1
|
Scarano N, Brullo C, Musumeci F, Millo E, Bruzzone S, Schenone S, Cichero E. Recent Advances in the Discovery of SIRT1/2 Inhibitors via Computational Methods: A Perspective. Pharmaceuticals (Basel) 2024; 17:601. [PMID: 38794171 PMCID: PMC11123952 DOI: 10.3390/ph17050601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/26/2024] Open
Abstract
Sirtuins (SIRTs) are classified as class III histone deacetylases (HDACs), a family of enzymes that catalyze the removal of acetyl groups from the ε-N-acetyl lysine residues of histone proteins, thus counteracting the activity performed by histone acetyltransferares (HATs). Based on their involvement in different biological pathways, ranging from transcription to metabolism and genome stability, SIRT dysregulation was investigated in many diseases, such as cancer, neurodegenerative disorders, diabetes, and cardiovascular and autoimmune diseases. The elucidation of a consistent number of SIRT-ligand complexes helped to steer the identification of novel and more selective modulators. Due to the high diversity and quantity of the structural data thus far available, we reviewed some of the different ligands and structure-based methods that have recently been used to identify new promising SIRT1/2 modulators. The present review is structured into two sections: the first includes a comprehensive perspective of the successful computational approaches related to the discovery of SIRT1/2 inhibitors (SIRTIs); the second section deals with the most interesting SIRTIs that have recently appeared in the literature (from 2017). The data reported here are collected from different databases (SciFinder, Web of Science, Scopus, Google Scholar, and PubMed) using "SIRT", "sirtuin", and "sirtuin inhibitors" as keywords.
Collapse
Affiliation(s)
- Naomi Scarano
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (F.M.); (S.S.)
| | - Chiara Brullo
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (F.M.); (S.S.)
| | - Francesca Musumeci
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (F.M.); (S.S.)
| | - Enrico Millo
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy; (E.M.); (S.B.)
| | - Santina Bruzzone
- Department of Experimental Medicine, Section of Biochemistry, University of Genoa, Viale Benedetto XV 1, 16132 Genoa, Italy; (E.M.); (S.B.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Silvia Schenone
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (F.M.); (S.S.)
| | - Elena Cichero
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (N.S.); (F.M.); (S.S.)
| |
Collapse
|
2
|
Barone V. Accurate structures and spectroscopic parameters of α,α-dialkylated α-amino acids in the gas-phase: a joint venture of DFT and wave-function composite methods. Phys Chem Chem Phys 2023; 25:22768-22774. [PMID: 37591810 DOI: 10.1039/d3cp02503a] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Accurate computations of structural, conformational and spectroscopic properties in the gas phase have been performed for two α,α-dialkylated α-amino acids, namely aminoisobutyric acid and cyclopropylglycine. Thanks to the integration of modern double hybrid functionals and wave-function methods, several low-energy structures of the title molecules could be analyzed employing standard computer resources. The computed features of all the most stable conformers of the target amino acids closely match the corresponding spectroscopic parameters issued from microwave spectroscopic studies in the gas-phase. Together with their intrinsic interest, the accuracy of the results obtained with reasonable computer times paves the way for accurate investigations of other flexible bricks of life.
Collapse
Affiliation(s)
- Vincenzo Barone
- Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy.
| |
Collapse
|
3
|
Rahnasto-Rilla M, Järvenpää J, Huovinen M, Schroderus AM, Ihantola EL, Küblbeck J, Khadeer M, Moaddel R, Lahtela-Kakkonen M. Effects of galloflavin and ellagic acid on sirtuin 6 and its anti-tumorigenic activities. Biomed Pharmacother 2020; 131:110701. [PMID: 32905943 DOI: 10.1016/j.biopha.2020.110701] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
Sirtuin 6 (SIRT6), a member of sirtuin family (SIRT1-7), regulates distinct cellular functions; genome stability, DNA repair, and inflammation related diseases. Recently, we demonstrated that anthocyanidins in berries induce the catalytic activity of SIRT6. In this study, we explored the effects of Galloflavin and Ellagic acid, the most common polyphenols in berries, on SIRT6. SIRT6 deacetylation was investigated using HPLC and immunoblotting assays. The expression levels of SIRT6, glycolytic proteins and cellular metabolism were studied on human colon adenocarcinoma cells (Caco2). Molecular docking studies were carried out to study possible interactions of the compounds with sirtuins. Ellagic acid increased the deacetylase activity of SIRT6 by up to 50-fold; it showed moderate inhibition of SIRT1-3. Galloflavin and Ellagic acid showed anti-proliferative effects on Caco2. The compounds also upregulated SIRT6 expression whereas key proteins in glycolysis were downregulated. Galloflavin decreased glucose transporter 1 (GLUT1) expression, and Ellagic acid affected the expression of protein dehydrogenase kinase 1 (PDK1). Interestingly, both compounds caused reduction in glucose uptake and lactate production. Both Galloflavin and Ellagic acid were able to form hydrogen bonds with Asp188 and Gly6 in SIRT6. In this study, we showed that Galloflavin and Ellagic acid increased SIRT6 activity and decreased the expression of SIRT6 associated proteins involved in cancer development. Taken together, Galloflavin and Ellagic acid targeting SIRT6 activity may provide a new insight in the development of anti-cancer therapy.
Collapse
Affiliation(s)
| | - Joni Järvenpää
- School of Pharmacy, University of Eastern Finland, 70210, Kuopio, Finland
| | - Marjo Huovinen
- School of Pharmacy, University of Eastern Finland, 70210, Kuopio, Finland
| | - Anna-Mari Schroderus
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Emmi-Leena Ihantola
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jenni Küblbeck
- School of Pharmacy, University of Eastern Finland, 70210, Kuopio, Finland
| | - Mohammed Khadeer
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, 21224, USA
| | - Ruin Moaddel
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, 21224, USA.
| | | |
Collapse
|
4
|
Tenhunen J, Kokkola T, Huovinen M, Rahnasto-Rilla M, Lahtela-Kakkonen M. Impact of structurally diverse BET inhibitors on SIRT1. Gene 2020; 741:144558. [PMID: 32165310 DOI: 10.1016/j.gene.2020.144558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/08/2020] [Indexed: 12/15/2022]
Abstract
The epigenetic regulation of gene expression is controlled by various processes, of which one is histone acetylation. Many proteins control gene expression via histone acetylation. Those proteins include sirtuins (SIRTs) and bromodomain and extraterminal proteins (BETs), which are known to regulate same cellular processes and pathways. The aim of this study was to explore BET inhibitors' effects on SIRT1. Previously we showed that BET inhibitor (+)-JQ1 increases SIRT1 levels, but in the current study we used also other, structurally diverse BET inhibitors, I-BET151 and Pfi-1, and examined their effects on SIRT1 levels in two breast cancer cell lines. The results differed between the inhibitors and also between the cell lines. (+)-JQ1 had opposite effects on SIRT1 levels in the two cell lines, I-BET151 increased the levels in both cell lines, and Pfi-1 had no effect. In conclusion, the effect of structurally diverse BET inhibitors on SIRT1 levels is divergent, and the responses might also be cell type-dependent. These findings are important for all SIRT1 and BET inhibitor-related research, and they show that different BET inhibitors might have important individual effects.
