51
|
Identification of novel potential scaffold for class I HDACs inhibition: An in-silico protocol based on virtual screening, molecular dynamics, mathematical analysis and machine learning. Biochem Biophys Res Commun 2017; 491:800-806. [PMID: 28705738 DOI: 10.1016/j.bbrc.2017.07.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 07/09/2017] [Indexed: 12/25/2022]
Abstract
Histone deacetylases (HDACs) family has been widely reported as an important class of enzyme targets for cancer therapy. Much effort has been made in discovery of novel scaffolds for HDACs inhibition besides existing hydroxamic acids, cyclic peptides, benzamides, and short-chain fatty acids. Herein we set up an in-silico protocol which not only could detect potential Zn2+ chelation bonds but also still adopted non-bonded model to be effective in discovery of Class I HDACs inhibitors, with little human's subjective visual judgment involved. We applied the protocol to screening of Chembridge database and selected out 7 scaffolds, 3 with probability of more than 99%. Biological assay results demonstrated that two of them exhibited HDAC-inhibitory activity and are thus considerable for structure modification to further improve their bio-activity.
Collapse
|
52
|
HDAC Inhibitor-Induced Mitotic Arrest Is Mediated by Eg5/KIF11 Acetylation. Cell Chem Biol 2017; 24:481-492.e5. [PMID: 28392145 DOI: 10.1016/j.chembiol.2017.03.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/08/2016] [Accepted: 03/10/2017] [Indexed: 11/24/2022]
Abstract
Histone deacetylase 1 (HDAC1) is an epigenetic enzyme that regulates key cellular processes, such as cell proliferation, apoptosis, and cell survival, by deacetylating histone substrates. Aberrant expression of HDAC1 is implicated in multiple diseases, including cancer. As a consequence, HDAC inhibitors have emerged as effective anti-cancer drugs. HDAC inhibitor-induced G0/G1 cell-cycle arrest has been attributed to epigenetic transcriptional changes mediated by histone acetylation. However, the mechanism of G2/M arrest remains poorly understood. Here, we identified mitosis-related protein Eg5 (KIF11) as an HDAC1 substrate using a trapping mutant strategy. HDAC1 colocalized with Eg5 during mitosis and influenced the ATPase activity of Eg5. Importantly, an HDAC1- and HDAC2-selective inhibitor caused mitotic arrest and monopolar spindle formation, consistent with a model in which Eg5 deacetylation by HDAC1 is critical for mitotic progression. These findings revealed a previously unknown mechanism of action of HDAC inhibitors involving Eg5 acetylation, and provide a compelling mechanistic hypothesis for HDAC inhibitor-mediated G2/M arrest.
Collapse
|
53
|
Mangiatordi GF, Alberga D, Pisani L, Gadaleta D, Trisciuzzi D, Farina R, Carotti A, Lattanzi G, Catto M, Nicolotti O. A rational approach to elucidate human monoamine oxidase molecular selectivity. Eur J Pharm Sci 2017; 101:90-99. [DOI: 10.1016/j.ejps.2017.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/27/2017] [Accepted: 02/05/2017] [Indexed: 02/05/2023]
|
54
|
Dines JA, Marson CM. A direct alkylation route to branched derivatives of suberoylanilide hydroxamic acid (SAHA), a potent non-selective inhibitor of histone deacetylases. Tetrahedron 2016. [DOI: 10.1016/j.tet.2016.11.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
55
|
Bieliauskas AV, Weerasinghe SVW, Negmeldin AT, Pflum MKH. Structural Requirements of Histone Deacetylase Inhibitors: SAHA Analogs Modified on the Hydroxamic Acid. Arch Pharm (Weinheim) 2016; 349:373-82. [PMID: 27062198 DOI: 10.1002/ardp.201500472] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/15/2016] [Accepted: 03/18/2016] [Indexed: 11/09/2022]
Abstract
Histone deacetylase (HDAC) proteins have emerged as targets for anti-cancer therapeutics, with several inhibitors used in the clinic, including suberoylanilide hydroxamic acid (SAHA, vorinostat). Because SAHA and many other inhibitors target all or most of the 11 human HDAC proteins, the creation of selective inhibitors has been studied intensely. Recently, inhibitors selective for HDAC1 and HDAC2 were reported where selectivity was attributed to interactions between substituents on the metal binding moiety of the inhibitor and residues in the 14-Å internal cavity of the HDAC enzyme structure. Based on this earlier work, we synthesized and tested SAHA analogs with substituents on the hydroxamic acid metal binding moiety. The N-substituted SAHA analogs displayed reduced potency and solubility, but greater selectivity, compared to SAHA. Docking studies suggested that the N-substituent accesses the 14-Å internal cavity to impart preferential inhibition of HDAC1. These studies with N-substituted SAHA analogs are consistent with the strategy exploiting the 14-Å internal cavity of HDAC proteins to create HDAC1/2 selective inhibitors.
Collapse
Affiliation(s)
| | | | | | - Mary Kay H Pflum
- Department of Chemistry, Wayne State University, Detroit, MI, USA
| |
Collapse
|
56
|
Target engagement and drug residence time can be observed in living cells with BRET. Nat Commun 2015; 6:10091. [PMID: 26631872 PMCID: PMC4686764 DOI: 10.1038/ncomms10091] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/30/2015] [Indexed: 12/18/2022] Open
Abstract
The therapeutic action of drugs is predicated on their physical engagement with cellular targets. Here we describe a broadly applicable method using bioluminescence resonance energy transfer (BRET) to reveal the binding characteristics of a drug with selected targets within intact cells. Cell-permeable fluorescent tracers are used in a competitive binding format to quantify drug engagement with the target proteins fused to Nanoluc luciferase. The approach enabled us to profile isozyme-specific engagement and binding kinetics for a panel of histone deacetylase (HDAC) inhibitors. Our analysis was directed particularly to the clinically approved prodrug FK228 (Istodax/Romidepsin) because of its unique and largely unexplained mechanism of sustained intracellular action. Analysis of the binding kinetics by BRET revealed remarkably long intracellular residence times for FK228 at HDAC1, explaining the protracted intracellular behaviour of this prodrug. Our results demonstrate a novel application of BRET for assessing target engagement within the complex milieu of the intracellular environment.
Collapse
|
57
|
Mendoza-Sanchez R, Cotnoir-White D, Kulpa J, Jutras I, Pottel J, Moitessier N, Mader S, Gleason JL. Design, synthesis and evaluation of antiestrogen and histone deacetylase inhibitor molecular hybrids. Bioorg Med Chem 2015; 23:7597-606. [DOI: 10.1016/j.bmc.2015.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/28/2015] [Accepted: 11/05/2015] [Indexed: 01/28/2023]
|
58
|
Toro TB, Watt TJ. KDAC8 substrate specificity quantified by a biologically relevant, label-free deacetylation assay. Protein Sci 2015; 24:2020-32. [PMID: 26402585 DOI: 10.1002/pro.2813] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 08/21/2015] [Accepted: 09/20/2015] [Indexed: 01/25/2023]
Abstract
Analysis of the human proteome has identified thousands of unique protein sequences that contain acetylated lysine residues in vivo. These modifications regulate a variety of biological processes and are reversed by the lysine deacetylase (KDAC) family of enzymes. Despite the known prevalence and importance of acetylation, the details of KDAC substrate recognition are not well understood. While several methods have been developed to monitor protein deacetylation, none are particularly suited for identifying enzyme-substrate pairs of label-free substrates across the entire family of lysine deacetylases. Here, we present a fluorescamine-based assay which is more biologically relevant than existing methods and amenable to probing substrate specificity. Using this assay, we evaluated the activity of KDAC8 and other lysine deacetylases, including a sirtuin, for several peptides derived from known acetylated proteins. KDAC8 showed clear preferences for some peptides over others, indicating that the residues immediately surrounding the acetylated lysine play an important role in substrate specificity. Steady-state kinetics suggest that the sequence surrounding the acetylated lysine affects binding affinity and catalytic rate independently. Our results provide direct evidence that potential KDAC8 substrates previously identified through cell based experiments can be directly deacetylated by KDAC8. Conversely, the data from this assay did not correlate well with predictions from previous screens for KDAC8 substrates using less biologically relevant substrates and assay conditions. Combining results from our assay with mass spectrometry-based experiments and cell-based experiments will allow the identification of specific KDAC-substrate pairs and lead to a better understanding of the biological consequences of these interactions.
