Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 69
1.
J Biomol Struct Dyn ; 42(11): 5642-5656, 2024 Jul.
Article En | MEDLINE | ID: mdl-38870352

Histone deacetylase 1 (HDAC1), a class I HDAC enzyme, is crucial for histone modification. Currently, it is emerged as one of the important biological targets for designing small molecule drugs through cancer epigenetics. Along with synthetic inhibitors different natural inhibitors are showing potential HDAC1 inhibitions. In order to gain insights into the relationship between the molecular structures of the natural inhibitors and HDAC1, different molecular modelling techniques (Bayesian classification, recursive partitioning, molecular docking and molecular dynamics simulations) have been applied on a dataset of 155 HDAC1 nature-inspired inhibitors with diverse scaffolds. The Bayesian study showed acceptable ROC values for both the training set and test sets. The Recursive partitioning study produced decision tree 1 with 6 leaves. Further, molecular docking study was processed for generating the protein ligand complex which identified some potential amino acid residues such as F205, H28, L271, P29, F150, Y204 for the binding interactions in case of natural inhibitors. Stability of these HDAC1-natutal inhibitors complexes has been also evaluated by molecular dynamics simulation study. The current modelling study is an attempt to get a deep insight into the different important structural fingerprints among different natural compounds modulating HDAC1 inhibition.Communicated by Ramaswamy H. Sarma.


Drug Discovery , Epigenesis, Genetic , Histone Deacetylase 1 , Histone Deacetylase Inhibitors , Molecular Docking Simulation , Molecular Dynamics Simulation , Neoplasms , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Drug Discovery/methods , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/enzymology , Protein Binding , Biological Products/chemistry , Biological Products/pharmacology , Ligands , Bayes Theorem , Structure-Activity Relationship , Binding Sites
2.
Amino Acids ; 55(5): 579-593, 2023 May.
Article En | MEDLINE | ID: mdl-36781452

Histone deacetylases are well-established target enzymes involved in the pathology of different diseases including cancer and neurodegenerative disorders. The approved HDAC inhibitor drugs are associated with cellular toxicities. Different phenolic compounds have been shown to possess inhibitory activities against HDACs and are, therefore, considered safer alternatives to synthetic compounds. Here, we elucidated the binding mode and calculated the binding propensity of some of the top phenolic compounds against different isoforms representing different classes of Zn2+ ion-containing HDACs using the molecular docking approach. Our data reaffirmed the activity of the studied phenolic compounds against HDACs. Binding interaction analysis suggested that these compounds can block the activity of HDACs with or without binding to the active site zinc metal ion. Furthermore, molecular dynamics (MD) simulations were carried out on the selected crystal and docking complexes of each selected HDAC isoform. Analysis of root-mean-square displacement (RMSD) showed that the phenolic compounds demonstrated a stable binding mode over 50 ns in a way that is comparable to the cocrystal ligands. Together, these findings can aid future efforts in the search for natural inhibitors of HDACs.


Histone Deacetylase Inhibitors , Molecular Dynamics Simulation , Molecular Docking Simulation , Histone Deacetylase Inhibitors/pharmacology , Protein Isoforms/chemistry , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism
3.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Article En | MEDLINE | ID: mdl-34815344

Carriers of heterozygous germline BAP1 mutations (BAP1+/-) are affected by the "BAP1 cancer syndrome." Although they can develop almost any cancer type, they are unusually susceptible to asbestos carcinogenesis and mesothelioma. Here we investigate why among all carcinogens, BAP1 mutations cooperate with asbestos. Asbestos carcinogenesis and mesothelioma have been linked to a chronic inflammatory process promoted by the extracellular release of the high-mobility group box 1 protein (HMGB1). We report that BAP1+/- cells secrete increased amounts of HMGB1, and that BAP1+/- carriers have detectable serum levels of acetylated HMGB1 that further increase when they develop mesothelioma. We linked these findings to our discovery that BAP1 forms a trimeric protein complex with HMGB1 and with histone deacetylase 1 (HDAC1) that modulates HMGB1 acetylation and its release. Reduced BAP1 levels caused increased ubiquitylation and degradation of HDAC1, leading to increased acetylation of HMGB1 and its active secretion that in turn promoted mesothelial cell transformation.