Collapse
Affiliation(s)
- Jonna Tenhunen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1C, 70211 Kuopio, Finland
| | - Tarja Kokkola
- Institute of Clinical Medicine, University of Eastern Finland, Yliopistonranta 1C, 70211 Kuopio, Finland
| | - Marjo Huovinen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1C, 70211 Kuopio, Finland
| | - Minna Rahnasto-Rilla
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1C, 70211 Kuopio, Finland
| | - Maija Lahtela-Kakkonen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1C, 70211 Kuopio, Finland.
| |
Collapse
|
5
|
Seifert T, Malo M, Kokkola T, Stéen EJL, Meinander K, Wallén EAA, Jarho EM, Luthman K. A scaffold replacement approach towards new sirtuin 2 inhibitors. Bioorg Med Chem 2020; 28:115231. [PMID: 31848116 DOI: 10.1016/j.bmc.2019.115231] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/20/2019] [Accepted: 11/20/2019] [Indexed: 10/25/2022]
Abstract
Sirtuins (SIRT1-SIRT7) are an evolutionary conserved family of NAD+-dependent protein deacylases regulating the acylation state of ε-N-lysine residues of proteins thereby controlling key biological processes. Numerous studies have found association of the aberrant enzymatic activity of SIRTs with various diseases like diabetes, cancer and neurodegenerative disorders. Previously, we have shown that substituted 2-alkyl-chroman-4-one/chromone derivatives can serve as selective inhibitors of SIRT2 possessing an antiproliferative effect in two human cancer cell lines. In this study, we have explored the bioisosteric replacement of the chroman-4-one/chromone core structure with different less lipophilic bicyclic scaffolds to overcome problems associated to poor physiochemical properties due to a highly lipophilic substitution pattern required for achieve a good inhibitory effect. Various new derivatives based on the quinolin-4(1H)-one scaffold, bicyclic secondary sulfonamides or saccharins were synthesized and evaluated for their SIRT inhibitory effect. Among the evaluated scaffolds, the benzothiadiazine-1,1-dioxide-based compounds showed the highest SIRT2 inhibitory activity. Molecular modeling studies gave insight into the binding mode of the new scaffold-replacement analogues.
Collapse
Affiliation(s)
- Tina Seifert
- Department of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, SE-412 96 Göteborg, Sweden.
| | - Marcus Malo
- Department of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, SE-412 96 Göteborg, Sweden
| | - Tarja Kokkola
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - E Johanna L Stéen
- Department of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, SE-412 96 Göteborg, Sweden
| | - Kristian Meinander
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI- 00014 Helsinki, Finland
| | - Erik A A Wallén
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI- 00014 Helsinki, Finland
| | - Elina M Jarho
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Kristina Luthman
- Department of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, SE-412 96 Göteborg, Sweden
| |
Collapse
|
6
|
Alonso JL, Peña I, López JC, Alonso ER, Vaquero V. The Shape of the Simplest Non-proteinogenic Amino Acid α-Aminoisobutyric Acid (Aib). Chemistry 2019; 25:2288-2294. [DOI: 10.1002/chem.201805038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 11/09/2018] [Indexed: 11/12/2022]
Affiliation(s)
- José L. Alonso
- Grupo de Espectroscopía Molecular (GEM); Edificio Quifima; Laboratorio de Espectroscopia y Bioespectroscopia; Unidad Asociada CSIC; Parque Científico Uva; Universidad de Valladolid; 47011 Valladolid Spain
| | - Isabel Peña
- Grupo de Espectroscopía Molecular (GEM); Edificio Quifima; Laboratorio de Espectroscopia y Bioespectroscopia; Unidad Asociada CSIC; Parque Científico Uva; Universidad de Valladolid; 47011 Valladolid Spain
| | - Juan C. López
- Departamento de Química Física e Inorgánica; Facultad de, Ciencias; Universidad de Valladolid; 47011 Valladolid Spain
| | - Elena R. Alonso
- Grupo de Espectroscopía Molecular (GEM); Edificio Quifima; Laboratorio de Espectroscopia y Bioespectroscopia; Unidad Asociada CSIC; Parque Científico Uva; Universidad de Valladolid; 47011 Valladolid Spain
| | - Vanesa Vaquero
- Grupo de Espectroscopía Molecular (GEM); Edificio Quifima; Laboratorio de Espectroscopia y Bioespectroscopia; Unidad Asociada CSIC; Parque Científico Uva; Universidad de Valladolid; 47011 Valladolid Spain
| |
Collapse
|
7
|
Omar AM, Abdelghany TM, Abdel-Bakky MS, Alahdal AM, Radwan MF, El-Araby ME. Design, Synthesis and Antiproliferative Activities of Oxidative Stress Inducers Based on 2-Styryl-3,5-dihydro-4 H-imidazol-4-one Scaffold. Chem Pharm Bull (Tokyo) 2018; 66:967-975. [DOI: 10.1248/cpb.c18-00398] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Abdelsattar M. Omar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University
| | | | - Mohamed S. Abdel-Bakky
- Department of Pharmacology, Faculty of Pharmacy, Al-Azhar University
- Department of Pharmacology, Faculty of Pharmacy, Aljouf University
| | | | - Mohamed F. Radwan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University
| | - Moustafa E. El-Araby
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Helwan University
| |
Collapse
|
8
|
Rahnasto-Rilla MK, McLoughlin P, Kulikowicz T, Doyle M, Bohr VA, Lahtela-Kakkonen M, Ferrucci L, Hayes M, Moaddel R. The Identification of a SIRT6 Activator from Brown Algae Fucus distichus. Mar Drugs 2017; 15:E190. [PMID: 28635654 PMCID: PMC5484140 DOI: 10.3390/md15060190] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/13/2017] [Accepted: 06/16/2017] [Indexed: 11/17/2022] Open
Abstract
Brown seaweeds contain many bioactive compounds, including polyphenols, polysaccharides, fucosterol, and fucoxantin. These compounds have several biological activities, including anti-inflammatory, hepatoprotective, anti-tumor, anti-hypertensive, and anti-diabetic activity, although in most cases their mechanisms of action are not understood. In this study, extracts generated from five brown algae (Fucus dichitus, Fucus vesiculosus (Linnaeus), Cytoseira tamariscofolia, Cytoseira nodacaulis, Alaria esculenta) were tested for their ability to activate SIRT6 resulting in H3K9 deacetylation. Three of the five macroalgal extracts caused a significant increase of H3K9 deacetylation, and the effect was most pronounced for F. dichitus. The compound responsible for this in vitro activity was identified by mass spectrometry as fucoidan.
Collapse
Affiliation(s)
- Minna K Rahnasto-Rilla
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA.
