glucose Tipo 1/metabolismo , Inibidores de Histona Desacetilases/síntese química , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Humanos , Simulação de Acoplamento Molecular , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Proteínas Repressoras/metabolismo , Relação Estrutura-Atividade , Tiazolidinedionas/metabolismo , Tiazolidinedionas/farmacologia
8.
J Med Chem ; 62(9): 4426-4443, 2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-30964290

RESUMO

The discovery of isozyme-selective histone deacetylase (HDAC) inhibitors is critical for understanding the biological functions of individual HDACs and for validating HDACs as drug targets. The isozyme HDAC10 contributes to chemotherapy resistance and has recently been described to be a polyamine deacetylase, but no studies toward selective HDAC10 inhibitors have been published. Using two complementary assays, we found Tubastatin A, an HDAC6 inhibitor, to potently bind HDAC10. We synthesized Tubastatin A derivatives and found that a basic amine in the cap group was required for strong HDAC10 binding. HDAC10 inhibitors mimicked knockdown by causing dose-dependent accumulation of acidic vesicles in a neuroblastoma cell line. Furthermore, docking into human HDAC10 homology models indicated that a hydrogen bond between a cap group nitrogen and the gatekeeper residue Glu272 was responsible for potent HDAC10 binding. Taken together, our data provide an optimal platform for the development of HDAC10-selective inhibitors, as exemplified with the Tubastatin A scaffold.


Assuntos
Benzamidas/metabolismo , Ácido Glutâmico/química , Inibidores de Histona Desacetilases/metabolismo , Histona Desacetilases/metabolismo , Ácidos Hidroxâmicos/metabolismo , Animais , Benzamidas/síntese química , Benzamidas/química , Transferência Ressonante de Energia de Fluorescência , Células HeLa , Desacetilase 6 de Histona/química , Desacetilase 6 de Histona/metabolismo , Inibidores de Histona Desacetilases/síntese química , Inibidores de Histona Desacetilases/química , Histona Desacetilases/química , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Ácidos Hidroxâmicos/síntese química , Ácidos Hidroxâmicos/química , Ligantes , Simulação de Acoplamento Molecular , Estrutura Molecular , Ligação Proteica , Relação Estrutura-Atividade , Peixe-Zebra
9.
J Med Chem ; 62(8): 3898-3923, 2019 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-30901208

RESUMO

Concurrent inhibition of Janus kinase (JAK) and histone deacetylase (HDAC) could potentially improve the efficacy of the HDAC inhibitors in the treatment of cancers and resolve the problem of HDAC inhibitor resistance in some tumors. Here, a novel series of pyrimidin-2-amino-pyrazol hydroxamate derivatives as JAK and HDAC dual inhibitors was designed, synthesized, and evaluated, among which 8m possessed potent and balanced activities against both JAK2 and HDAC6 with half-maximal inhibitory concentration at the nanomolar level. 8m exhibited improved antiproliferative and proapoptotic activities over SAHA and ruxolitinib in several hematological cell lines. Remarkably, 8m exhibited more potent antiproliferation effect than the combination of SAHA and ruxolitinib in HEL cells bearing JAK2V617F mutation. Pharmacokinetic studies in mice showed that 8m possessed good bioavailability after intraperitoneal administration. Finally, 8m showed antitumor efficacy with no significant toxicity in a HEL xenograft model. Collectively, the results confirm the therapeutic potential of JAK and HDAC dual inhibitors in hematological malignancies and provide valuable leads for further structural optimization and antitumor mechanism study.


Assuntos
Inibidores de Histona Desacetilases/química , Histona Desacetilases/química , Janus Quinases/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Animais , Sítios de Ligação , Domínio Catalítico , Avaliação Pré-Clínica de Medicamentos , Meia-Vida , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/patologia , Inibidores de Histona Desacetilases/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Histona Desacetilases/metabolismo , Humanos , Janus Quinases/metabolismo , Masculino , Camundongos , Camundongos Nus , Simulação de Acoplamento Molecular , Nitrilas , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Pirazóis/química , Pirazóis/metabolismo , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Pirimidinas , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Mol Biol Rep ; 46(2): 2307-2325, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30771146