Asbestos , HMGB1 Protein/chemistry , Histone Deacetylase 1/chemistry , Tumor Suppressor Proteins/chemistry , Ubiquitin Thiolesterase/chemistry , Animals , Biomarkers, Tumor/metabolism , Carcinogenesis , Cell Nucleus/metabolism , Female , Gene-Environment Interaction , Germ-Line Mutation , HMGB1 Protein/genetics , Heterozygote , Histone Deacetylase 1/genetics , Incidence , Inflammation , Male , Mesothelioma/metabolism , Mice , Mutation , Prognosis , Protein Binding , Tumor Suppressor Proteins/metabolism , Ubiquitin/chemistry , Ubiquitin Thiolesterase/metabolism
4.
Methods Mol Biol ; 2266: 155-170, 2021.
Article En | MEDLINE | ID: mdl-33759126

Medicinal chemistry society has enough arguments to justify the usage of fragment-based drug design (FBDD) methodologies for the identification of lead compounds. Since the FDA approval of three kinase inhibitors - vemurafenib, venetoclax, and erdafitinib, FBDD has become a challenging alternative to high-throughput screening methods in drug discovery. The following protocol presents in silico drug design of selective histone deacetylase 6 (HDAC6) inhibitors through a fragment-based approach. To date, structural motifs that are important for HDAC inhibitory activity and selectivity are described as: surface recognition group (CAP group), aliphatic or aromatic linker, and zinc-binding group (ZBG). The main idea of this FBDD method is to identify novel and target-selective CAP groups by virtual scanning of publicly available fragment databases. Template structure used to search for novel heterocyclic and carbocyclic fragments is 1,8-naphthalimide (CAP group of scriptaid, a potent HDAC inhibitor). Herein, the design of HDAC6 inhibitors is based on linking the identified fragments with the aliphatic or aromatic linker and hydroxamic acid (ZBG) moiety. Final selection of potential selective HDAC6 inhibitors is based on combined structure-based (molecular docking) and ligand-based (three-dimensional quantitative structure-activity relationships, 3D-QSAR) techniques. Designed compounds are docked in the active site pockets of human HDAC1 and HDAC6 isoforms, and their docking conformations used to predict their HDAC inhibitory and selectivity profiles through two developed 3D-QSAR models (describing HDAC1 and HDAC6 inhibitory activities).


Drug Discovery/methods , Histone Deacetylase 6/chemistry , Histone Deacetylase Inhibitors/chemistry , Molecular Docking Simulation/methods , Naphthalimides/chemistry , Amino Acid Motifs , Catalytic Domain , Databases, Chemical , Drug Design , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/chemistry , Histone Deacetylase 6/antagonists & inhibitors , In Vitro Techniques , Ligands , Molecular Conformation , Molecular Dynamics Simulation , Quantitative Structure-Activity Relationship , Small Molecule Libraries , Structure-Activity Relationship
5.
Int J Mol Sci ; 21(16)2020 Aug 16.
Article En | MEDLINE | ID: mdl-32824279

N-(2'-hydroxyphenyl)-2-propylpentanamide (HO-AAVPA) is a VPA derivative designed to be a histone deacetylase (HDAC) inhibitor. HO-AAVPA has better antiproliferative effect than VPA in cancer cell lines. Therefore, in this work, the inhibitory effect of HO-AAVPA on HDAC1, HDAC6, and HDAC8 was determined by in silico and in vitro enzymatic assay. Furthermore, its antiproliferative effect on the cervical cancer cell line (SiHa) and the translocation of HMGB1 and ROS production were evaluated. The results showed that HO-AAVPA inhibits HDAC1, which could be related with HMGB1 translocation from the nucleus to the cytoplasm due to HDAC1 being involved in the deacetylation of HMGB1. Furthermore, an increase in ROS production was observed after the treatment with HO-AAVPA, which also could contribute to HMGB1 translocation. Therefore, the results suggest that one of the possible antiproliferative mechanisms of HO-AAVPA is by HDAC1 inhibition which entails HMGB1 translocation and ROS increased levels that could trigger the cell apoptosis.