- School of Pharmacy, University of Eastern Finland, Kuopio FI-70210, Finland.
| | - Padraig McLoughlin
- Food Biosciences Department, Teagasc Food Research Centre, Ashtown, Dublin 15, Ireland.
| | - Tomasz Kulikowicz
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA.
| | - Maire Doyle
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA.
| | - Vilhelm A Bohr
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA.
| | | | - Luigi Ferrucci
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA.
| | - Maria Hayes
- Food Biosciences Department, Teagasc Food Research Centre, Ashtown, Dublin 15, Ireland.
| | - Ruin Moaddel
- Biomedical Research Center, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA.
| |
Collapse
|
9
|
Khanfar MA, Taha MO. Unsupervised pharmacophore modeling combined with QSAR analyses revealed novel low micromolar SIRT2 inhibitors. J Mol Recognit 2017; 30. [PMID: 28299833 DOI: 10.1002/jmr.2623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 01/18/2017] [Accepted: 02/13/2017] [Indexed: 11/10/2022]
Abstract
Situin 2 (SIRT2) enzyme is a histone deacetylase that has important role in neuronal development. SIRT2 is clinically validated target for neurodegenerative diseases and some cancers. In this study, exhaustive unsupervised pharmacophore modeling was combined with quantitative structure-activity relationship (QSAR) analysis to explore the structural requirements for potent SIRT2 inhibitors using 146 known SIRT2 ligands. A computational workflow that combines genetic function algorithm with k-nearest neighbor or multiple linear regression was implemented to build self-consistent and predictive QSAR models based on combinations of pharmacophores and physicochemical descriptors. Successful pharmacophores were complemented with exclusion spheres to optimize their receiver operating characteristic curve profiles. Optimal QSAR models and their associated pharmacophore hypotheses were experimentally validated by identification and in vitro evaluation of several new promising SIRT2 inhibitory leads retrieved from the National Cancer Institute structural database. The most potent hit illustrated IC50 value of 5.4μM. The chemical structures of active hits were validated by proton nuclear magnetic resonance and mass spectroscopy.
Collapse
Affiliation(s)
- Mohammad A Khanfar
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Univerity of Jordan, Amman, Jordan
| | - Mutasem O Taha
- Drug Discovery Unit, Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, Jordan
| |
Collapse
|
10
|
Dietz C, Hart TK, Nemati R, Yao X, Nichols FC, Smith MB. Structural verification via convergent total synthesis of dipeptide–lipids isolated from Porphyromonas gingivalis. Tetrahedron 2016. [DOI: 10.1016/j.tet.2016.10.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
11
|
Sun Y, Zhou H, Zhu H, Leung SW. Ligand-based virtual screening and inductive learning for identification of SIRT1 inhibitors in natural products. Sci Rep 2016; 6:19312. [PMID: 26805727 PMCID: PMC4726279 DOI: 10.1038/srep19312] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 12/09/2015] [Indexed: 02/04/2023] Open
Abstract
Sirtuin 1 (SIRT1) is a nicotinamide adenine dinucleotide-dependent deacetylase, and its dysregulation can lead to ageing, diabetes, and cancer. From 346 experimentally confirmed SIRT1 inhibitors, an inhibitor structure pattern was generated by inductive logic programming (ILP) with DMax Chemistry Assistant software. The pattern contained amide, amine, and hetero-aromatic five-membered rings, each of which had a hetero-atom and an unsubstituted atom at a distance of 2. According to this pattern, a ligand-based virtual screening of 1 444 880 active compounds from Chinese herbs identified 12 compounds as inhibitors of SIRT1. Three compounds (ZINC08790006, ZINC08792229, and ZINC08792355) had high affinity (-7.3, -7.8, and -8.6 kcal/mol, respectively) for SIRT1 as estimated by molecular docking software AutoDock Vina. This study demonstrated a use of ILP and background knowledge in machine learning to facilitate virtual screening.
Collapse
Affiliation(s)
- Yunan Sun
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Hui Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Hongmei Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Siu-wai Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.,School of Informatics, University of Edinburgh, Edinburgh EH8 9AB, United Kingdom
| |
Collapse
|
12
|
Boehmeriasin A as new lead compound for the inhibition of topoisomerases and SIRT2. Eur J Med Chem 2015; 92:766-75. [DOI: 10.1016/j.ejmech.2015.01.038] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 01/15/2015] [Accepted: 01/19/2015] [Indexed: 12/12/2022]
|
13
|
DeBerardinis AM, Raccuia DS, Thompson EN, Maschinot CA, Kyle Hadden M. Vitamin D3 analogues that contain modified A- and seco-B-rings as hedgehog pathway inhibitors. Eur J Med Chem 2015; 93:156-71. [PMID: 25676864 DOI: 10.1016/j.ejmech.2015.01.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/21/2015] [Accepted: 01/24/2015] [Indexed: 10/24/2022]
Abstract
The hedgehog (Hh) signaling pathway is a developmental signaling pathway that has been implicated as a target for anti-cancer drug development in a variety of human malignancies. Several natural and synthetic vitamin D-based seco-steroids have been identified as potent inhibitors of Hh signaling with chemotherapeutic potential. These include the previously characterized analogue 4, which contains the northern CD-ring/side chain region of vitamin D3 (VD3) linked to an aromatic A-ring mimic through an ester bond. To further explore structure-activity relationships for this class of VD3-based Hh pathway inhibitors, we have designed, synthesized and evaluated several series of compounds that modify the length, composition, and stereochemical orientation of the ester linker. These studies have identified compounds 54 and 55, which contain an amine linker and an aromatic A-ring incorporating a para-phenol, as new lead compounds with enhanced potency against the Hh pathway (IC50 values = 0.40 and 0.32 μM, respectively).
Collapse
Affiliation(s)
- Albert M DeBerardinis
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Unit 3092, Storrs, CT 06269-3092, USA
| | - Daniel S Raccuia
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Unit 3092, Storrs, CT 06269-3092, USA
| | - Evrett N Thompson
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Unit 3092, Storrs, CT 06269-3092, USA
| | - Chad A Maschinot
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Unit 3092, Storrs, CT 06269-3092, USA
| | - M Kyle Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Unit 3092, Storrs, CT 06269-3092, USA.
| |
Collapse
|
14
|
Chuang YC, Chang CH, Lin JT, Yang CN. Molecular modelling studies of sirtuin 2 inhibitors using three-dimensional structure-activity relationship analysis and molecular dynamics simulations. MOLECULAR BIOSYSTEMS 2014; 11:723-33. [PMID: 25502412 DOI: 10.1039/c4mb00620h] [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
Sirtuin 2 (SIRT2) is a nicotinamide-adenine-dinucleotide-dependent histone deacetylase that plays a vital role in various biological processes related to DNA regulation, metabolism, and longevity. Recent studies on SIRT2 have indicated its therapeutic potential for neurodegenerative diseases such as Parkinson's disease. In this study, a series of SIRT2 inhibitors with a 2-anilinobenzamide core was analysed using a combination of molecular modelling techniques. A three-dimensional structure-activity relationship (3D-QSAR) model adopting a comparative molecular field analysis (CoMFA) method with a non-cross-validated correlation coefficient R(2) = 0.992 (for training set) and a correlation coefficient Rtest(2) = 0.804 (for test set) was generated to determine the structural requirements for inhibitory activity. Furthermore, we employed molecular dynamics (MD) simulations and the molecular mechanics/generalized Born surface area (MM/GBSA) method to compare the binding modes of a potent and selective compound interacting with SIRT1, SIRT2, and SIRT3 and also their binding free energies to shed light on the selectivity of the footing of structural and energetic investigations. The steric and electrostatic contour maps from the 3D-QSAR analysis identified several key interactions also observed in the MD simulations. According to these results, we provide guidelines for developing novel potent and selective SIRT2 inhibitors.