Collapse
Affiliation(s)
- Tasha B Toro
- Department of Chemistry, Xavier University of Louisiana, New Orleans, Louisiana, 70125
| | - Terry J Watt
- Department of Chemistry, Xavier University of Louisiana, New Orleans, Louisiana, 70125
| |
Collapse
|
59
|
Noor Z, Afzal N, Rashid S. Exploration of Novel Inhibitors for Class I Histone Deacetylase Isoforms by QSAR Modeling and Molecular Dynamics Simulation Assays. PLoS One 2015; 10:e0139588. [PMID: 26431201 PMCID: PMC4592208 DOI: 10.1371/journal.pone.0139588] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/15/2015] [Indexed: 12/20/2022] Open
Abstract
Histone deacetylases (HDAC) are metal-dependent enzymes and considered as important targets for cell functioning. Particularly, higher expression of class I HDACs is common in the onset of multiple malignancies which results in deregulation of many target genes involved in cell growth, differentiation and survival. Although substantial attempts have been made to control the irregular functioning of HDACs by employing various inhibitors with high sensitivity towards transformed cells, limited success has been achieved in epigenetic cancer therapy. Here in this study, we used ligand-based pharmacophore and 2-dimensional quantitative structure activity relationship (QSAR) modeling approaches for targeting class I HDAC isoforms. Pharmacophore models were generated by taking into account the known IC50 values and experimental energy scores with extensive validations. The QSAR model having an external R2 value of 0.93 was employed for virtual screening of compound libraries. 10 potential lead compounds (C1-C10) were short-listed having strong binding affinities for HDACs, out of which 2 compounds (C8 and C9) were able to interact with all members of class I HDACs. The potential binding modes of HDAC2 and HDAC8 to C8 were explored through molecular dynamics simulations. Overall, bioactivity and ligand efficiency (binding energy/non-hydrogen atoms) profiles suggested that proposed hits may be more effective inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Zainab Noor
- National Center for Bioinformatics, Quaid I Azam University, Islamabad, Pakistan
| | - Noreen Afzal
- National Center for Bioinformatics, Quaid I Azam University, Islamabad, Pakistan
| | - Sajid Rashid
- National Center for Bioinformatics, Quaid I Azam University, Islamabad, Pakistan
| |
Collapse
|
60
|
Clausen DJ, Smith WB, Haines BE, Wiest O, Bradner JE, Williams RM. Modular synthesis and biological activity of pyridyl-based analogs of the potent Class I Histone Deacetylase Inhibitor Largazole. Bioorg Med Chem 2015; 23:5061-5074. [PMID: 26054247 PMCID: PMC4626217 DOI: 10.1016/j.bmc.2015.03.063] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 11/26/2022]
Abstract
The formation of a series of analogs containing a pyridine moiety in place of the natural thiazole heterocycle, based on the potent, naturally occurring HDAC inhibitor Largazole has been accomplished. The synthetic strategy was designed modularly to access multiple inhibitors with different aryl functionalities containing both the natural depsipeptide and peptide isostere variant of the macrocycle. The cytotoxicity and biochemical activity of the library of HDAC inhibitors is described herein.
Collapse
Affiliation(s)
- Dane J Clausen
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | - William B Smith
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Brandon E Haines
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Olaf Wiest
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Robert M Williams
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; University of Colorado Cancer Center, Aurora, CO 80045, USA.
| |
Collapse
|
61
|
Gupta SP. QSAR Studies on Hydroxamic Acids: A Fascinating Family of Chemicals with a Wide Spectrum of Activities. Chem Rev 2015; 115:6427-90. [DOI: 10.1021/cr500483r] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Satya P. Gupta
- Department of Applied Sciences, National Institute of Technical Teachers’ Training and Research, Shamla
Hills, Bhopal-462002, India
| |
Collapse
|
62
|
Hou X, Du J, Liu R, Zhou Y, Li M, Xu W, Fang H. Enhancing the Sensitivity of Pharmacophore-Based Virtual Screening by Incorporating Customized ZBG Features: A Case Study Using Histone Deacetylase 8. J Chem Inf Model 2015; 55:861-71. [DOI: 10.1021/ci500762z] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Xuben Hou
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology of Natural
Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong 250012, China
| | - Jintong Du
- Shandong Cancer Hospital and Institute, Jinan, Shandong 250117, China
| | - Renshuai Liu
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology of Natural
Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong 250012, China
| | - Yi Zhou
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology of Natural
Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong 250012, China
| | - Minyong Li
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology of Natural
Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong 250012, China
| | - Wenfang Xu
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology of Natural
Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong 250012, China
| | - Hao Fang
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology of Natural
Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
63
|
Deschamps N, Simões-Pires CA, Carrupt PA, Nurisso A. How the flexibility of human histone deacetylases influences ligand binding: an overview. Drug Discov Today 2015; 20:736-42. [PMID: 25597521 DOI: 10.1016/j.drudis.2015.01.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 12/12/2014] [Accepted: 01/09/2015] [Indexed: 12/17/2022]
Abstract
Over the past decade, human histone deacetylases (HDACs) have become interesting as therapeutic targets because of the benefits that their modulation might provide in aging-related disorders. Recently, studies using crystallography and computational chemistry have provided information on the structure and conformational changes related to HDAC-mediated recognition events. Through the description of the key mass and one-off movements observed in metal-dependent HDACs, here we highlight the impact of flexibility on drug-binding affinity and specificity. The collected information will be useful for not only a better understanding of the biological functions of HDACs, but also the conception of new selective binders.