Amides/pharmacology , Antineoplastic Agents/pharmacology , HMGB1 Protein/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Pentanes/pharmacology , Uterine Cervical Neoplasms/metabolism , Active Transport, Cell Nucleus/drug effects , Amides/chemistry , Antineoplastic Agents/chemistry , Binding Sites , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Female , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/chemistry , Histone Deacetylase Inhibitors/chemistry , Humans , Molecular Docking Simulation , Pentanes/chemistry , Protein Binding
6.
Anal Chem ; 92(12): 8031-8036, 2020 06 16.
Article En | MEDLINE | ID: mdl-32420730

Isoprenoid pyrophosphates are involved in protein prenylation and assume regulatory roles in cells; however, little is known about the cellular proteins that can interact with isoprenoid pyrophosphates. Here, we devised a chemical proteomic strategy, capitalizing on the use of a desthiobiotin-geranyl pyrophosphate (GPP) acyl phosphate probe for the enrichment and subsequent identification of GPP-binding proteins using liquid chromatography-tandem mass spectrometry (LC-MS/MS). By combining stable isotope labeling by amino acids in cell culture (SILAC) and competitive labeling with low vs high concentrations of GPP probe, with ATP vs GPP acyl phosphate probes, or with the GPP probe in the presence of different concentrations of free GPP, we uncovered a number of candidate GPP-binding proteins. We also discovered, for the first time, histone deacetylase 1 (HDAC1) as a GPP-binding protein. Furthermore, we found that the enzymatic activity of HDAC1 could be modulated by isoprenoid pyrophosphates. Together, we developed a novel chemical proteomic method for the proteome-wide discovery of GPP-binding proteins, which sets the stage for a better understanding about the biological functions of isoprenoids.


Biotin/analogs & derivatives , Histone Deacetylase 1/chemistry , Polyisoprenyl Phosphates/chemistry , Proteomics , Biotin/chemistry , Histone Deacetylase 1/metabolism , Humans , Molecular Structure
7.
FEBS Lett ; 594(14): 2322-2330, 2020 07.
Article En | MEDLINE | ID: mdl-32391601

The Sin3L/Rpd3L histone deacetylase (HDAC) complex is one of six major HDAC complexes in the nucleus, and its recruitment by promoter-bound transcription factors is an important step in many gene transcription regulatory pathways. Here, we investigate how the Myt1L zinc finger transcription factor, important for neuronal differentiation and the maintenance of neuronal identity, recruits this complex at the molecular level. We show that Myt1L, through a highly conserved segment shared with its paralogs, interacts directly and specifically with the Sin3 PAH1 domain, binding principally to the canonical hydrophobic cleft found in paired amphipathic helix domain (PAH) domains. Our findings are relevant not only for other members of the Myt family but also for enhancing our understanding of the rules of protein-protein interactions involving Sin3 PAH domains.


Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism , Nerve Tissue Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/chemistry , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Transcription Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Humans , Mice , Models, Molecular , Protein Binding , Protein Domains
8.
J Med Chem ; 63(11): 5734-5751, 2020 06 11.
Article En | MEDLINE | ID: mdl-32392053

Leishmania (L.) infantum causes visceral, cutaneous, and mucosal leishmaniasis in humans and canine leishmaniasis in dogs. Herein, we describe that O-alkyl hydroxamate derivatives displayed potent and selective in vitro activity against the amastigote stage of L. infantum while no activity was observed against promastigotes. Compound 5 showed potent in vivo activity against L. infantum. Moreover, the combination of compound 5 supported on gold nanoparticles and meglumine antimoniate was also effective in vivo and improved the activity of these compounds compared to that of the individual treatment. Docking studies showed that compound 5 did not reach highly conserved pocket C and established interactions with the semiconserved residues V44, A45, R242, and E243 in pocket A of LiSIR2rp1. The surface space determined by these four amino acids is not conserved in human sirtuins. Compound 5 represents a new class of selective ligands with antileishmanial activity.


Antiprotozoal Agents/pharmacology , Hydroxamic Acids/chemistry , Leishmania infantum/drug effects , Animals , Antiprotozoal Agents/chemistry , Binding Sites , Female , Gold/chemistry , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism , Humans , Hydroxamic Acids/pharmacology , Leishmania infantum/growth & development , Life Cycle Stages/drug effects , Meglumine Antimoniate/pharmacology , Metal Nanoparticles/chemistry , Mice , Mice, Inbred BALB C , Microsomes, Liver/metabolism , Molecular Docking Simulation , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Spleen/parasitology
9.
J Med Chem ; 63(9): 4701-4715, 2020 05 14.
Article En | MEDLINE | ID: mdl-32267687