RESUMO

The important role of histone deacetylases (HDACs) in the development of cancer has been demonstrated by various studies. Thus targeting HDACs with inhibitors is a major focus in anticancer drug research. Although few synthetic HDAC inhibitors (HDIs) have been approved for cancer treatment, they have significant undesirable side effects. Therefore emphases have been placed on natural HDIs as substitutes for the synthetic ones. In a bid to identify more HDIs, this study evaluated the binding tendency of compounds derived from Morinda lucida Benth. towards selected HDACs for the discovery of potent HDIs as potential candidates for anticancer therapeutics, based on the report of anticancer potentials of Morinda lucida-derived extracts and compounds. Givinostat and 49 Morinda-lucida derived compounds were docked against selected HDAC isoforms using AutodockVina, while binding interactions were viewed with Discovery Studio Visualizer, BIOVIA, 2016. Druglikeness and Absorption-Distribution-Metabolism-Excretion (ADME) parameters of the top 7 compounds were evaluated using the Swiss online ADME web tool. The results revealed that out of the 49 compounds, 3 phytosterols (campesterol, cycloartenol, and stigmasterol) and 2 triterpenes (oleanolic acid and ursolic acid) exhibited high HDAC inhibitory activity compared to givinostat. These 5 compounds also fulfill oral drugability of Lipinski rule of five. Morinda lucida-derived phytosterols and triterpenes show high binding tendency towards the selected HDACs and exhibited good drugability characteristics and are therefore good candidates for further studies in the search for therapies against abnormalities linked with over-activity of HDACs.


Assuntos
Inibidores de Histona Desacetilases/isolamento & purificação , Morinda/metabolismo , Morinda/fisiologia , Colesterol/análogos & derivados , Inibidores de Histona Desacetilases/metabolismo , Histona Desacetilases/metabolismo , Humanos , Simulação de Acoplamento Molecular/métodos , Ácido Oleanólico , Fitosteróis/química , Fitosteróis/isolamento & purificação , Extratos Vegetais/farmacologia , Folhas de Planta/metabolismo , Isoformas de Proteínas , Estigmasterol , Triterpenos/classificação , Triterpenos/isolamento & purificação , Ácido Ursólico
11.
Vet Med Sci ; 4(4): 357-363, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30117668

RESUMO

The role of epigenetic alterations during cancer has gained increasing attention and has resulted in a paradigm shift in our understanding of mechanisms leading to cancer susceptibility. Sulforaphane (SFN) is a naturally occurring isothiocyanate derived from the precursor glucosinolate, glucoraphanin (GFN), which is found in cruciferous vegetables such as broccoli. Sulforaphane has been shown to suppress tumour growth by several mechanisms including inhibiting histone deacetylases. The objective of this study was to provide a detailed analysis of sulforaphane absorption following a single oral dose of a broccoli sprout supplement in normal dogs. A single dose of broccoli sprout supplement (with active myrosinase) was orally administered to 10 healthy adult dogs. Blood and urine samples were collected prior to dosing, and at various time points post-dosing. Plasma total SFN metabolite levels peaked at 4 h post-consumption and were cleared by 24 h post-consumption. Urinary SFN metabolites peaked at 4 h post-consumption, and remained detectable at 24 and 48 h post-supplement consumption. A trend for decrease in histone deacetylase activity was observed at 1 h post-consumption and a significant decrease was observed at 24 h post-consumption. The data presented herein indicate that oral SFN is absorbed in dogs, SFN metabolites are detectable in plasma and urine post-dosing, and SFN and its metabolites have some effect on histone deacetylase activity following a single dose.


Assuntos
Brassica/química , Cães , Histona Desacetilases/metabolismo , Isotiocianatos/farmacocinética , Extratos Vegetais/farmacologia , Animais , Cães/sangue , Cães/urina , Inibidores de Histona Desacetilases/metabolismo , Inibidores de Histona Desacetilases/farmacocinética , Inibidores de Histona Desacetilases/farmacologia , Isotiocianatos/metabolismo , Isotiocianatos/farmacologia , Sulfóxidos
12.
Insect Mol Biol ; 27(4): 512-521, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29693770