Collapse
Affiliation(s)
- Nathalie Deschamps
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Quai Ernest Ansermet, 30, CH-1211 Geneva 4, Switzerland
| | - Claudia Avello Simões-Pires
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Quai Ernest Ansermet, 30, CH-1211 Geneva 4, Switzerland
| | - Pierre-Alain Carrupt
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Quai Ernest Ansermet, 30, CH-1211 Geneva 4, Switzerland
| | - Alessandra Nurisso
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Quai Ernest Ansermet, 30, CH-1211 Geneva 4, Switzerland.
| |
Collapse
|
64
|
Nakada Y, Kageyama K, Sugiyama A, Desaki R, Takayasu S, Niioka K, Murasawa S, Ishigame N, Asari Y, Iwasaki Y, Daimon M. Inhibitory effects of trichostatin A on adrenocorticotropic hormone production and proliferation of corticotroph tumor AtT-20 cells. Endocr J 2015; 62:1083-90. [PMID: 26497760 DOI: 10.1507/endocrj.ej15-0369] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Cushing's disease is primarily caused by adrenocorticotropic hormone (ACTH)-producing pituitary adenomas. Pituitary tumor-transforming gene 1 (PTTG1) expression, a hallmark of pituitary tumors, stimulates pituitary cell proliferation. Histone deacetylases (HDACs) play an important role in regulating gene transcription and HDAC inhibitors induce cellular differentiation and suppress tumor cell proliferation. HDAC inhibitors also repress PTTG1 mRNA levels. Trichostatin A (TSA) is a potent cell-permeable HDAC inhibitor that blocks cell cycle progression. In the present study, we determined the effect of TSA on ACTH production and cellular proliferation in mouse AtT-20 corticotroph tumor cells. TSA decreased proopiomelanocortin (POMC) mRNA levels in AtT-20 cells and reduced ACTH levels in the culture medium of these cells. The TSA-induced decreases in POMC mRNA levels were not modulated when TSA and dexamethasone were simultaneously administered. Drug treatment also decreased AtT-20 cell proliferation, induced apoptosis, and increased the percentage of cells in G0/G1 phase using flow cytometry. TSA decreased PTTG1 mRNA levels. Furthermore, PTTG1 knockdown inhibited cellular proliferation. Its knockdown also inhibited POMC mRNA and ACTH levels. TSA inhibits ACTH production and corticotroph tumor cell proliferation. TSA may inhibit cellular proliferation, and ACTH synthesis and secretion by decreasing PTTG1 expression.
Collapse
Affiliation(s)
- Yuki Nakada
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, Aomori 036-8562, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Kuzmochka C, Abdou HS, Haché RJG, Atlas E. Inactivation of histone deacetylase 1 (HDAC1) but not HDAC2 is required for the glucocorticoid-dependent CCAAT/enhancer-binding protein α (C/EBPα) expression and preadipocyte differentiation. Endocrinology 2014; 155:4762-73. [PMID: 25203139 DOI: 10.1210/en.2014-1565] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Several drugs currently used in the management of mood disorders, epilepsy (ie, valproic acid), or the control of inflammation (ie, corticosteroids) have been shown to promote visceral obesity in humans by increasing the number of newly formed adipocytes. Valproic acid is classified as a nonspecific histone deacetylase (HDAC) inhibitor, along with trichostatin A and butyric acid. In vitro experiments have demonstrated that such molecules greatly enhance the rate of preadipocyte differentiation, similarly to the effect of corticosteroids. The glucocorticoid receptor stimulates adipogenesis in part by enhancing the transcription of C/ebpa through the titration, and subsequent degradation, of HDAC1 from the C/ebpα promoter. There is, however, controversy in the literature as to the role of HDACs during adipogenesis. In this study, we sought to demonstrate, using 2 different strategies, the definite role of HDAC1 in adipogenesis. By using small interference RNA-mediated knockdown of HDAC1 and by generating an enzymatically inactive HDAC1D181A by site-directed mutagenesis, we were able to show that HDAC1, but not HDAC2, suppresses glucocorticoid receptor-potentiated preadipocyte differentiation by decreasing CCAAT/enhancer-binding protein (C/ebp)α and Pparγ expression levels at the onset of differentiation. Finally, we demonstrate that HDAC1D181A acts as a dominant negative mutant of HDAC1 during adipogenesis by modulating C/EBPβ transcriptional activity on the C/ebpα promoter.
Collapse
Affiliation(s)
- Claire Kuzmochka
- Northern Ontario School of Medicine, Ontario, Canada ON POM Reproduction, Mother and Youth Health (H.-S.A.), CHUQ Research Centre, Quebec city, Quebec, Canada G1R2J6; Environmental Health Science and Research Bureau (E.A.), Health Canada, Ottawa, Ontario, Canada M3J1P3; and York University (R.J.G.H.), Toronto, Ontario, Canada K1A0K9
| | | | | | | |
Collapse
|
66
|
|
67
|
Zang LL, Wang XJ, Li XB, Wang SQ, Xu WR, Xie XB, Cheng XC, Ma H, Wang RL. SAHA-based novel HDAC inhibitor design by core hopping method. J Mol Graph Model 2014; 54:10-8. [DOI: 10.1016/j.jmgm.2014.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 08/08/2014] [Accepted: 08/25/2014] [Indexed: 10/24/2022]
|
68
|
Kim B, Park H, Salvador LA, Serrano PE, Kwan JC, Zeller SL, Chen QY, Ryu S, Liu Y, Byeon S, Luesch H, Hong J. Evaluation of class I HDAC isoform selectivity of largazole analogues. Bioorg Med Chem Lett 2014; 24:3728-31. [PMID: 25070421 DOI: 10.1016/j.bmcl.2014.07.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 06/27/2014] [Accepted: 07/01/2014] [Indexed: 10/25/2022]
Abstract
Largazole is a potent class I selective histone deacetylase (HDAC) inhibitor. The majority of largazole analogues to date have modified the thiazole-thiazoline and the warhead moiety. In order to elucidate class I-specific structure-activity relationships, a series of analogues with modifications in the valine or the linker region were prepared and evaluated for their class I isoform selectivity. The inhibition profile showed that the C2 position of largazole has an optimal steric requirement for efficient HDAC inhibition and that substitution of the trans-alkene in the linker with an aromatic group results in complete loss of activity. This data will aid the design of class I isoform selective HDAC inhibitors.
Collapse
Affiliation(s)
- Bumki Kim
- Department of Chemistry, Duke University, Durham, NC 27708, United States
| | - Heekwang Park
- Department of Chemistry, Duke University, Durham, NC 27708, United States
| | - Lilibeth A Salvador
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States; Marine Science Institute, College of Science, University of the Philippines, Diliman, Quezon City 1100, Philippines
| | - Patrick E Serrano
- Department of Chemistry, Duke University, Durham, NC 27708, United States
| | - Jason C Kwan
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States
| | - Sabrina L Zeller
- Department of Chemistry, Duke University, Durham, NC 27708, United States
| | - Qi-Yin Chen
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States; Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL 32610, United States
| | - Soyoung Ryu
- Department of Chemistry, Duke University, Durham, NC 27708, United States
| | - Yanxia Liu
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States; Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL 32610, United States
| | - Seongrim Byeon
- Department of Chemistry, Duke University, Durham, NC 27708, United States
| | - Hendrik Luesch
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States; Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL 32610, United States.
| | - Jiyong Hong
- Department of Chemistry, Duke University, Durham, NC 27708, United States; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, United States.
| |
Collapse
|
69
|
A short antisense oligonucleotide ameliorates symptoms of severe mouse models of spinal muscular atrophy. MOLECULAR THERAPY. NUCLEIC ACIDS 2014; 3:e174. [PMID: 25004100 PMCID: PMC4121513 DOI: 10.1038/mtna.2014.23] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 05/23/2014] [Indexed: 12/22/2022]
Abstract
Recent reports underscore the unparalleled potential of antisense-oligonucleotide (ASO)-based approaches to ameliorate various pathological conditions. However, in vivo studies validating the effectiveness of a short ASO (<10-mer) in the context of a human disease have not been performed. One disease with proven amenability to ASO-based therapy is spinal muscular atrophy (SMA). SMA is a neuromuscular disease caused by loss-of-function mutations in the survival motor neuron 1 (SMN1) gene. Correction of aberrant splicing of the remaining paralog, SMN2, can rescue mouse models of SMA. Here, we report the therapeutic efficacy of an 8-mer ASO (3UP8i) in two severe models of SMA. While 3UP8i modestly improved survival and function in the more severe Taiwanese SMA model, it dramatically increased survival, improved neuromuscular junction pathology, and tempered cardiac deficits in a new, less severe model of SMA. Our results expand the repertoire of ASO-based compounds for SMA therapy, and for the first time, demonstrate the in vivo efficacy of a short ASO in the context of a human disease.