While proteasome inhibitors such as bortezomib showed satisfactory clinical benefits in the initial treatment of multiple myeloma (MM), drug resistance and relapse are unavoidable. Recent studies suggested inhibition of histone deacetylases (HDACs) restored sensitivity of bortezomib-resistant MM. Hence, we designed dual inhibitors targeting both HDACs and proteasomes to address the resistance of bortezomib. The most potent inhibitors, ZY-2 and ZY-13 showed excellent inhibition against proteasome and good selectivity against HDACs. In particular, ZY-2 not only exhibited good antiproliferative activities on the MM cell lines RPMI-8226, U266, and KM3 (IC50 values of 6.66, 4.31, and 10.1 nM, respectively) but also showed more potent antiproliferative activities against the bortezomib-resistant MM cell line KM3/BTZ compared with bortezomib (IC50 values of 8.98 vs. 226 nM, P < 0.01) and even better than the combination of the HDAC inhibitor MS-275 and bortezomib (1:1) (IC50 values of 8.98 vs. 98.0 nM, P < 0.01).


Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Drug Resistance, Neoplasm/drug effects , Histone Deacetylase Inhibitors/pharmacology , Peptides/pharmacology , Proteasome Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Boronic Acids/chemical synthesis , Boronic Acids/metabolism , Boronic Acids/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Discovery , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/toxicity , Humans , Molecular Docking Simulation , Multiple Myeloma/drug therapy , Peptides/chemical synthesis , Peptides/metabolism , Peptides/toxicity , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemical synthesis , Proteasome Inhibitors/metabolism , Proteasome Inhibitors/toxicity , Protein Binding , Protein Subunits/chemistry , Protein Subunits/metabolism , S Phase Cell Cycle Checkpoints/drug effects
10.
J Comput Aided Mol Des ; 34(8): 857-878, 2020 08.
Article En | MEDLINE | ID: mdl-32180123

Valproic acid (VPA) is a compound currently used in clinical practice for the treatment of epilepsy as well as bipolar and mood disorders. VPA targets histone deacetylases (HDACs), which participate in the removal of acetyl groups from lysine in several proteins, regulating a wide variety of functions within the organism. An imbalance or malfunction of these enzymes is associated with the development and progression of several diseases, such as cancer and neurodegenerative diseases. HDACs are divided into four classes, but VPA only targets Class I (HDAC1-3 and 8) and Class IIa (HDAC4-5, 7 and 9) HDACs; however, structural and energetic information regarding the manner by which VPA inhibits these HDACs is lacking. Here, the structural and energetic features that determine this recognition were studied using molecular docking and molecular dynamics (MD) simulation. It was found that VPA reaches the catalytic site in HDAC1-3 and 7, whereas in HDAC6, VPA only reaches the catalytic tunnel. In HDAC4, VPA was bound adjacent to L1 and L2, a zone that participates in corepressor binding, and in HDAC8, VPA was bound to the hydrophobic active site channel (HASC), in line with previous reports.


Histone Deacetylase Inhibitors/pharmacology , Molecular Docking Simulation/methods , Valproic Acid/pharmacology , Crystallography, X-Ray , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism , Histone Deacetylase 6/chemistry , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemistry , Humans , Molecular Dynamics Simulation , Principal Component Analysis , Protein Conformation , Reproducibility of Results , Valproic Acid/chemistry
11.
J Biol Chem ; 295(51): 17738-17751, 2020 12 18.
Article En | MEDLINE | ID: mdl-33454011

Distinct cell types emerge from embryonic stem cells through a precise and coordinated execution of gene expression programs during lineage commitment. This is established by the action of lineage specific transcription factors along with chromatin complexes. Numerous studies have focused on epigenetic factors that affect embryonic stem cells (ESC) self-renewal and pluripotency. However, the contribution of chromatin to lineage decisions at the exit from pluripotency has not been as extensively studied. Using a pooled epigenetic shRNA screen strategy, we identified chromatin-related factors critical for differentiation toward mesodermal and endodermal lineages. Here we reveal a critical role for the chromatin protein, ARID4B. Arid4b-deficient mESCs are similar to WT mESCs in the expression of pluripotency factors and their self-renewal. However, ARID4B loss results in defects in up-regulation of the meso/endodermal gene expression program. It was previously shown that Arid4b resides in a complex with SIN3A and HDACS 1 and 2. We identified a physical and functional interaction of ARID4B with HDAC1 rather than HDAC2, suggesting functionally distinct Sin3a subcomplexes might regulate cell fate decisions Finally, we observed that ARID4B deficiency leads to increased H3K27me3 and a reduced H3K27Ac level in key developmental gene loci, whereas a subset of genomic regions gain H3K27Ac marks. Our results demonstrate that epigenetic control through ARID4B plays a key role in the execution of lineage-specific gene expression programs at pluripotency exit.