RESUMO

Apis mellifera plays crucial roles in maintaining the balance of global ecosystems and stability of agricultural systems by helping pollination of flowering plants, including many crops. In recent years, this balance has been disrupted greatly by some pesticides, which results in great losses of honeybees worldwide. Previous studies have found that pesticide-caused memory loss might be one of the major reasons for colony loss. Histone deacetylase inhibitors (HDACis) are chemical compounds that inhibit the activity of histone deacetylases and are known to cause hyperacetylation of histone cores and influence gene expression. In our study, the HDACi sodium butyrate was applied to honeybees as a dietary supplement. The effect of sodium butyrate on the expression profiles of memory-related genes was analysed by quantitative reverse transcription PCR. The results revealed that this HDACi had up-regulation effects on most of the memory-related genes in bees, even in bees treated with imidacloprid. In addition, using the proboscis extension reflex to evaluate olfactory learning in bees, we found that this HDACi boosted the memory formation of bees after impairment owing to imidacloprid exposure. This study investigated the association between gene expression and memory formation from an epigenetic perspective. Additionally, we further demonstrate the possibility of enhancing bee learning using HDACis and provide initial data for future research.


Assuntos
Abelhas/fisiologia , Ácido Butírico/farmacologia , Expressão Gênica , Antagonistas dos Receptores Histamínicos/farmacologia , Inibidores de Histona Desacetilases/metabolismo , Proteínas de Insetos/genética , Memória , Acetilação , Animais , Abelhas/enzimologia , Abelhas/genética , Proteínas de Insetos/metabolismo , Inseticidas/toxicidade , Aprendizagem , Neonicotinoides/toxicidade , Nitrocompostos/toxicidade
13.
J Biomol Struct Dyn ; 36(8): 1966-1978, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28632421

RESUMO

Three series of novel urushiol derivatives were designed by introducing a hydroxamic acid moiety into the tail of an alkyl side chain and substituents with differing electronic properties or steric bulk onto the benzene ring and alkyl side chain. The compounds' binding affinity toward HDAC8 was screened by Glide docking. The highest-scoring compounds were processed further with molecular docking, MD simulations, and binding free energy studies to analyze the binding modes and mechanisms. Ten compounds had Glide scores of -8.2 to -10.2, which revealed that introducing hydroxy, carbonyl, amino, or methyl ether groups into the alkyl side chain or addition of -F, -Cl, sulfonamide, benzamido, amino, or hydroxy substituents on the benzene ring could significantly increase binding affinity. Molecular docking studies revealed that zinc ion coordination, hydrogen bonding, and hydrophobic interactions contributed to the high calculated binding affinities of these compounds toward HDAC8. MD simulations and binding free energy studies showed that all complexes possessed good stability, as characterized by low RMSDs, low RMSFs of residues, moderate hydrogen bonding and zinc ion coordination and low values of binding free energies. Hie147, Tyr121, Phe175, Hip110, Phe119, Tyr273, Lys21, Gly118, Gln230, Leu122, Gly269, and Gly107 contributed favorably to the binding; and Van der Waals and electrostatic interactions provided major contributions to the stability of these complexes. These results show the potential of urushiol derivatives as HDAC8 binding lead compounds, which have great therapeutic potential in the treatment of various malignancies, neurological disorders, and human parasitic diseases.


Assuntos
Catecóis/química , Inibidores de Histona Desacetilases/química , Histona Desacetilases/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas Repressoras/química , Biocatálise/efeitos dos fármacos , Catecóis/metabolismo , Catecóis/farmacologia , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores de Histona Desacetilases/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Ácidos Hidroxâmicos/química , Estrutura Molecular , Ligação Proteica , Conformação Proteica , Proteínas Repressoras/metabolismo , Eletricidade Estática
14.
Sci Rep ; 7(1): 3228, 2017 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-28607401