Collapse
|
70
|
Zhao WW, Liu FF, Shi QH, Dong XY, Sun Y. Biomimetic design of affinity peptide ligands for human IgG based on protein A-IgG complex. Biochem Eng J 2014. [DOI: 10.1016/j.bej.2014.03.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
71
|
Exploring the potential binding sites of some known HDAC inhibitors on some HDAC8 conformers by docking studies. Appl Biochem Biotechnol 2014; 173:1907-26. [PMID: 24888409 DOI: 10.1007/s12010-014-0976-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 05/19/2014] [Indexed: 01/03/2023]
Abstract
We describe the conformational behavior of histone deacetylase 8 (HDAC8) using molecular dynamics (MD) simulations. HDAC8 conformers were used for the docking studies using some known HDAC inhibitors (HDACi) suberoylanilide hydroxamic acid (SAHA), valproic acid (VPA), aroyl-pyrrole-hydroxy-amide (APHA-8) and tubacin to explore their interactions, binding modes, free energy values. The MD simulation show that HDAC8 make important surface changes at the catalytic site (CS) entrance as well as at two entrances locations in the 14-Å tunnel. In addition, we identify an alternate entrance to the 14-Å tunnel named adjacent to the catalytic site pocket (ACSP). By using docking studies, it was possible to elucidate the importance of hydrophobic and π-π interactions that are the most important for the ligand-HDAC8 complex structural stabilization. In conclusion, the ligand flexibility, molecular weight and chemical moieties (hydroxamic acid, aryl and aliphatic moieties) are the principal properties required to increase the binding affinity on HDAC8.
Collapse
|
72
|
Vellore NA, Baron R. Epigenetic molecular recognition: a biomolecular modeling perspective. ChemMedChem 2014; 9:484-94. [PMID: 24616246 DOI: 10.1002/cmdc.201300510] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Indexed: 01/23/2023]
Abstract
The abnormal regulation of epigenetic protein families is associated with the onset and progression of various human diseases. However, epigenetic processes remain relatively obscure at the molecular level, thus preventing the rational design of chemical therapeutics. An array of robust computational and modeling approaches can complement experiments to shed light on the complex mechanisms of epigenetic molecular recognition and can guide medicinal chemists in designing selective and potent drug molecules. Herein we present a review of studies focused on epigenetic molecular recognition from a biomolecular modeling viewpoint. Although the known epigenetic targets are numerous, this review focuses on the more limited protein families on which computational modeling has been successfully applied. Therefore, we review three main topics: 1) histone deacetylases, 2) histone demethylases, and 3) histone tail dynamics. A brief review of the biological background and biomedical relevance is presented for each topic, followed by a detailed discussion of the computational studies and their relevance.
Collapse
Affiliation(s)
- Nadeem A Vellore
- Department of Medicinal Chemistry, College of Pharmacy and The Henry Eyring Center for Theoretical Chemistry, The University of Utah, 30 South 2000 East, Salt Lake City, UT 84112 (USA)
| | | |
Collapse
|
73
|
Ganai SA, Shanmugam K, Mahadevan V. Energy-optimised pharmacophore approach to identify potential hotspots during inhibition of Class II HDAC isoforms. J Biomol Struct Dyn 2014; 33:374-87. [PMID: 24460542 DOI: 10.1080/07391102.2013.879073] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Histone deacetylases (HDACs) are conjugated enzymes that modulate chromatin architecture by deacetylating lysine residues on the histone tails leading to transcriptional repression. Pharmacological interventions of these enzymes with small molecule inhibitors called Histone deacetylase inhibitors (HDACi) have shown enhanced acetylation of the genome and are hence emerging as potential targets at the clinic. Type-specific inhibition of Class II HDACs has shown enhanced therapeutic benefits against developmental and neurodegenerative disorders. However, the structural identity of class-specific isoforms limits the potential of their inhibitors in precise targeting of their enzymes. Diverse strategies have been implemented to recognise the features in HDAC enzymes which may help in identifying isoform specificity factors. This work attempts a computational approach that combines in silico docking and energy-optimised pharmacophore (E-pharmacophore) mapping of 18 known HDAC inhibitors and has identified structural variations that regulate their interactions against the six Class II HDAC enzymes considered for the study. This combined approach establishes that inhibitors possessing higher number of aromatic rings in different structural regions might function as potent inhibitors, while inhibitors with scarce ring structures might point to compromised potency. This would aid the rationale for chemical optimisation and design of isoform selective HDAC inhibitors with enhanced affinity and therapeutic efficiency.
Collapse
Affiliation(s)
- Shabir Ahmad Ganai
- a Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical & Biotechnology , SASTRA University , Thanjavur 613401 , India
| | | | | |
Collapse
|
74
|
Wambua MK, Nalawansha DA, Negmeldin AT, Pflum MKH. Mutagenesis studies of the 14 Å internal cavity of histone deacetylase 1: insights toward the acetate-escape hypothesis and selective inhibitor design. J Med Chem 2014; 57:642-50. [PMID: 24405391 PMCID: PMC3983352 DOI: 10.1021/jm401837e] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
![]()
Histone
deacetylase (HDAC) proteins are promising targets for cancer
treatment, as shown by the approval of two HDAC inhibitors for the
treatment of cutaneous T-cell lymphoma. HDAC1 in particular has been
linked to cell growth and cell cycle regulation and is therefore an
attractive target for anticancer drugs. The HDAC1 active site contains
a hydrophobic 11 Å active-site channel, with a 14 Å internal
cavity at the bottom of the active site. Several computational and
biochemical studies have proposed an acetate-escape hypothesis where
the acetate byproduct of the deacetylation reaction escapes via the
14 Å internal cavity. Selective HDAC inhibitors that bind to
the 14 Å cavity have also been created. To understand the influence
of amino acids lining the HDAC1 14 Å cavity in acetate escape
and inhibitor binding, we used mutagenesis coupled with acetate competition
assays. The results indicate that amino acids lining the 14 Å
cavity are critical for catalytic activity and acetate competition,
confirming the role of the cavity in acetate escape. In addition,
these mutagenesis studies will aid in HDAC1-inhibitor design that
exploits the 14 Å cavity.