DNA-Binding Proteins/metabolism , Animals , Cell Differentiation , Cell Line , Cell Lineage , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Endoderm/cytology , Endoderm/metabolism , Epigenesis, Genetic , Gene Expression Regulation , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/chemistry , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Histones/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/chemistry , Sin3 Histone Deacetylase and Corepressor Complex/metabolism
12.
Adv Biol Regul ; 75: 100667, 2020 01.
Article En | MEDLINE | ID: mdl-31648945

The higher-order inositol phosphate second messengers inositol tetrakisphosphate (IP4), inositol pentakisphosphate (IP5) and inositol hexakisphosphate (IP6) are important signaling molecules that regulate DNA-damage repair, cohesin dynamics, RNA-editing, retroviral assembly, nuclear transport, phosphorylation, acetylation, crotonylation, and ubiquitination. This functional diversity has made understanding how inositol polyphosphates regulate cellular processes challenging to dissect. However, some inositol phosphates have been unexpectedly found in X-ray crystal structures, occasionally revealing structural and mechanistic details of effector protein regulation before functional consequences have been described. This review highlights a sampling of crystal structures describing the interaction between inositol phosphates and protein effectors. This list includes the RNA editing enzyme "adenosine deaminase that acts on RNA 2" (ADAR2), the Pds5B regulator of cohesin dynamics, the class 1 histone deacetylases (HDACs) HDAC1 and HDAC3, and the PH domain of Bruton's tyrosine kinase (Btk). One of the most important enzymes responsible for higher-order inositol phosphate synthesis is inositol polyphosphate multikinase (IPMK), which plays dual roles in both inositol and phosphoinositide signaling. Structures of phosphoinositide lipid binding proteins have also revealed new aspects of protein effector regulation, as mediated by the nuclear receptors Steroidogenic Factor-1 (SF-1, NR5A2) and Liver Receptor Homolog-1 (LRH-1, NR5A2). Together, these studies underscore the structural diversity in binding interactions between effector proteins and inositol phosphate small signaling molecules, and further support that detailed structural studies can lead to new biological discovery.


Inositol Phosphates/chemistry , Second Messenger Systems , Adenosine Deaminase/chemistry , Adenosine Deaminase/metabolism , Agammaglobulinaemia Tyrosine Kinase/chemistry , Agammaglobulinaemia Tyrosine Kinase/metabolism , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Humans , Inositol Phosphates/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
13.
Sci Rep ; 9(1): 13187, 2019 Sep 12.
Article En | MEDLINE | ID: mdl-31515509

To develop novel CNS penetrant HDAC inhibitors, a new series of HDAC inhibitors having benzoheterocycle were designed, synthesized, and biologically evaluated. Among the synthesized compounds, benzothiazole derivative 9b exhibited a remarkable anti-proliferative activity (GI50 = 2.01 µM) against SH-SY5Y cancer cell line in a dose and time-dependent manner, better than the reference drug SAHA (GI50 = 2.90 µM). Moreover, compound 9b effectively promoted the accumulation of acetylated Histone H3 and α-tubulin through inhibition of HDAC1 and HDAC6 enzymes, respectively. HDAC enzyme assay also confirmed that compound 9b efficiently inhibited HDAC1 and HDAC6 isoforms with IC50 values of 84.9 nM and 95.9 nM. Furthermore, compound 9b inhibited colony formation capacity of SH-SY5Y cells, which is considered a hallmark of cell carcinogenesis and metastatic potential. The theoretical prediction, in vitro PAMPA-BBB assay, and in vivo brain pharmacokinetic studies confirmed that compound 9b had much higher BBB permeability than SAHA. In silico docking study demonstrated that compound 9b fitted in the substrate binding pocket of HDAC1 and HDAC6. Taken together, compound 9b provided a novel scaffold for developing CNS penetrant HDAC inhibitors and therapeutic potential for CNS-related diseases.