RESUMO

Histone deacetylases (HDAC) contain eighteen isoforms that can be divided into four classes. Of these isoform enzymes, class IIa (containing HDAC4, 5, 7 and 9) target unique substrates, some of which are client proteins associated with epigenetic control. Class IIa HDACs are reportedly associated with some neuronal disorders, making HDACs therapeutic targets for treating neurodegenerative diseases. Additionally, some reported HDAC inhibitors contain hydroxamate moiety that chelates with zinc ion to become the cofactor of HDAC enzymes. However, the hydroxamate functional group is shown to cause undesirable effects and has poor pharmacokinetic profile. This study used in silico virtual screening methodology to identify several nonhydroxamate compounds, obtained from National Cancer Institute database, which potentially inhibited HDAC4. Comparisons of the enzyme inhibitory activity against a panel of HDAC isoforms revealed these compounds had strong inhibitory activity against class IIa HDACs, but weak inhibitory activity against class I HDACs. Further analysis revealed that a single residue affects the cavity size between class I and class IIa HDACs, thus contributing to the selectivity of HDAC inhibitors discovered in this study. The discovery of these inhibitors presents the possibility of developing new therapeutic treatments that can circumvent the problems seen in traditional hydroxamate-based drugs.


Assuntos
Simulação por Computador , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores de Histona Desacetilases/química , Histona Desacetilases/química , Biocatálise/efeitos dos fármacos , Inibidores de Histona Desacetilases/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Modelos Moleculares , Estrutura Molecular , Ligação Proteica , Domínios Proteicos
15.
SAR QSAR Environ Res ; 28(3): 199-220, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28332438

RESUMO

Histone deacetylases (HDAC) are emerging as promising targets in cancer, neuronal diseases and immune disorders. Computational modelling approaches have been widely applied for the virtual screening and rational design of novel HDAC inhibitors. In this study, different machine learning (ML) techniques were applied for the development of models that accurately discriminate HDAC2 inhibitors form non-inhibitors. The obtained models showed encouraging results, with the global accuracy in the external set ranging from 0.83 to 0.90. Various aspects related to the comparison of modelling techniques, applicability domain and descriptor interpretations were discussed. Finally, consensus predictions of these models were used for screening HDAC2 inhibitors from four chemical libraries whose bioactivities against HDAC1, HDAC3, HDAC6 and HDAC8 have been known. According to the results of virtual screening assays, structures of some hits with pair-isoform-selective activity (between HDAC2 and other HDACs) were revealed. This study illustrates the power of ML-based QSAR approaches for the screening and discovery of potent, isoform-selective HDACIs.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/metabolismo , Relação Quantitativa Estrutura-Atividade , Descoberta de Drogas/métodos , Concentração Inibidora 50 , Aprendizado de Máquina , Estrutura Molecular , Bibliotecas de Moléculas Pequenas
16.
J Gen Virol ; 98(4): 799-809, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28113052

RESUMO

Antiretroviral therapy (ART) can control human immunodeficiency virus-1 (HIV-1) replication in infected individuals. Unfortunately, patients remain persistently infected owing to the establishment of latent infection requiring that ART be maintained indefinitely. One strategy being pursued involves the development of latency-reversing agents (LRAs) to eliminate the latent arm of the infection. One class of molecules that has been tested for LRA activity is the epigenetic modulating compounds histone deacetylases inhibitors (HDACis). Previously, initial screening of these molecules typically commenced using established cell models of viral latency, and although certain drugs such as the HDACi suberoylanilide hydroxamic acid demonstrated strong activity in these models, it did not translate to comparable activity with patient samples. Here we developed a primary cell model of viral latency using primary resting CD4+ T cells infected with Vpx-complemented HIV-1 and found that the activation profile using previously described LRAs mimicked that obtained with patient samples. This primary cell model was used to evaluate 94 epigenetic compounds. Not surprisingly, HDACis were found to be the strongest activators. However, within the HDACi class, the most active LRAs with the least pronounced toxicity contained a benzamide functional moiety with a pyridyl cap group, as exemplified by the HDACi chidamide. The results indicate that HDACis with a benzamide moiety and pyridyl cap group should be considered for further drug development in the pursuit of a successful viral clearance strategy.