Collapse
Affiliation(s)
- Magdalene K Wambua
- Department of Chemistry, Wayne State University , 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | | | | | | |
Collapse
|
75
|
Kaiser FJ, Ansari M, Braunholz D, Concepción Gil-Rodríguez M, Decroos C, Wilde JJ, Fincher CT, Kaur M, Bando M, Amor DJ, Atwal PS, Bahlo M, Bowman CM, Bradley JJ, Brunner HG, Clark D, Del Campo M, Di Donato N, Diakumis P, Dubbs H, Dyment DA, Eckhold J, Ernst S, Ferreira JC, Francey LJ, Gehlken U, Guillén-Navarro E, Gyftodimou Y, Hall BD, Hennekam R, Hudgins L, Hullings M, Hunter JM, Yntema H, Innes AM, Kline AD, Krumina Z, Lee H, Leppig K, Lynch SA, Mallozzi MB, Mannini L, McKee S, Mehta SG, Micule I, Mohammed S, Moran E, Mortier GR, Moser JAS, Noon SE, Nozaki N, Nunes L, Pappas JG, Penney LS, Pérez-Aytés A, Petersen MB, Puisac B, Revencu N, Roeder E, Saitta S, Scheuerle AE, Schindeler KL, Siu VM, Stark Z, Strom SP, Thiese H, Vater I, Willems P, Williamson K, Wilson LC, Hakonarson H, Quintero-Rivera F, Wierzba J, Musio A, Gillessen-Kaesbach G, Ramos FJ, Jackson LG, Shirahige K, Pié J, Christianson DW, Krantz ID, Fitzpatrick DR, Deardorff MA. Loss-of-function HDAC8 mutations cause a phenotypic spectrum of Cornelia de Lange syndrome-like features, ocular hypertelorism, large fontanelle and X-linked inheritance. Hum Mol Genet 2014; 23:2888-900. [PMID: 24403048 DOI: 10.1093/hmg/ddu002] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Cornelia de Lange syndrome (CdLS) is a multisystem genetic disorder with distinct facies, growth failure, intellectual disability, distal limb anomalies, gastrointestinal and neurological disease. Mutations in NIPBL, encoding a cohesin regulatory protein, account for >80% of cases with typical facies. Mutations in the core cohesin complex proteins, encoded by the SMC1A, SMC3 and RAD21 genes, together account for ∼5% of subjects, often with atypical CdLS features. Recently, we identified mutations in the X-linked gene HDAC8 as the cause of a small number of CdLS cases. Here, we report a cohort of 38 individuals with an emerging spectrum of features caused by HDAC8 mutations. For several individuals, the diagnosis of CdLS was not considered prior to genomic testing. Most mutations identified are missense and de novo. Many cases are heterozygous females, each with marked skewing of X-inactivation in peripheral blood DNA. We also identified eight hemizygous males who are more severely affected. The craniofacial appearance caused by HDAC8 mutations overlaps that of typical CdLS but often displays delayed anterior fontanelle closure, ocular hypertelorism, hooding of the eyelids, a broader nose and dental anomalies, which may be useful discriminating features. HDAC8 encodes the lysine deacetylase for the cohesin subunit SMC3 and analysis of the functional consequences of the missense mutations indicates that all cause a loss of enzymatic function. These data demonstrate that loss-of-function mutations in HDAC8 cause a range of overlapping human developmental phenotypes, including a phenotypically distinct subgroup of CdLS.
Collapse
Affiliation(s)
- Frank J Kaiser
- Sektion für Funktionelle Genetik am Institut für Humangenetik, Universität zu Lübeck, Lübeck 23538, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Wang J, Sun F, Han L, Hou X, Pan X, Liu R, Tang W, Fang H. Design, synthesis, and preliminary bioactivity studies of substituted purine hydroxamic acid derivatives as novel histone deacetylase (HDAC) inhibitors. MEDCHEMCOMM 2014. [DOI: 10.1039/c4md00203b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Histone deacetylase (HDAC) is a clinically validated target for anti-tumor therapy. In order to increase HDAC inhibition and efficiency, we developed a series of novel substituted purine hydroxamic acids as potent HDAC inhibitors.
Collapse
Affiliation(s)
- Junhua Wang
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (Ministry of Education)
- School of Pharmacy
- Shandong University
- Jinan 250012
| | - Feng'e Sun
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (Ministry of Education)
- School of Pharmacy
- Shandong University
- Jinan 250012
| | - Leiqiang Han
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (Ministry of Education)
- School of Pharmacy
- Shandong University
- Jinan 250012
| | - Xuben Hou
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (Ministry of Education)
- School of Pharmacy
- Shandong University
- Jinan 250012
| | - Xiaole Pan
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (Ministry of Education)
- School of Pharmacy
- Shandong University
- Jinan 250012
| | - Renshuai Liu
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (Ministry of Education)
- School of Pharmacy
- Shandong University
- Jinan 250012
| | - Weiping Tang
- Division of Pharmaceutical Sciences
- School of Pharmacy
- University of Wisconsin
- Madison 53705
- USA
| | - Hao Fang
- Department of Medicinal Chemistry
- Key Laboratory of Chemical Biology (Ministry of Education)
- School of Pharmacy
- Shandong University
- Jinan 250012
| |
Collapse
|
77
|
Newman JC, Verdin E. Ketone bodies as signaling metabolites. Trends Endocrinol Metab 2014; 25:42-52. [PMID: 24140022 PMCID: PMC4176946 DOI: 10.1016/j.tem.2013.09.002] [Citation(s) in RCA: 652] [Impact Index Per Article: 65.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 09/14/2013] [Accepted: 09/17/2013] [Indexed: 12/13/2022]
Abstract
Traditionally, the ketone body β-hydroxybutyrate (βOHB) has been looked upon as a carrier of energy from liver to peripheral tissues during fasting or exercise. However, βOHB also signals via extracellular receptors and acts as an endogenous inhibitor of histone deacetylases (HDACs). These recent findings support a model in which βOHB functions to link the environment, in this case the diet, and gene expression via chromatin modifications. We review the regulation and functions of ketone bodies, the relationship between ketone bodies and calorie restriction, and the implications of HDAC inhibition by the ketone body βOHB in the modulation of metabolism and in diseases of aging.
Collapse
Affiliation(s)
- John C Newman
- Gladstone Institutes and University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158, USA
| | - Eric Verdin
- Gladstone Institutes and University of California San Francisco, 1650 Owens Street, San Francisco, CA 94158, USA.
| |
Collapse
|
78
|
Lactam based 7-amino suberoylamide hydroxamic acids as potent HDAC inhibitors. Bioorg Med Chem Lett 2014; 24:61-4. [DOI: 10.1016/j.bmcl.2013.11.072] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 11/25/2013] [Accepted: 11/29/2013] [Indexed: 10/25/2022]
|
79
|
Kunze MBA, Wright DW, Werbeck ND, Kirkpatrick J, Coveney PV, Hansen DF. Loop interactions and dynamics tune the enzymatic activity of the human histone deacetylase 8. J Am Chem Soc 2013; 135:17862-8. [PMID: 24171457 PMCID: PMC3926704 DOI: 10.1021/ja408184x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
![]()
The human histone deacetylase 8 (HDAC8)
is a key hydrolase in gene
regulation and has been identified as a drug target for the treatment
of several cancers. Previously the HDAC8 enzyme has been extensively
studied using biochemical techniques, X-ray crystallography, and computational
methods. Those investigations have yielded detailed information about
the active site and have demonstrated that the substrate entrance
surface is highly dynamic. Yet it has remained unclear how the dynamics
of the entrance surface tune and influence the catalytic activity
of HDAC8. Using long time scale all atom molecular dynamics simulations
we have found a mechanism whereby the interactions and dynamics of
two loops tune the configuration of functionally important residues
of HDAC8 and could therefore influence the activity of the enzyme.
We subsequently investigated this hypothesis using a well-established
fluorescence activity assay and a noninvasive real-time progression
assay, where deacetylation of a p53 based peptide was observed by
nuclear magnetic resonance spectroscopy. Our work delivers detailed
insight into the dynamic loop network of HDAC8 and provides an explanation
for a number of experimental observations.
Collapse
Affiliation(s)
- Micha B A Kunze
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London , Gower Street, London WC1E 6BT, United Kingdom
| | | | | | | | | | | |
Collapse
|
80
|
McCarthy MM, Nugent BM. Epigenetic contributions to hormonally-mediated sexual differentiation of the brain. J Neuroendocrinol 2013; 25:1133-40. [PMID: 23919286 PMCID: PMC5330673 DOI: 10.1111/jne.12072] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 06/18/2013] [Accepted: 06/29/2013] [Indexed: 12/28/2022]
Abstract
It has been long established that hormones exert enduring influences on the developing brain that direct the reproductive response in adulthood, although the cellular mechanisms by which organisational effects are maintained have not been determined satisfactorily. Recent interest in epigenetic modifications to the nervous system has highlighted the potential for hormone-induced changes to the genome that could endure for the lifespan but not be transmitted to the next generation. Preliminary evidence suggests that this is indeed possible because sex differences in the histone code and in the methylation of CpGs in the promoters of specific genes have been identified and, at times, functionally correlated with behaviour. The present review provides an overview of epigenetic processes and discusses the current state-of-the-art, and also identifies future directions.
Collapse
Affiliation(s)
- M M McCarthy
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | | |
Collapse
|
81
|
Papakyriakou A, Zervoudi E, Theodorakis EA, Saveanu L, Stratikos E, Vourloumis D. Novel selective inhibitors of aminopeptidases that generate antigenic peptides. Bioorg Med Chem Lett 2013; 23:4832-6. [PMID: 23916253 DOI: 10.1016/j.bmcl.2013.07.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/12/2013] [Accepted: 07/15/2013] [Indexed: 11/15/2022]
Abstract
Endoplasmic reticulum aminopeptidases, ERAP1 and ERAP2, as well as Insulin regulated aminopeptidase (IRAP) play key roles in antigen processing, and have recently emerged as biologically important targets for manipulation of antigen presentation. Taking advantage of the available structural and substrate-selectivity data for these enzymes, we have rationally designed a new series of inhibitors that display low micromolar activity. The selectivity profile for these three highly homologous aminopeptidases provides a promising avenue for modulating intracellular antigen processing.
Collapse
Affiliation(s)
- Athanasios Papakyriakou
- Department of Chemistry & Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0358, USA
| | | | | | | | | | | |
Collapse
|
82
|
Yuan J, Pu Y, Yin L. Docking-based three-dimensional quantitative structure-activity relationship (3D-QSAR) predicts binding affinities to aryl hydrocarbon receptor for polychlorinated dibenzodioxins, dibenzofurans, and biphenyls. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2013; 32:1453-1458. [PMID: 23424013 DOI: 10.1002/etc.2191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/14/2013] [Accepted: 02/06/2013] [Indexed: 06/01/2023]
Abstract
Polychlorinated dibenzodioxins (PCDDs), polychlorinated dibenzofurans (PCDFs), and polychlorinated biphenyls (PCBs) cause toxic effects after binding to an intracellular cytosolic receptor called the aryl hydrocarbon receptor (AhR). Thymic atrophy, weight loss, immunotoxicity, acute lethality, and induction of cytochrome P4501A1 have all been correlated with the binding affinity to AhR. To study the key molecular features for determining binding affinity to AhR, a homology model of AhR ligand-binding domains was developed, a molecular docking approach was employed to obtain docking-based conformations of all molecules in the whole set, and 3-dimensional quantitative structure-activity relationship (3D-QSAR) methodology, namely, comparative molecular field analysis (CoMFA), was applied. A partial least square analysis was performed, and QSAR models were generated for a training set of 59 compounds. The generated QSAR model showed good internal and external statistical reliability, and in a comparison with other reported CoMFA models using different alignment methods, the docking-based CoMFA model showed some advantages.
Collapse
Affiliation(s)
- Jintao Yuan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | | | | |
Collapse
|
83
|
Di Micco S, Chini MG, Terracciano S, Bruno I, Riccio R, Bifulco G. Structural basis for the design and synthesis of selective HDAC inhibitors. Bioorg Med Chem 2013; 21:3795-807. [DOI: 10.1016/j.bmc.2013.04.036] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 04/09/2013] [Accepted: 04/10/2013] [Indexed: 10/26/2022]
|
84
|
Kim J, Chun P, Moon HR. Synthesis of Novel N-(2-Hydroxyphenyl)arylsulfonamides as Selective HDAC Inhibitory and Cytotoxic Agents. B KOREAN CHEM SOC 2013. [DOI: 10.5012/bkcs.2013.34.5.1487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
85
|
Simões-Pires C, Zwick V, Nurisso A, Schenker E, Carrupt PA, Cuendet M. HDAC6 as a target for neurodegenerative diseases: what makes it different from the other HDACs? Mol Neurodegener 2013; 8:7. [PMID: 23356410 PMCID: PMC3615964 DOI: 10.1186/1750-1326-8-7] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 01/21/2013] [Indexed: 01/04/2023] Open
Abstract
Histone deacetylase (HDAC) inhibitors have been demonstrated to be beneficial in animal models of neurodegenerative diseases. Such results were mainly associated with the epigenetic modulation caused by HDACs, especially those from class I, via chromatin deacetylation. However, other mechanisms may contribute to the neuroprotective effect of HDAC inhibitors, since each HDAC may present distinct specific functions within the neurodegenerative cascades. Such an example is HDAC6 for which the role in neurodegeneration has been partially elucidated so far. The strategy to be adopted in promising therapeutics targeting HDAC6 is still controversial. Specific inhibitors exert neuroprotection by increasing the acetylation levels of α-tubulin with subsequent improvement of the axonal transport, which is usually impaired in neurodegenerative disorders. On the other hand, an induction of HDAC6 would theoretically contribute to the degradation of protein aggregates which characterize various neurodegenerative disorders, including Alzheimer’s, Parkinson’s and Hutington’s diseases. This review describes the specific role of HDAC6 compared to the other HDACs in the context of neurodegeneration, by collecting in silico, in vitro and in vivo results regarding the inhibition and/or knockdown of HDAC6 and other HDACs. Moreover, structure, function, subcellular localization, as well as the level of HDAC6 expression within brain regions are reviewed and compared to the other HDAC isoforms. In various neurodegenerative diseases, the mechanisms underlying HDAC6 interaction with other proteins seem to be a promising approach in understanding the modulation of HDAC6 activity.
Collapse
Affiliation(s)
- Claudia Simões-Pires
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Quai Ernest-Ansermet 30, CH-1211 Geneva 4, Switzerland
| | | | | | | | | | | |
Collapse
|
86
|
A novel two-step QSAR modeling work flow to predict selectivity and activity of HDAC inhibitors. Bioorg Med Chem Lett 2013; 23:929-33. [PMID: 23321563 DOI: 10.1016/j.bmcl.2012.12.067] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/17/2012] [Accepted: 12/20/2012] [Indexed: 11/22/2022]
Abstract
A two-step modeling approach was employed to study the selectivity and activity of histone deacetylase inhibitors. First, according to the activity difference against HDAC1 and HDAC6, a binary classification model was established to classify two kinds of inhibitors. Then two continuous models were built for each subclass to predict the activity value of HDAC1 and HDAC6 inhibitors. The three models were all built with the GA-kNN method combined with dragon descriptors. They were external validated by using external prediction set and Y-randomization test. The highly predictive models were generated for all three data sets. For the classification model, the classification accuracies of the models were as high as 100% for the external test set. For HDAC1 and HDAC6 inhibitor consecutive models, external R(2) values are 0.947 and 0.911, respectively. The results proved the reliability of these models. All models were used to screen 1000 compounds included in PubMed dataset. Virtual screening resulted in 8 and 13 structurally unique consensus hits that were considered novel putative HDAC1 and HDAC6 inhibitors, respectively.
Collapse
|
87
|
Salvador LA, Luesch H. Discovery and mechanism of natural products as modulators of histone acetylation. Curr Drug Targets 2012; 13:1029-47. [PMID: 22594471 DOI: 10.2174/138945012802008973] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 01/30/2012] [Accepted: 05/15/2012] [Indexed: 12/31/2022]
Abstract
Small molecules that modulate histone acetylation by targeting key enzymes mediating this posttranslational modification - histone acetyltransferases and histone deacetylases - are validated chemotherapeutic agents for the treatment of cancer. This area of research has seen a rapid increase in interest in the past decade, with the structurally diverse natural products-derived compounds at its forefront. These secondary metabolites from various biological sources target this epigenetic modification through distinct mechanisms of enzyme regulation by utilizing a diverse array of pharmacophores. We review the discovery of these compounds and discuss their modes of inhibition together with their downstream biological effects.
Collapse
Affiliation(s)
- Lilibeth A Salvador
- Department of Medicinal Chemistry, University of Florida, Gainesville, 32610, USA
| | | |
Collapse
|
88
|
Vyas VK, Ukawala RD, Ghate M, Chintha C. Homology modeling a fast tool for drug discovery: current perspectives. Indian J Pharm Sci 2012. [PMID: 23204616 PMCID: PMC3507339 DOI: 10.4103/0250-474x.102537] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Major goal of structural biology involve formation of protein-ligand complexes; in which the protein molecules act energetically in the course of binding. Therefore, perceptive of protein-ligand interaction will be very important for structure based drug design. Lack of knowledge of 3D structures has hindered efforts to understand the binding specificities of ligands with protein. With increasing in modeling software and the growing number of known protein structures, homology modeling is rapidly becoming the method of choice for obtaining 3D coordinates of proteins. Homology modeling is a representation of the similarity of environmental residues at topologically corresponding positions in the reference proteins. In the absence of experimental data, model building on the basis of a known 3D structure of a homologous protein is at present the only reliable method to obtain the structural information. Knowledge of the 3D structures of proteins provides invaluable insights into the molecular basis of their functions. The recent advances in homology modeling, particularly in detecting and aligning sequences with template structures, distant homologues, modeling of loops and side chains as well as detecting errors in a model contributed to consistent prediction of protein structure, which was not possible even several years ago. This review focused on the features and a role of homology modeling in predicting protein structure and described current developments in this field with victorious applications at the different stages of the drug design and discovery.
Collapse
Affiliation(s)
- V K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad-382 481, India
| | | | | | | |
Collapse
|
89
|
Thangapandian S, John S, Lee Y, Arulalapperumal V, Lee KW. Molecular modeling study on tunnel behavior in different histone deacetylase isoforms. PLoS One 2012; 7:e49327. [PMID: 23209570 PMCID: PMC3510210 DOI: 10.1371/journal.pone.0049327] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 10/07/2012] [Indexed: 12/11/2022] Open
Abstract
Histone deacetylases (HDACs) have emerged as effective therapeutic targets in the treatment of various diseases including cancers as these enzymes directly involved in the epigenetic regulation of genes. However the development of isoform-selective HDAC inhibitors has been a challenge till date since all HDAC enzymes possess conserved tunnel-like active site. In this study, using molecular dynamics simulation we have analyzed the behavior of tunnels present in HDAC8, 10, and 11 enzymes of class I, II, and IV, respectively. We have identified the equivalent tunnel forming amino acids in these three isoforms and found that they are very much conserved with subtle differences to be utilized in selective inhibitor development. One amino acid, methionine of HDAC8, among six tunnel forming residues is different in isoforms of other classes (glutamic acid (E) in HDAC10 and leucine (L) in HDAC 11) based on which mutations were introduced in HDAC11, the less studied HDAC isoform, to observe the effects of this change. The HDAC8-like (L268M) mutation in the tunnel forming residues has almost maintained the deep and narrow tunnel as present in HDAC8 whereas HDAC10-like (L268E) mutation has changed the tunnel wider and shallow as observed in HDAC10. These results explained the importance of the single change in the tunnel formation in different isoforms. The observations from this study can be utilized in the development of isoform-selective HDAC inhibitors.
Collapse
Affiliation(s)
| | | | | | | | - Keun Woo Lee
- Division of Applied Life Science (BK21 Program), Systems and Synthetic Agrobiotech Center (SSAC), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), Gazwa-dong, Jinju, Republic of Korea.
- * E-mail:
| |
Collapse
|
90
|
Targeted cancer therapy: giving histone deacetylase inhibitors all they need to succeed. Future Med Chem 2012; 4:505-24. [PMID: 22416777 DOI: 10.4155/fmc.12.3] [Citation(s) in RCA: 296] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Histone deacetylase inhibitors (HDACis) have now emerged as a powerful new class of small-molecule therapeutics acting through the regulation of the acetylation states of histone proteins (a form of epigenetic modulation) and other non-histone protein targets. Over 490 clinical trials have been initiated in the last 10 years, culminating in the approval of two structurally distinct HDACis - SAHA (vorinostat, Zolinza™) and FK228 (romidepsin, Istodax™). However, the current HDACis have serious limitations, including ineffectively low concentrations in solid tumors and cardiac toxicity, which is hindering their progress in the clinic. Herein, we review the primary paradigms being pursued to overcome these hindrances, including HDAC isoform selectivity, localized administration, and targeting cap groups to achieve selective tissue and cell type distribution.
Collapse
|
91
|
Design, synthesis and preliminary bioactivity studies of 1,3,4-thiadiazole hydroxamic acid derivatives as novel histone deacetylase inhibitors. Bioorg Med Chem 2012; 20:3865-72. [DOI: 10.1016/j.bmc.2012.04.032] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 04/13/2012] [Accepted: 04/16/2012] [Indexed: 11/23/2022]
|
92
|
Stolfa DA, Stefanachi A, Gajer JM, Nebbioso A, Altucci L, Cellamare S, Jung M, Carotti A. Design, Synthesis, and Biological Evaluation of 2-Aminobenzanilide Derivatives as Potent and Selective HDAC Inhibitors. ChemMedChem 2012; 7:1256-66. [DOI: 10.1002/cmdc.201200193] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Indexed: 11/11/2022]
|
93
|
Saravanan P, Dubey VK, Patra S. Potential selective inhibitors against Rv0183 of Mycobacterium tuberculosis targeting host lipid metabolism. Chem Biol Drug Des 2012; 79:1056-62. [PMID: 22405030 DOI: 10.1111/j.1747-0285.2012.01373.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Tuberculosis is the second leading infectious killer with 9 million new cases in 2009. Extensive use of pathogen's lipid metabolism especially in utilizing the host lipids and virulence highlights the importance of exported lipid-catabolizing enzymes. Current study aims to emphasize the importance of Rv0183, an exported monoacylglycerol lipase, involved in metabolizing the host cell membrane lipids. Sequence analysis and homology modeling shows Rv0183 is highly conserved throughout mycobacterial species even in Mycobacterium leprae and also significantly divergent from mammalian lipases. Additionally, employing virtual screening using NCI diversity set and ZINC database with criteria of molecules with higher predicted free energy of binding toward Rv0183 than human lipase, potential inhibitors have been identified for Rv0183. A tautomer of ZINC13451138, known inhibitor for HIV-1 integrase is the best hit with difference in free energy of binding of 8.72 kcal/mol. The sequence and structure analysis were helpful in identifying the ligand binding sites and molecular function of the mycobacterial specific monoacylglycerol lipase. Rv0183 represents a suitable and promising drug target and is also a step towards understanding dormancy development and reactivation, thereby addressing pathogen's drug resistance. Experimental studies on the discovered potential inhibitors in this virtual screen should further validate the therapeutic utility of Rv0183.
Collapse
|
94
|
Abstract
Suberoylanilide hydroxamic acid (vorinostat) was the first of the histone deacetylase inhibitors (HDACi) to be entered as therapy for the treatment of cutaneous T-cell lymphoma. Since then, a number of HDACi belonging to the short-chain fatty acid, hydroxamate, cyclic peptide or benzamide classes have been investigated in Phase II or III clinical trials (alone or in combination) for the treatment of many kinds of tumors. In addition, HDACi can be useful in antimalarial and antifungal therapies, and can reactivate HIV-1 expression in latent cellular reservoirs, thus suggesting that they could be used in combination with highly active antiretroviral therapy. Moreover, they have also proved their efficacy in neurodegenerative diseases, such as Huntington's disease, Parkinson's disease and Friedreich's ataxia. In particular, a new series of bis-anilides demonstrating a peculiar mechanism of action displayed highly beneficial effects against Huntington's disease and Friedreich's ataxia. In addition, a number of sirtuin inhibitors demonstrated antiproliferative effects in cell assays as well as in mouse tumor models, thus suggesting a role of such compounds in therapy against cancer. Furthermore, the SIRT2-selective AGK-2 has been reported to have protective effects against Parkinson's disease, and resveratrol and other sirtuin activators can be useful for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Antonello Mai
- Pasteur Institute-Cenci Bolognetti Foundation, Drug Chemistry and Technologies Department, University of Rome Sapienza, Piazzale Aldo Moro 5, Rome, Italy.
| |
Collapse
|
95
|
Cai X, Qian C. Discovery of HDAC-Inhibiting Multi-Target Inhibitors. DESIGNING MULTI-TARGET DRUGS 2012. [DOI: 10.1039/9781849734912-00221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Several HDAC-inhibiting multiple-target inhibitors have been reported. In this chapter, the current progress in investigating HDAC-inhibiting multi-target inhibitors is briefly reviewed, with a focus on the first and only clinical candidate CUDC-101 as the case study example. CUDC-101 is a novel small molecule potently inhibiting activities of the EGFR and HER2 kinases and HDAC enzymes with IC50 values of 2.4, 15.7, and 4.4nM, respectively. CUDC-101's rational design and synthesis, superior in vitro potency, broad anti-proliferative and pro-apoptotic activities in cultured tumor cells including RTK inhibitor-resistant cell lines, effective network disruption in survival signaling pathways, high efficacy in in vivo xenograft animal models, favorable safety profile, and preliminary evidence of anti-tumor activity in phase I trials are presented in this chapter. This case study provides proof-of-principle that a single molecule with multiple targeted specificities can improve the effectiveness of current anticancer therapeutics preclinically.
Collapse
|
96
|
Insights from comprehensive multiple receptor docking to HDAC8. J Mol Model 2012; 18:3927-39. [PMID: 22431224 DOI: 10.1007/s00894-011-1297-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 11/02/2011] [Indexed: 12/29/2022]
Abstract
A systematic investigation of the available crystal structures of HDAC8 and of the influence of different receptor structures and docking protocols is presented. The study shows that the open conformation of HDAC8 may be preferred by ligands with flexible surface binding groups, as such a conformation allows the ligands to minimize their exposure to solvent upon binding. This observation allowed us to rationalize the excellent potency of pyrazole-based inhibitors compared to that of isoxazole-based inhibitors.
Collapse
|
97
|
Thaler F, Varasi M, Carenzi G, Colombo A, Abate A, Bigogno C, Boggio R, Carrara S, Cataudella T, Dal Zuffo R, Reali V, Vultaggio S, Dondio G, Gagliardi S, Minucci S, Mercurio C. Spiro[chromane-2,4′-piperidine]-Based Histone Deacetylase Inhibitors with Improved in vivo Activity. ChemMedChem 2012; 7:709-21. [DOI: 10.1002/cmdc.201200024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Indexed: 11/06/2022]
|
98
|
Thangapandian S, John S, Lee KW. Molecular Dynamics Simulation Study Explaining Inhibitor Selectivity in Different Class of Histone Deacetylases. J Biomol Struct Dyn 2012; 29:677-98. [DOI: 10.1080/07391102.2012.10507409] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
99
|
Huang D, Li X, Sun L, Xiu Z, Nishino N. Synthesis, evaluation and molecular modeling of cyclic tetrapeptide histone deacetylase inhibitors as anticancer agents. J Pept Sci 2012; 18:242-51. [PMID: 22253009 DOI: 10.1002/psc.2392] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 10/24/2011] [Accepted: 12/01/2011] [Indexed: 11/07/2022]
Abstract
Histone deacetylase inhibitors (HDACIs) are a promising class of anticancer agents. To examine whether a slight change in the recognition domain could alter their inhibitory activity, we synthesized a series of cyclo(-L-Am7(S2Py)-Aib-L-Phe(n-Me)-D-Pro)derivatives and evaluated their HDAC inhibitory and anticancer activities. The peptides exhibited potent HDAC inhibitory activity and inhibited three human cancer cell lines with IC₅₀ in the micromolar range. Docking and molecular dynamics simulation were conducted to explore the interaction mechanisms of class I and II HDACs with these inhibitors. It revealed that the zinc ion in the active site coordinated five atoms of HDACs and the sulfur atom of the inhibitor. The metal binding domains of these compounds interacted with HDAC2, and the surface recognition domains of these compounds interacted with HDAC4 through hydrogen bonding. The hydrophobic interactions also provided favorable contributions to stabilize the complexes. The results obtained from this study would be helpful for us to design some novel cyclic tetrapeptides that may act as potent HDACIs.
Collapse
Affiliation(s)
- Dawei Huang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, China
| | | | | | | | | |
Collapse
|
100
|
Design, synthesis and biological evaluation of novel histone deacetylase inhibitors based on virtual screening. Acta Pharm Sin B 2011. [DOI: 10.1016/j.apsb.2011.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|