Amyloid beta-Peptides/chemistry , Histone Deacetylase 1 , Histone Deacetylase 6 , Histone Deacetylase Inhibitors , Molecular Docking Simulation , Neoplasm Proteins , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/chemistry , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase 6/chemistry , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasms/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism
14.
Molecules ; 24(14)2019 Jul 15.
Article En | MEDLINE | ID: mdl-31311163

A series of novel coumarin-based hydroxamate derivatives were designed and synthesized as histone deacetylase inhibitors (HDACis). Selective compounds showed a potent HDAC inhibition with nM IC50 values, with the best compound (10e) being nearly 90 times more active than vorinostat (SAHA) against HDAC1. Compounds 10e and 11d also increased the levels of acetylated histone H3 and H4, which is consistent with their strong HDAC inhibition. In addition, 10e and 11d displayed a higher potency toward human A549 and Hela cancer cell lines compared with SAHA. Moreover, 10e and 11d significantly arrested A549 cells at the G2/M phase and enhanced apoptosis. Molecular docking studies revealed the possible mode of interaction of compounds 10e and 12a with HDAC1. Our findings suggest that these novel coumarin-based HDAC inhibitors provide a promising scaffold for the development of new potential cancer chemotherapies.


Antineoplastic Agents/chemical synthesis , Coumarins/chemical synthesis , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Hydroxamic Acids/chemical synthesis , A549 Cells , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Coumarins/chemistry , Coumarins/pharmacology , Drug Design , Drug Screening Assays, Antitumor , HeLa Cells , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/chemistry , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Molecular Docking Simulation , Structure-Activity Relationship
15.
Eur J Med Chem ; 177: 457-466, 2019 Sep 01.
Article En | MEDLINE | ID: mdl-31181405

Histone deacetylases (HDACs) play an important role in cancer, degenerative diseases and inflammation. The currently applied HDAC inhibitors in the clinic lack selectivity among HDAC isoforms, which limits their application for novel indications such as inflammatory diseases. Recent, literature indicates that HDAC 3 plays an important role among class I HDACs in gene expression in inflammation. In this perspective, the development and understanding of inhibitory selectivity among HDACs 1, 2 and 3 and their respective influence on gene expression need to be characterized to facilitate drug discovery. Towards this aim, we synthesized nine structural analogues of the class I HDAC inhibitor Entinostat and investigated their selectivity profile among HDACs 1, 2 and 3. We found that we can explain the observed structure activity relationships by small structural and conformational differences between HDAC 1 and HDAC 3 in the 'lid' interacting region. Cell-based studies indicated, however, that application of inhibitors with improved HDAC 3 selectivity did not provide an anti-inflammatory response in contrast to expectations from biochemical evidence in literature. Altogether, in this study, we identified structure activity relationships among class I HDACs and we connected isoform selectivity among class I HDACs with pro- and anti-inflammatory gene transcription in macrophages.


Anilides/pharmacology , Benzamides/pharmacology , Gene Expression/drug effects , Histone Deacetylase Inhibitors/pharmacology , Macrophages/drug effects , Anilides/chemical synthesis , Anilides/chemistry , Anilides/metabolism , Animals , Benzamides/chemical synthesis , Benzamides/chemistry , Benzamides/metabolism , Catalytic Domain , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Humans , Inflammation/genetics , Interleukin-10/genetics , Interleukin-6/genetics , Mice , Molecular Docking Simulation , NF-kappa B p50 Subunit/metabolism , Nitric Oxide Synthase Type II/genetics , Protein Binding , RAW 264.7 Cells , Stereoisomerism , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/genetics
16.
Eur J Med Chem ; 178: 116-130, 2019 Sep 15.
Article En | MEDLINE | ID: mdl-31177073

In this study, a series of novel HDAC inhibitors, using 1,2,4-oxadiazole-containing as the cap group, were synthesized and evaluated in vitro. Compound 14b, N-hydroxy-2-(methyl((3-(1-(4-methylbenzyl)piperidin-4-yl)-1,2,4-oxadiazol-5-yl)methyl)amino)pyrimidine-5-carboxamide, displayed the most potent histone deacetylase (HDAC) inhibition, especially against HDAC1, 2, and 3 with IC50 values of 1.8, 3.6 and 3.0 nM, respectively. In vitro antiproliferative studies confirmed that 14b was more potent than SAHA, with IC50 values against 12 types of cancer cell lines ranging from 9.8 to 44.9 nM. The results of Western blot assays showed that compound 14b can significantly up-regulate the acetylation of the biomarker his-H3 and molecular docking analyses revealed the mode of action of compound 14b against HDAC1. The results of flow-cytometry analysis suggested that compound 14b induces cell cycle arrest at the G1 phase and has apoptotic effects. Further investigation of the activity of 14b on the primary cells of three patients, showed IC50 values of 21.3, 61.1, and 77.4 nM. More importantly, an oral bioavailability of up to 53.52% was observed for 14b. An in vivo pharmacodynamic evaluation demonstrated that compound 14b can significantly inhibit tumor growth in a Daudi Burkitt's lymphoma xenograft model, with tumor inhibition rates of 53.8 and 46.1% observed at 20 and 10 mg/kg when administered p.o. and i.v., respectively. These results indicate that compound 14b may be a suitable lead for further evaluation and development as an HDAC inhibitor and a potent anticancer agent.


Antineoplastic Agents/therapeutic use , Burkitt Lymphoma/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Oxadiazoles/therapeutic use , Acetylation/drug effects , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Discovery , Drug Screening Assays, Antitumor , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/pharmacokinetics , Histones/metabolism , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacokinetics , Mice, Inbred NOD , Mice, SCID , Molecular Docking Simulation , Molecular Structure , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacokinetics , Protein Binding , Rats, Sprague-Dawley , Structure-Activity Relationship , Tubulin/metabolism , Xenograft Model Antitumor Assays
17.
J Nat Prod ; 82(6): 1442-1450, 2019 06 28.
Article En | MEDLINE | ID: mdl-31120744

Naturally occurring ß-carbolines are known to have antitumor activities but with limited effectiveness. In order to improve their efficacy, a series of new hydroxamic-acid-containing ß-carbolines connected via a hydroxycinnamic acid moitey (12a-f) were developed to incorporate histone deacetylase (HDAC) inhibition for possible synergistic effects. When evaluated in in vitro assays, most of the analogues showed significant antitumor activities against four human cancer cells. In particular, 12b showed the highest cytotoxic potency of the series, including drug-resistant Bel7402 cells, but had minimal effect on normal hepatic LO2 cells. These compounds also showed excellent inhibitory effects against HDAC1/6, which appear to contribute greatly to their antiproliferative properties. Compound 12b enhanced the acetylation levels of histone H3 and α-tubulin and induced greater cancer cell apoptosis than the FDA-approved HDAC inhibitor SAHA by regulating expression of apoptotic proteins Bax, Bcl-2, and caspase 3. Importantly, 12b also induced a significant amount of autophagic flux activity in Bel7402 cells by increasing the expression of Beclin-1 and LC3-II proteins and decreasing that of LC3-I and p62. Finally, 12b significantly inhibited PI3K/Akt/mTOR signaling, an important cell-growth-promoting pathway aberrantly activated in many cancers. Together, the results suggest that these hydroxamic-acid-containing ß-carboline derivatives may be new leads for the discovery of agents for the treatment of human carcinoma cancers.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Beclin-1/chemistry , Carbolines/pharmacology , Caspase 3/pharmacology , Cell Proliferation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/chemistry , TOR Serine-Threonine Kinases/metabolism , Acetylation , Antineoplastic Agents/chemistry , Carbolines/chemistry , Caspase 3/chemistry , Cell Line, Tumor , Coumaric Acids , Histone Deacetylase 1/chemistry , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/chemistry , Humans , Hydroxamic Acids/chemistry , Molecular Structure , Phosphatidylinositol 3-Kinases/chemistry , Proto-Oncogene Proteins c-akt/chemistry , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/chemistry , bcl-2-Associated X Protein/chemistry , bcl-2-Associated X Protein/metabolism
18.
ACS Chem Neurosci ; 10(5): 2467-2480, 2019 05 15.
Article En | MEDLINE | ID: mdl-30784262