Assuntos
Benzamidas/metabolismo , Linfócitos T CD4-Positivos/virologia , Avaliação Pré-Clínica de Medicamentos/métodos , HIV-1/fisiologia , Inibidores de Histona Desacetilases/metabolismo , Ativação Viral/efeitos dos fármacos , Latência Viral/efeitos dos fármacos , Aminopiridinas/química , Aminopiridinas/metabolismo , Benzamidas/química , Células Cultivadas , Inibidores de Histona Desacetilases/química , Humanos
17.
J Chem Inf Model ; 56(1): 173-87, 2016 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-26636371

RESUMO

Silent mating-type information regulation 2 homologue 1 (SIRT1), being the homologous enzyme of silent information regulator-2 gene in yeast, has multifaceted functions. It deacetylates a wide range of histone and nonhistone proteins; hence, it has good therapeutic importance. SIRT1 was believed to be overexpressed in many cancers (prostate, colon) and inflammatory disorders (rheumatoid arthritis). Hence, designing inhibitors against SIRT1 could be considered valuable. Both structure-based and ligand-based drug design strategies were employed to design novel inhibitors utilizing high-throughput virtual screening of chemical databases. An energy-based pharmacophore was generated using the crystal structure of SIRT1 bound with a small molecule inhibitor and compared with a ligand-based pharmacophore model that showed four similar features. A three-dimensional quantitative structure-activity relationship (3D-QSAR) model was developed and validated to be employed in the virtual screening protocol. Among the designed compounds, Lead 17 emerged as a promising SIRT1 inhibitor with IC50 of 4.34 µM and, at nanomolar concentration (360 nM), attenuated the proliferation of prostate cancer cells (LnCAP). In addition, Lead 17 significantly reduced production of reactive oxygen species, thereby reducing pro inflammatory cytokines such as IL6 and TNF-α. Furthermore, the anti-inflammatory potential of the compound was ascertained using an animal paw inflammation model induced by carrageenan. Thus, the identified SIRT1 inhibitors could be considered as potent leads to treat both cancer and inflammation.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Simulação de Acoplamento Molecular , Relação Quantitativa Estrutura-Atividade , Sirtuína 1/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Interface Usuário-Computador , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Humanos , Interleucina-6/metabolismo , Masculino , Conformação Proteica , Ratos , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 1/química , Sirtuína 1/metabolismo , Bibliotecas de Moléculas Pequenas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo
18.
PLoS Negl Trop Dis ; 9(9): e0004026, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26402733

RESUMO

Due to an increasing problem of drug resistance among almost all parasites species ranging from protists to worms, there is an urgent need to explore new drug targets and their inhibitors to provide new and effective parasitic therapeutics. In this regard, there is growing interest in exploring known drug leads of human epigenetic enzymes as potential starting points to develop novel treatments for parasitic diseases. This approach of repurposing (starting with validated targets and inhibitors) is quite attractive since it has the potential to reduce the expense of drug development and accelerate the process of developing novel drug candidates for parasite control. Lysine deacetylases (KDACs) are among the most studied epigenetic drug targets of humans, and a broad range of small-molecule inhibitors for these enzymes have been reported. In this work, we identify the KDAC protein families in representative species across important classes of parasites, screen a compound library of 23 hydroxamate- or benzamide-based small molecules KDAC inhibitors, and report their activities against a range of parasitic species, including the pathogen of malaria (Plasmodium falciparum), kinetoplastids (Trypanosoma brucei and Leishmania donovani), and nematodes (Brugia malayi, Dirofilaria immitis and Haemonchus contortus). Compound activity against parasites is compared to that observed against the mammalian cell line (L929 mouse fibroblast) in order to determine potential parasite-versus-host selectivity). The compounds showed nanomolar to sub-nanomolar potency against various parasites, and some selectivity was observed within the small panel of compounds tested. The possible binding modes of the active compounds at the different protein target sites within different species were explored by docking to homology models to help guide the discovery of more selective, parasite-specific inhibitors. This current work supports previous studies that explored the use of KDAC inhibitors in targeting Plasmodium to develop new anti-malarial treatments, and also pioneers experiments with these KDAC inhibitors as potential new anthelminthics. The selectivity observed begins to address the challenges of targeting specific parasitic diseases while limiting host toxicity.