Collapse
Affiliation(s)
- Yu-Chung Chuang
- Department of Life Sciences, National University of Kaohsiung, 700, Kaohsiung University Road, Nan-Tzu District 811, Kaohsiung, Taiwan.
| | | | | | | |
Collapse
|
15
|
Seifert T, Malo M, Kokkola T, Engen K, Fridén-Saxin M, Wallén EAA, Lahtela-Kakkonen M, Jarho EM, Luthman K. Chroman-4-one- and Chromone-Based Sirtuin 2 Inhibitors with Antiproliferative Properties in Cancer Cells. J Med Chem 2014; 57:9870-88. [DOI: 10.1021/jm500930h] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Tina Seifert
- Department
of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, Kemivagen 10, SE-412
96 Göteborg, Sweden
| | - Marcus Malo
- Department
of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, Kemivagen 10, SE-412
96 Göteborg, Sweden
| | - Tarja Kokkola
- School
of Pharmacy, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Karin Engen
- Department
of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, Kemivagen 10, SE-412
96 Göteborg, Sweden
| | - Maria Fridén-Saxin
- Department
of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, Kemivagen 10, SE-412
96 Göteborg, Sweden
| | - Erik A. A. Wallén
- Division
of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FIN-00014 Helsinki, Finland
| | | | - Elina M. Jarho
- School
of Pharmacy, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Kristina Luthman
- Department
of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, Kemivagen 10, SE-412
96 Göteborg, Sweden
| |
Collapse
|
16
|
Sensitive and photo-triggered transformation of hierarchical helices assembled from achiral bolaamphiphiles. J Colloid Interface Sci 2014; 435:1-7. [DOI: 10.1016/j.jcis.2014.08.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/06/2014] [Accepted: 08/07/2014] [Indexed: 11/20/2022]
|
17
|
Pulla VK, Alvala M, Sriram DS, Viswanadha S, Sriram D, Yogeeswari P. Structure-based drug design of small molecule SIRT1 modulators to treat cancer and metabolic disorders. J Mol Graph Model 2014; 52:46-56. [DOI: 10.1016/j.jmgm.2014.06.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 05/05/2014] [Accepted: 06/17/2014] [Indexed: 11/29/2022]
|
18
|
Barontini M, Bernini R, Carastro I, Gentili P, Romani A. Synthesis and DPPH radical scavenging activity of novel compounds obtained from tyrosol and cinnamic acid derivatives. NEW J CHEM 2014. [DOI: 10.1039/c3nj01180a] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Novel compounds exhibiting DPPH radical scavenging activity were synthesised. The key step was the trifluoroacetic acid-mediated hydroarylation of cinnamic ester with tyrosol.
Collapse
Affiliation(s)
- Maurizio Barontini
- Dipartimento di Scienze e Tecnologie per l'Agricoltura, le Foreste, la Natura e l'Energia (DAFNE)
- Università degli Studi della Tuscia
- 01100 Viterbo, Italy
| | - Roberta Bernini
- Dipartimento di Scienze e Tecnologie per l'Agricoltura, le Foreste, la Natura e l'Energia (DAFNE)
- Università degli Studi della Tuscia
- 01100 Viterbo, Italy
| | - Isabella Carastro
- Dipartimento di Scienze e Tecnologie per l'Agricoltura, le Foreste, la Natura e l'Energia (DAFNE)
- Università degli Studi della Tuscia
- 01100 Viterbo, Italy
| | - Patrizia Gentili
- Dipartimento di Chimica and IMC-CNR Sezione Meccanismi di Reazione
- Università degli Studi La Sapienza
- 00185 Roma, Italy
| | | |
Collapse
|
19
|
Christodoulou MS, Sacchetti A, Ronchetti V, Caufin S, Silvani A, Lesma G, Fontana G, Minicone F, Riva B, Ventura M, Lahtela-Kakkonen M, Jarho E, Zuco V, Zunino F, Martinet N, Dapiaggi F, Pieraccini S, Sironi M, Dalla Via L, Gia OM, Passarella D. Quinazolinecarboline alkaloid evodiamine as scaffold for targeting topoisomerase I and sirtuins. Bioorg Med Chem 2013; 21:6920-8. [PMID: 24103429 DOI: 10.1016/j.bmc.2013.09.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/09/2013] [Accepted: 09/11/2013] [Indexed: 12/12/2022]
Abstract
This paper reports the synthesis of a series of evodiamine derivatives. We assayed the ability to inhibit cell growth on three human tumour cell lines (H460, MCF-7 and HepG2) and we evaluated the capacity to interfere with the catalytic activity of topoisomerase I both by the relaxation assay and the occurrence of the cleavable complex. Moreover, whose effect on sirtuins 1, 2 and 3 was investigated. Finally, molecular docking analyses were performed in an attempt to rationalize the biological results.
Collapse
Affiliation(s)
- Michael S Christodoulou
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Mellini P, Kokkola T, Suuronen T, Salo HS, Tolvanen L, Mai A, Lahtela-Kakkonen M, Jarho EM. Screen of pseudopeptidic inhibitors of human sirtuins 1-3: two lead compounds with antiproliferative effects in cancer cells. J Med Chem 2013; 56:6681-95. [PMID: 23927550 DOI: 10.1021/jm400438k] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In the past few years sirtuins have gained growing attention for their involvement in many biological processes such as cellular metabolism, apoptosis, aging and inflammation. In this contribution, we report the synthesis of a library of thioacetylated pseudopeptides that were screened against human sirtuins 1-3 to reveal their in vitro inhibition activities. Molecular modeling studies were performed to acquire data about the binding modes of the inhibitors. Three sirtuin inhibitors were subjected to cellular studies, and all of them showed an increase in acetylation of Lys382 of p53 after DNA damage. Furthermore, two of the compounds were able to inhibit both A549 lung carcinoma and MCF-7 breast carcinoma cell growth in micromolar concentration with the ability to arrest cancer cell cycle in the G1 phase.
Collapse
Affiliation(s)
- Paolo Mellini
- School of Pharmacy and ∥Department of Neurology, Institute of Clinical Medicine , University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Seidel C, Florean C, Schnekenburger M, Dicato M, Diederich M. Chromatin-modifying agents in anti-cancer therapy. Biochimie 2012; 94:2264-79. [DOI: 10.1016/j.biochi.2012.05.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 05/14/2012] [Indexed: 01/12/2023]
|
22
|
Fridén-Saxin M, Seifert T, Landergren MR, Suuronen T, Lahtela-Kakkonen M, Jarho EM, Luthman K. Synthesis and evaluation of substituted chroman-4-one and chromone derivatives as sirtuin 2-selective inhibitors. J Med Chem 2012; 55:7104-13. [PMID: 22746324 PMCID: PMC3426190 DOI: 10.1021/jm3005288] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
A series of substituted chromone/chroman-4-one derivatives
has
been synthesized and evaluated as novel inhibitors of SIRT2, an enzyme
involved in aging-related diseases, e.g., neurodegenerative disorders.
The analogues were efficiently synthesized in a one-step procedure
including a base-mediated aldol condensation using microwave irradiation.
The most potent compounds, with inhibitory concentrations in the low
micromolar range, were substituted in the 2-, 6-, and 8-positions.
Larger, electron-withdrawing substituents in the 6- and 8-positions
were favorable. The most potent inhibitor of SIRT2 was 6,8-dibromo-2-pentylchroman-4-one
with an IC50 of 1.5 μM. The synthesized compounds
show high selectivity toward SIRT2 over SIRT1 and SIRT3 and represent
an important starting point for the development of novel SIRT2 inhibitors.
Collapse
Affiliation(s)
- Maria Fridén-Saxin
- Department of Chemistry and Molecular Biology, Medicinal Chemistry, University of Gothenburg, SE-412 96 Göteborg, Sweden
| | | | | | | | | | | | | |
Collapse
|
23
|
Sakkiah S, Thangapandian S, Park C, Son M, Lee KW. Molecular docking and dynamics simulation, receptor-based hypothesis: application to identify novel sirtuin 2 inhibitors. Chem Biol Drug Des 2012; 80:315-27. [PMID: 22564257 DOI: 10.1111/j.1747-0285.2012.01406.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Sirtuin, NAD(+)-dependent histone deacetylase enzyme, emerged as a potential therapeutic target, and modulations by small molecules could be effective drugs for various diseases. Owing to the absence of complex structure of sirtuin 2 (SIRT2), sirtinol was docked in the NAD(+) binding site and subjected to 5-nseconds molecular dynamics (MD) simulation. LigandScout was used to develop hypotheses based on 3-representative SIRT2 complex structures from MD. Three structure-based hypotheses are generated and merged to form dynamics hypothesis. The dynamics hypothesis was validated using test and decoy sets. The results showed that dynamic hypothesis represents the complementary features of SIRT2 active site. Dynamic hypothesis was used to screen ChemDiv database, and hits were filtered through ADMET, rule of five, and two different molecular docking studies. Finally, 21 molecules were selected as potent leads based on consensus score from LigandFit, Gold fitness score, binding affinity from VINA as well as based on the important interactions with critical residues in SIRT2 active site. Hence, we suggest that the dynamic hypothesis will be reliable in the identification of SIRT2 new lead as well as to reduce time and cost in the drug discovery process.
Collapse
Affiliation(s)
- Sugunadevi Sakkiah
- 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), 501 Jinju-daero, Gazha-dong, Jinju 660-701, Korea
| | | | | | | | | |
Collapse
|
24
|
Suzuki T, Khan MNA, Sawada H, Imai E, Itoh Y, Yamatsuta K, Tokuda N, Takeuchi J, Seko T, Nakagawa H, Miyata N. Design, synthesis, and biological activity of a novel series of human sirtuin-2-selective inhibitors. J Med Chem 2012; 55:5760-73. [PMID: 22642300 DOI: 10.1021/jm3002108] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Selective inhibitors of human sirtuin 2 (SIRT2), a deacetylase, are candidate therapeutic agents for neurodegenerative diseases such as Parkinson's disease and Huntington's disease as well as potential tools for elucidating the biological functions of SIRT2. On the basis of homology models of SIRT1 and SIRT2, we designed and prepared a series of 2-anilinobenzamide analogues. Enzyme assays using recombinant SIRT1 and SIRT2 revealed that 3'-phenethyloxy-2-anilinobenzamide analogues such as 33a and 33i are potent and selective SIRT2 inhibitors, showing more than 3.5-fold greater SIRT2-inhibitory activity and more than 35-fold greater SIRT2-selectivity compared with AGK2 (3), a previously reported SIRT2-selective inhibitor. Compound 33a also induced a dose-dependent selective increase of α-tubulin acetylation in human colon cancer HCT116 cells, indicating selective inhibition of SIRT2 in the cells. These 3'-phenethyloxy-2-anilinobenzamide derivatives represent an entry into a new class of SIRT2-selective inhibitors.
Collapse
Affiliation(s)
- Takayoshi Suzuki
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 13 Taishogun Nishitakatsukasa-cho, Kita-ku, Kyoto 603-8334, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Alvala M, Bhatnagar S, Ravi A, Jeankumar VU, Manjashetty TH, Yogeeswari P, Sriram D. Novel acridinedione derivatives: design, synthesis, SIRT1 enzyme and tumor cell growth inhibition studies. Bioorg Med Chem Lett 2012; 22:3256-60. [PMID: 22464458 DOI: 10.1016/j.bmcl.2012.03.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 02/27/2012] [Accepted: 03/07/2012] [Indexed: 10/28/2022]
Abstract
A new scaffold N-(9-(ortho/meta/para-(benzyloxy)phenyl)-3,3,6,6-tetramethyl-1,8-dioxo-1,2,3,4,5,6,7,8-octahydroacridin-10(9H)-yl) isonicotinamide (H1-3) was discovered as a hSIRT1 inhibitor through virtual screening of in-house database. Based on these hits, a library of compounds were designed, synthesized and tested for in vitro hSIRT1 activity. The most potent compound 4d in the series showed a significant inhibition of SIRT1 activity. Further antitumor studies of compound 4d, showed a dose dependent increase in acetylation of p53K382 and decrease in SIRT1 with an IC(50) of 0.25 μM in MDA-MB231 breast cancer cell lines. Individual 3D-QSAR analysis using Schrödinger showed distribution of hydrophobic and non polar positive co-efficient at ortho position essential for bioactivity based on 4d.
Collapse
Affiliation(s)
- Mallika Alvala
- Drug Discovery Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science, Pilani-Hyderabad Campus, Hyderabad 500 078, India
| | | | | | | | | | | | | |
Collapse
|
26
|
Liu X, Wang T, Liu M. Interfacial assembly of a series of cinnamoyl-containing bolaamphiphiles: spacer-controlled packing, photochemistry, and odd-even effect. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2012; 28:3474-3482. [PMID: 22272776 DOI: 10.1021/la204653b] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
A series of bolaamphiphiles with 4-hydroxycinnamoyl head groups and different length of the alkyl spacers (n = 6-12) were designed to investigate their photochemistry in the organized films obtained from the air/water interface. It has been found that both the length and odd-even number of the spacers can finely tune the molecular packing as well as the photochemistry. When the spacer length was changed from 6 to 12 methylene units, the assemblies changed from J aggregate to H aggregate. The molecules with even-numbered polymethylene spacer tend to form three-dimensional nanorod structure at the air/water interface. For the assembly of derivatives with odd-numbered spacers, diverse morphologies such as nanospirals and nanofibers were observed depending on the chain length and the surface pressures. The different packing of bolaamphiphiles could subsequently affect the photochemistry of the cinnamoyl groups in the organized films. The spacer effect in the assembly can be understood from the cooperation between H-bond of the phenolic hydroxyl and the amide groups, π-π stacking as well as the hydrophobic interactions of the alkyl spacer. A packing model was proposed to explain the phenomenon.
Collapse
Affiliation(s)
- Xufei Liu
- Beijing National Laboratory for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | | | | |
Collapse
|
27
|
Abstract
This review focuses on the progress in the development of the second generation of epigenetic modifiers able to modulate histone marks, and restore normal gene transcription.
Collapse
Affiliation(s)
- Philip Jones
- Institute for Applied Cancer Sciences
- MD Anderson Cancer Center
- Houston
- USA
| |
Collapse
|
28
|
Huhtiniemi T, Salo HS, Suuronen T, Poso A, Salminen A, Leppänen J, Jarho E, Lahtela-Kakkonen M. Structure-based design of pseudopeptidic inhibitors for SIRT1 and SIRT2. J Med Chem 2011; 54:6456-68. [PMID: 21895016 DOI: 10.1021/jm200590k] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The lack of substrate-bound crystal structures of SIRT1 and SIRT2 complicates the drug design for these targets. In this work, we aim to study whether SIRT3 could serve as a target structure in the design of substrate based pseudopeptidic inhibitors of SIRT1 and SIRT2. We created a binding hypothesis for pseudopeptidic inhibitors, synthesized a series of inhibitors, and studied how well the fulfillment of the binding criteria proposed by the hypothesis correlated with the in vitro inhibitory activities. The chosen approach was further validated by studying docking results between 12 different SIRT3, Sir2Tm, SIRT1 and SIRT2 X-ray structures and homology models in different conformational forms. It was concluded that the created binding hypothesis can be used in the design of the substrate based inhibitors of SIRT1 and SIRT2 although there are some reservations, and it is better to use the substrate-bound structure of SIRT3 instead of the available apo-SIRT2 as the target structure.
Collapse
Affiliation(s)
- Tero Huhtiniemi
- School of Pharmacy, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Liu X, Wang T, Liu M. Interfacial assembly of cinnamoyl-terminated bolaamphiphiles through the air/water interface: headgroup-dependent assembly, supramolecular nanotube and photochemical sewing. Phys Chem Chem Phys 2011; 13:16520-9. [PMID: 21860882 DOI: 10.1039/c1cp21561b] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A series of cinnamoyl-terminated bolaamphiphiles were synthesized and their assemblies at the air/water interface were investigated. It was found that the assembly behaviour depended on the substituted groups on the cinnamoyl unit. The bolaamphiphile with 4-hydroxycinnamoyl head groups (HCDA) was found to assemble into a supramolecular nanotube, while the others formed only layer-structured films. Moreover, the nanotube formed from HCDA showed supramolecular chirality due to the symmetry breaking. Both the layered films and the nanotubes showed photochemical dimerization upon UV irradiation, which were studied from the UV-Vis, FT-IR spectral and MALDI-TOF MS analysis. Interestingly, such dimerization behavior of the cinnamoyl group could be used to stabilize the nanotube of HCDAvia photochemical sewing. During such a process both the supramolecular chirality and the tubular shapes were kept. Remarkably, such a photochemical sewed chiral nanotube could further induce the chirality of an achiral porphyrin derivative assembled on it, and produced the induced chirality without using any chiral molecules.
Collapse
Affiliation(s)
- Xufei Liu
- Beijing National Laboratory for Molecular Science (BNLMS), CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, The Chinese Academy of Sciences, Beijing, People's Republic of China
| | | | | |
Collapse
|
30
|
Design of a novel nucleoside analog as potent inhibitor of the NAD dependent deacetylase, SIRT2. SYSTEMS AND SYNTHETIC BIOLOGY 2011; 4:257-63. [PMID: 22132052 DOI: 10.1007/s11693-011-9069-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 02/03/2011] [Indexed: 10/18/2022]
Abstract
Sirtuins (class III histone deacetylase) are evolutionarily conserved NAD(+)-dependent enzymes that catalyze the deacetylation of acetyl-lysine residues of histones and other target proteins. Because of their associations in various pathophysiological conditions, the identification of small molecule modulators has been of significant interest. In the present study, virtual screening was carried out with NCI Diversity Set II using crystal structure of hSIRT2 (PDB ID: 1J8F) as a model for the docking procedure to find potential compounds, which were then subjected to experimental tests for their in vitro SIRT2 inhibitory activity. One of the 40 compounds tested, NSC671136 (IUPAC name: 6-Acetyl-4-oxo-1,3-diphenyl-2-thioxo-1,2,3,4-tetrahydrothieno[2,3-d]pyrimidin-5-yl 2,4-dichlorobenzoate) has structurally unique scaffold, showed strong inhibitory activity towards SIRT2 with IC(50) of ~8.7 μM and to a lesser extent on SIRT1 activity. The reported compound is substantially potent compared to the published SIRT2 inhibitors and serves as an excellent base for future lead development.
Collapse
|
31
|
Huhtiniemi T, Suuronen T, Lahtela-Kakkonen M, Bruijn T, Jääskeläinen S, Poso A, Salminen A, Leppänen J, Jarho E. N(epsilon)-Modified lysine containing inhibitors for SIRT1 and SIRT2. Bioorg Med Chem 2010; 18:5616-25. [PMID: 20630764 DOI: 10.1016/j.bmc.2010.06.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 06/10/2010] [Accepted: 06/11/2010] [Indexed: 10/19/2022]
Abstract
Sirtuins catalyze the NAD(+) dependent deacetylation of N(epsilon)-acetyl lysine residues to nicotinamide, O'-acetyl-ADP-ribose (OAADPR) and N(epsilon)-deacetylated lysine. Here, an easy-to-synthesize Ac-Ala-Lys-Ala sequence has been used as a probe for the screening of novel N(epsilon)-modified lysine containing inhibitors against SIRT1 and SIRT2. N(epsilon)-Selenoacetyl and N(epsilon)-isothiovaleryl were the most potent moieties found in this study, comparable to the widely studied N(epsilon)-thioacetyl group. The N(epsilon)-3,3-dimethylacryl and N(epsilon)-isovaleryl moieties gave significant inhibition in comparison to the N(epsilon)-acetyl group present in the substrates. In addition, the studied N(epsilon)-alkanoyl, N(epsilon)-alpha,beta-unsaturated carbonyl and N(epsilon)-aroyl moieties showed that the acetyl binding pocket can accept rather large groups, but is sensitive to even small changes in electronic and steric properties of the N(epsilon)-modification. These results are applicable for further screening of N(epsilon)-acetyl analogues.
Collapse
Affiliation(s)
- Tero Huhtiniemi
- School of Pharmacy, University of Eastern Finland, Kuopio Campus, PO Box 1627, 70211 Kuopio, Finland.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
|
33
|
Cen Y. Sirtuins inhibitors: the approach to affinity and selectivity. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2009; 1804:1635-44. [PMID: 19931429 DOI: 10.1016/j.bbapap.2009.11.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2009] [Revised: 11/07/2009] [Accepted: 11/10/2009] [Indexed: 10/20/2022]
Abstract
Accumulating evidence has indicated the importance of sirtuins (class III histone deacetylases) in various biological processes. Their potential roles in metabolic and neurodegenerative diseases have encouraged scientists to seek potent and selective sirtuin inhibitors to investigate their biological functions with a view to eventual new therapeutic treatments. This article surveys current knowledge of sirtuin inhibitors including those discovered via high-throughput screening (HST) or via mechanism-based drug design from synthetic or natural sources. Their inhibitory affinity, selectivities, and possible inhibition mechanisms are discussed.
Collapse
Affiliation(s)
- Yana Cen
- Department of Pharmacology, Weill Medical College of Cornell University, New York, NY 10065, USA.
| |
Collapse
|
34
|
Sanders BD, Jackson B, Brent M, Taylor AM, Schreiber SL, Howitz K, Marmorstein R. Identification and characterization of novel sirtuin inhibitor scaffolds. Bioorg Med Chem 2009; 17:7031-41. [PMID: 19734050 PMCID: PMC2929362 DOI: 10.1016/j.bmc.2009.07.073] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 07/23/2009] [Accepted: 07/26/2009] [Indexed: 12/31/2022]
Abstract
The sirtuin proteins are broadly conserved NAD(+)-dependent deacetylases that are implicated in diverse biological processes including DNA recombination and repair, transcriptional silencing, longevity, apoptosis, axonal protection, insulin signaling, and fat mobilization. Because of these associations, the identification of small molecule sirtuin modulators has been of significant interest. Here we report on high throughput screening against the yeast sirtuin, Hst2, leading to the identification of four unique inhibitor scaffolds that also inhibit the human sirtuins, SIRT1-3, and are able to inhibit telomeric silencing of yeast Sir2 in vivo. The identified inhibitor scaffolds range in potency from IC(50) values of 6.5-130 microM against Hst2. Each of the inhibitor scaffolds binds reversibly to the enzyme, and kinetic analysis reveals that each of the inhibitors is non-competitive with respect to both acetyl-lysine and NAD(+) binding. Limited SAR analysis of the scaffolds also identifies which functional groups may be important for inhibition. These sirtuin inhibitors are low molecular weight and well-suited for lead molecule optimization, making them useful chemical probes to study the mechanism and biological roles of sirtuins and potential starting points for optimization into therapeutics.
Collapse
Affiliation(s)
- Brandi D. Sanders
- The Wistar Institute, University of Pennsylvania, Philadelphia, PA, 19104 USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Brittany Jackson
- The Wistar Institute, University of Pennsylvania, Philadelphia, PA, 19104 USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Michael Brent
- The Wistar Institute, University of Pennsylvania, Philadelphia, PA, 19104 USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Alexander M. Taylor
- Howard Hughes Medical Institute, Broad Institute of Harvard and MIT, Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138
| | - Stuart L. Schreiber
- Howard Hughes Medical Institute, Broad Institute of Harvard and MIT, Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138
| | - Konrad Howitz
- ENZO Life Sciences, Inc., 5120 Butler Pike Plymouth Meeting, PA 19462
| | - Ronen Marmorstein
- The Wistar Institute, University of Pennsylvania, Philadelphia, PA, 19104 USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104 USA
| |
Collapse
|
35
|
Shoba B, Lwin ZM, Ling LS, Bay BH, Yip GW, Kumar SD. Function of sirtuins in biological tissues. Anat Rec (Hoboken) 2009; 292:536-43. [PMID: 19301279 DOI: 10.1002/ar.20875] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Sirtuins are protein deacetylases, which are dependent on nicotine adenine dinucleotide. They are phylogenetically conserved from bacteria to humans. Seven sirtuin proteins localized in a wide variety of subcellular locations have been identified in the human genome. The most important known function of sirtuins is their regulation of transcriptional repression, mediated through binding of a complex containing sirtuins and other proteins. Studies have shown that sirtuins have pathophysiological relevance to neurodegeneration, muscle differentiation, inflammation, obesity, and cancer. In addition, sirtuin activity extends the lifespan of several organisms. In this review, we discuss the mode(s) of action of sirtuins, and their biological role(s) in health and disease.
Collapse
Affiliation(s)
- Balaji Shoba
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
BACKGROUND The sirtuin family of deacetylase enzymes comprises seven proteins (SIRT1-7) that are dependent on NAD(+) for their activity. Three proteins are located in the nucleus, three in the mitochondria and only one is predominantly cytoplasmic. Caloric restriction and oxidative stress generally up-regulate their expression. SIRT1, the orthologue of yeast Sir2, is the mammalian sirtuin that has been most extensively studied to date. Among other targets, SIRT1 down-regulates the activity of the nuclear transcription factor p53, being this related with an increase in lifespan and cell survival associated to stress resistance. OBJECTIVE Because sirtuin modulation could have beneficial effects on several human diseases, there is a growing interest in the discovery and development of small molecules that modify its activity. This review will be focused on sirtuin inhibitors. CONCLUSIONS Several specific inhibitors of SIRT1 have been described. These compounds could be mainly useful for the treatment of cancers by increasing p53 activity that stops the formation of tumours and induces apoptosis. A p53-independent massive induction of apoptosis has been also described for one inhibitor. In addition, a potent and selective SIRT2 inhibitor that ameliorates the alpha-synuclein fibril formation in Parkinson disease has been proposed to treat this kind of neurodegenerative disease.
Collapse
Affiliation(s)
- Francisco J Alcaín
- Universidad de Córdoba, Departamento de Biología Celular, Fisiología e Inmunología, Facultad de Ciencias, Campus Rabanales, Edificio Severo Ochoa, Córdoba, Spain
| | | |
Collapse
|
37
|
Struble JR, Bode JW. Formal synthesis of salinosporamide A via NHC-catalyzed intramolecular lactonization. Tetrahedron 2009; 65:4957-4967. [PMID: 20606761 PMCID: PMC2896295 DOI: 10.1016/j.tet.2009.03.103] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
An N-heterocyclic carbene (NHC) catalyzed intramolecular lactonization to prepare densely functionalized bicyclic γ-lactam-γ-lactone adducts from enals is reported. This method has been applied to the formal synthesis of salinosporamide A, a potent 20S proteasome inhibitor and anti-cancer therapeutic.
Collapse
Affiliation(s)
- Justin R. Struble
- Roy and Diana Vagelos Laboratories Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6354, USA Dedicated to Professor Michael Krische in honor of his 2008 Tetrahedron Young Investigator Award
| | - Jeffrey W. Bode
- Roy and Diana Vagelos Laboratories Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6354, USA Dedicated to Professor Michael Krische in honor of his 2008 Tetrahedron Young Investigator Award
| |
Collapse
|
38
|
Zhang Y, Au Q, Zhang M, Barber JR, Ng SC, Zhang B. Identification of a small molecule SIRT2 inhibitor with selective tumor cytotoxicity. Biochem Biophys Res Commun 2009; 386:729-33. [PMID: 19559674 DOI: 10.1016/j.bbrc.2009.06.113] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Accepted: 06/21/2009] [Indexed: 12/17/2022]
Abstract
As a member of the class III histone deacetylases, Sirtuin-2 (SIRT2) is critical in cell cycle regulation which makes it a potential target for cancer therapeutics. In this study, we identified a novel SIRT2 inhibitor, AC-93253, with IC(50) of 6 microM in vitro. The compound is selective, inhibiting SIRT2 7.5- and 4-fold more potently than the closely related SIRT1 and SIRT3, respectively. AC-93253 significantly enhanced acetylation of tubulin, p53, and histone H4, confirming SIRT2 and SIRT1 as its cellular targets. AC-93253 as a single agent exhibited submicromolar selective cytotoxicity towards all four tumor cell lines tested with a therapeutic window up to 200-fold, comparing to any of the three normal cell types tested. Results from high content analysis suggested that AC-93253 significantly triggered apoptosis. Taken together, SIRT2 selective inhibitor AC-93253 may serve as a novel chemical scaffold for structure-activity relationship study and future lead development.
Collapse
Affiliation(s)
- Yingjia Zhang
- Department of Biology, CytRx Corporation, 3030 Bunker Hill Street, Suite 101, San Diego, CA 92109, USA
| | | | | | | | | | | |
Collapse
|
39
|
Epi-drugs to fight cancer: from chemistry to cancer treatment, the road ahead. Int J Biochem Cell Biol 2008; 41:199-213. [PMID: 18790076 DOI: 10.1016/j.biocel.2008.08.020] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Revised: 08/08/2008] [Accepted: 08/08/2008] [Indexed: 12/16/2022]
Abstract
In addition to genetic events, a variety of epigenetic events have been widely reported to contribute to the onset of many diseases including cancer. DNA methylation and histone modifications (such as acetylation, methylation, sumoylation, and phosphorylation) involving chromatin remodelling are among the most studied epigenetic mechanisms for regulation of gene expression leading, when altered, to some diseases. Epigenetic therapy tries to reverse the aberrations followed to the disruption of the balance of the epigenetic signalling ways through the use of both natural compounds and synthetic molecules, active on specific epi-targets. Such epi-drugs are, for example, inhibitors of DNA methyltransferases, histone deacetylases, histone acetyltransferases, histone methyltransferases, and histone demethylases. In this review we will focus on the chemical aspects of such molecules, joined to their effective (or potential) application in cancer therapy.
Collapse
|
40
|
Kiviranta PH, Salo HS, Leppänen J, Rinne VM, Kyrylenko S, Kuusisto E, Suuronen T, Salminen A, Poso A, Lahtela-Kakkonen M, Wallén EAA. Characterization of the binding properties of SIRT2 inhibitors with a N-(3-phenylpropenoyl)-glycine tryptamide backbone. Bioorg Med Chem 2008; 16:8054-62. [PMID: 18701307 DOI: 10.1016/j.bmc.2008.07.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Accepted: 07/22/2008] [Indexed: 12/01/2022]
Abstract
SIRT2 inhibitors with a N-(3-phenylpropenoyl)-glycine tryptamide backbone were studied. This backbone has been developed in our group, and it is derived from a compound originally found by virtual screening. In addition, compounds with a smaller 3-phenylpropenoic acid tryptamide backbone were also included in the study. Binding modes for the new compounds and the previously reported compounds were analyzed with molecular modelling methods. The approach, which included a combination of molecular dynamics, molecular docking and cluster analysis, showed that certain docking poses were favourable despite the conformational variation in the target protein. The N-(3-phenylpropenoyl)-glycine tryptamide backbone is also a good backbone for SIRT2 inhibitors, and the series of compounds includes several potent SIRT2 inhibitors.
Collapse
Affiliation(s)
- Päivi H Kiviranta
- Department of Pharmaceutical Chemistry, University of Kuopio, PO Box 1627, 70211 Kuopio, Finland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Huhtiniemi T, Suuronen T, Rinne VM, Wittekindt C, Lahtela-Kakkonen M, Jarho E, Wallén EAA, Salminen A, Poso A, Leppänen J. Oxadiazole-carbonylaminothioureas as SIRT1 and SIRT2 inhibitors. J Med Chem 2008; 51:4377-80. [PMID: 18642893 DOI: 10.1021/jm800639h] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A new inhibitor for human sirtuin type proteins 1 and 2 (SIRT1 and SIRT2) was discovered through virtual database screening in search of new scaffolds. A series of compounds was synthesized based on the hit compound (3-[[3-(4-tert-butylphenyl)1,2,4-oxadiazole-5-carbonyl]amino]-1-[3-(trifluoromethyl)phenyl]thiourea). The most potent compound in the series was nearly as potent as the reference compound (6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide).
Collapse
Affiliation(s)
- Tero Huhtiniemi
- Department of Pharmaceutical Chemistry, University of Kuopio, PO Box 1627, 70211 Kuopio, Finland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
The Sirtuin family: therapeutic targets to treat diseases of aging. Curr Opin Chem Biol 2008; 12:11-7. [DOI: 10.1016/j.cbpa.2008.01.019] [Citation(s) in RCA: 219] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Accepted: 01/15/2008] [Indexed: 12/20/2022]
|
43
|
Gardiner J, Barton D, Marc J, Overall R. Potential Role of Tubulin Acetylation and Microtubule-Based Protein Trafficking in Familial Dysautonomia. Traffic 2007; 8:1145-9. [PMID: 17605759 DOI: 10.1111/j.1600-0854.2007.00605.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Familial dysautonomia (FD), a disease of the autonomic and sensory nervous systems, involves mutations in the protein IkappaB kinase complex-associated protein, which is a component of the human Elongator acetylase complex. We suggest a hypothesis in which defects in tubulin acetylation and impairment of microtubule-based protein trafficking may be an underlying cause of FD. In addition, an Arabidopsis homolog of the Elongator subunit ELP3 has been found to bind to the alphabeta-tubulin heterodimer, suggesting that alpha-tubulin may be a cytoplasmic target of Elongator acetylase activity. Studies of synergistic double mutants in yeast indicate a novel role for Elongator in cytoskeletal dynamics, although this is probably because of an effect on actin rather than microtubules. Finally, we suggest that tubulin deacetylase inhibitors may prove useful in the treatment of FD.
Collapse
Affiliation(s)
- John Gardiner
- School of Biological Sciences, Macleay Building (A12), Science Road, The University of Sydney, Camperdown 2006, Australia.
| | | | | | | |
Collapse
|