Histone deacetylase 6 (HDAC6) plays a key role in a variety of neurological disorders, which makes it attractive drug target for the treatment of Alzheimer's disease, Parkinson's disease, and memory/learning impairment. The selectivity of HDAC6 inhibitors (sHDAC6Is) are widely considered to be susceptible to the sizes of their Cap group and the physicochemical properties of their linker or zinc-binding group, which makes the discovery of new sHDAC6Is extremely difficult. With the discovery of the distinct selectivity between Trichostatin A (TSA) enantiomers, the chirality residing in the connective units between TSA's Cap and linker shows a great impact on its selectivity. However, the mechanism underlining ( S)-TSA's selectivity is still elusive, and the way chirality switches the selective ( S)-TSA to nonselective ( R)-TSA is unknown. In this study, multiple computational approaches were collectively applied to explore, validate, and differentiate the binding modes of two TSA enantiomers in HDACs (especially the HDAC6) at atomic level. First, two nonconservative residues (G200/M205 and Y197/F202 in HDAC1/6) in loop3 and four conservative residues deep inside the hydrophobic binding pocket were discovered as the decisive residues of ( S)-TSA's selectivity toward HDAC6. Then, a novel mechanism underlying the selectivity of ( S)-TSA toward HDAC6 was proposed, which was composed of the trigger by two nonconservative residues F202 and M205 in HDAC6 and a subsequently improved fit of ( S)-TSA deep inside HDAC6's hydrophobic binding pocket. TSA enantiomers were used as a molecular probe to explore the mechanism underlying sHDAC6Is' selectivity in this study. Because of their decisive roles in ( S)-TSA's selectivity to HDAC6, both F202 and M205 in HDAC6 should be especially considered in the discovery of novel sHDAC6Is.


Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Neurodegenerative Diseases/physiopathology , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/chemistry , Histone Deacetylase 6/chemistry , Humans , Molecular Dynamics Simulation , Protein Conformation
19.
Chembiochem ; 20(11): 1444-1449, 2019 06 03.
Article En | MEDLINE | ID: mdl-30701667

Histone deacetylase 1 (HDAC1) regulates transcription by deacetylating histones. In addition to histones, several non-histone proteins are HDAC1 substrates, which suggests a role for HDAC1 beyond epigenetics. Unfortunately, the identification of non-histone substrates has been largely serendipitous, which makes full characterization of HDAC1 functions difficult. To overcome this challenge, inactive "trapping" mutants were recently developed to identify HDAC1 substrates. To optimize substrate trapping, the relative trapping abilities of 17 inactive HDAC1 mutants was assessed. HDAC1 H141A, F150A, and C151A showed strong binding to substrates LSD1 and p53. Interestingly, each mutant preferentially trapped a different substrate. By combining several inactive mutants, the trapping strategy will facilitate the discovery of new HDAC1 substrates and shed light on the variety of HDAC1-related functions in cell biology.


Histone Deacetylase 1/chemistry , Histone Demethylases/metabolism , Tumor Suppressor Protein p53/metabolism , Epigenesis, Genetic , HEK293 Cells , Histone Deacetylase 1/genetics , Histones/metabolism , Humans , Mutation , Protein Binding , Substrate Specificity
20.
Exp Parasitol ; 198: 7-16, 2019 Mar.
Article En | MEDLINE | ID: mdl-30682336

Plasmodium falciparum histone deacetylases (PfHDACs) are an important class of epigenetic regulators that alter protein lysine acetylation, contributing to regulation of gene expression and normal parasite growth and development. PfHDACs are therefore under investigation as drug targets for malaria. Despite this, our understanding of the biological roles of these enzymes is only just beginning to emerge. In higher eukaryotes, HDACs function as part of multi-protein complexes and act on both histone and non-histone substrates. Here, we present a proteomics analysis of PfHDAC1 immunoprecipitates, identifying 26 putative P. falciparum complex proteins in trophozoite-stage asexual intraerythrocytic parasites. The co-migration of two of these (P. falciparum heat shock proteins 70-1 and 90) with PfHDAC1 was validated using Blue Native PAGE combined with Western blot. These data provide a snapshot of possible PfHDAC1 interactions and a starting point for future studies focused on elucidating the broader function of PfHDACs in Plasmodium parasites.


Histone Deacetylase 1/analysis , Plasmodium falciparum/enzymology , Proteomics , Protozoan Proteins/chemistry , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Histone Deacetylase 1/chemistry , Immunoprecipitation , Mass Spectrometry/methods
...