Assuntos
Antiparasitários/metabolismo , Inibidores de Histona Desacetilases/metabolismo , Histona Desacetilases/metabolismo , Kinetoplastida/enzimologia , Nematoides/enzimologia , Plasmodium falciparum/enzimologia , Animais , Antiparasitários/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Fibroblastos/efeitos dos fármacos , Inibidores de Histona Desacetilases/toxicidade , Histona Desacetilases/química , Kinetoplastida/efeitos dos fármacos , Camundongos , Simulação de Acoplamento Molecular , Nematoides/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Ligação Proteica
19.
J Nutr Biochem ; 25(6): 665-8, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24746830

RESUMO

Sulforaphane is a naturally occurring isothiocyanate in cruciferous vegetables. Sulforaphane inhibits histone deacetylases, leading to the transcriptional activation of genes including tumor suppressor genes. The compound has attracted considerable attention in the chemoprevention of prostate cancer. Here we tested the hypothesis that sulforaphane is not specific for tumor suppressor genes but also activates loci such as long terminal repeats (LTRs), which might impair genome stability. Studies were conducted using chemically pure sulforaphane in primary human IMR-90 fibroblasts and in broccoli sprout feeding studies in healthy adults. Sulforaphane (2.0 µM) caused an increase in LTR transcriptional activity in cultured cells. Consumption of broccoli sprouts (34, 68 or 102 g) by human volunteers caused a dose dependent elevation in LTR mRNA in circulating leukocytes, peaking at more than a 10-fold increase. This increase in transcript levels was associated with an increase in histone H3 K9 acetylation marks in LTR 15 in peripheral blood mononuclear cells from subjects consuming sprouts. Collectively, this study suggests that sulforaphane has off-target effects that warrant further investigation when recommending high levels of sulforaphane intake, despite its promising activities in chemoprevention.


Assuntos
Antineoplásicos Fitogênicos/efeitos adversos , Inibidores de Histona Desacetilases/efeitos adversos , Histonas/metabolismo , Isotiocianatos/efeitos adversos , Pulmão/metabolismo , Sequências Repetidas Terminais , Regulação para Cima , Acetilação , Adulto , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/metabolismo , Brassica/efeitos adversos , Células Cultivadas , Suplementos Nutricionais/efeitos adversos , Feminino , Inibidores de Histona Desacetilases/administração & dosagem , Inibidores de Histona Desacetilases/metabolismo , Humanos , Isotiocianatos/administração & dosagem , Isotiocianatos/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Pulmão/citologia , Masculino , Brotos de Planta/efeitos adversos , Processamento de Proteína Pós-Traducional , RNA Mensageiro/metabolismo , Sulfóxidos , Adulto Jovem
20.
Chem Soc Rev ; 42(23): 8870-94, 2013 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-24030774

RESUMO

The biological activity of selenium is dependent upon its speciation. We aim to integrate selenium speciation and metabolism into a discussion of the mechanisms by which selenium exerts its biological activity. First, we present the current status of selenium in the prevention of cancer, cardiovascular and neurodegenerative diseases with particular attention paid to the results of major chemoprevention trials involving selenium supplementation. A comprehensive review of the current understanding of the metabolism of common dietary selenium compounds - selenite, selenomethionine, methylselenocysteine and selenocystine - is presented, with discussion of the evidence for the various metabolic pathways and their products. The antioxidant, prooxidant and other mechanisms of the dietary selenium compounds have been linked to their disease prevention and treatment properties. The evidence for these various mechanisms -in vitro, in cells and in vivo- is evaluated with emphasis on the selenium metabolites involved. We conclude that dietary selenium compounds should be considered prodrugs, whose biological activity will depend on the activity of the various metabolic pathways in, and the redox status of, cells and tissues. These factors should be considered in future laboratory research and in selecting selenium compounds for trials of disease prevention and treatment by selenium supplementation.


Assuntos
Neoplasias/prevenção & controle , Compostos de Selênio/metabolismo , Ensaios Clínicos como Assunto , Cistina/análogos & derivados , Cistina/metabolismo , Cistina/uso terapêutico , Suplementos Nutricionais , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/metabolismo , Humanos , Compostos Organosselênicos/metabolismo , Compostos Organosselênicos/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Compostos de Selênio/uso terapêutico , Selenocisteína/análogos & derivados , Selenocisteína/metabolismo , Selenocisteína/uso terapêutico , Selenometionina/metabolismo , Selenometionina/uso terapêutico , Selenoproteínas/química , Selenoproteínas/metabolismo , Compostos de Sulfidrila/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA