1
|
Wang L, Xiao J, Zhang B, Hou A. Epigenetic modifications in the development of bronchopulmonary dysplasia: a review. Pediatr Res 2024; 96:632-642. [PMID: 38570557 DOI: 10.1038/s41390-024-03167-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 02/25/2024] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
While perinatal medicine advancements have bolstered survival outcomes for premature infants, bronchopulmonary dysplasia (BPD) continues to threaten their long-term health. Gene-environment interactions, mediated by epigenetic modifications such as DNA methylation, histone modification, and non-coding RNA regulation, take center stage in BPD pathogenesis. Recent discoveries link methylation variations across biological pathways with BPD. Also, the potential reversibility of histone modifications fuels new treatment avenues. The review also highlights the promise of utilizing mesenchymal stem cells and their exosomes as BPD therapies, given their ability to modulate non-coding RNA, opening novel research and intervention possibilities. IMPACT: The complexity and universality of epigenetic modifications in the occurrence and development of bronchopulmonary dysplasia were thoroughly discussed. Both molecular and cellular mechanisms contribute to the diverse nature of epigenetic changes, suggesting the need for deeper biochemical techniques to explore these molecular alterations. The utilization of innovative cell-specific drug delivery methods like exosomes and extracellular vesicles holds promise in achieving precise epigenetic regulation.
Collapse
Affiliation(s)
- Lichuan Wang
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Jun Xiao
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Bohan Zhang
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China
| | - Ana Hou
- Department of Pediatrics, Sheng Jing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Cheng T, Liu C, Wang Y, Li G, Feng L, Zhang S, Qi B, Cui J, Guo L, Cao L, Wang Y, Qi Z, Yang L. A novel histone deacetylase inhibitor Se-SAHA attenuates isoproterenol-induced heart failure via antioxidative stress and autophagy inhibition. Toxicol Appl Pharmacol 2024; 487:116957. [PMID: 38735590 DOI: 10.1016/j.taap.2024.116957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/27/2024] [Accepted: 05/06/2024] [Indexed: 05/14/2024]
Abstract
Heart failure is associated with histone deacetylase (HDAC) regulation of gene expression, the inhibition of which is thought to be beneficial for heart failure therapy. Here, we explored the cardioprotective effects and underlying mechanism of a novel selenium-containing HDAC inhibitor, Se-SAHA, on isoproterenol (ISO)-induced heart failure. We found that pretreatment with Se-SAHA attenuated ISO-induced cardiac hypertrophy and fibrosis in neonatal rat ventricular myocytes (NRVMs). Se-SAHA significantly attenuated the generation of ISO-induced reactive oxygen species (ROS) and restored the expression levels of superoxide dismutase 2 (SOD2) and heme oxygenase-1 (HO-1) in vitro. Furthermore, Se-SAHA pretreatment prevented the accumulation of autophagosomes. Se-SAHA reversed the high expression of HDAC1 and HDAC6 induced by ISO incubation. However, after the addition of the HDAC agonist, the effect of Se-SAHA on blocking autophagy was inhibited. Using ISO-induced mouse models, cardiac ventricular contractile dysfunction, hypertrophy, and fibrosis was reduced treated by Se-SAHA. In addition, Se-SAHA inhibited HDAC1 and HDAC6 overexpression in ISO-treated mice. Se-SAHA treatment significantly increased the activity of SOD2 and improved the ability to eliminate free radicals. Se-SAHA hindered the excessive levels of the microtubule-associated protein 1 light chain 3 (LC3)-II and Beclin-1 in heart failure mice. Collectively, our results indicate that Se-SAHA exerts cardio-protection against ISO-induced heart failure via antioxidative stress and autophagy inhibition.
Collapse
Affiliation(s)
- Tianwei Cheng
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Chang Liu
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yufei Wang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Guangru Li
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Lifeng Feng
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Shengzheng Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Bing Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jianlin Cui
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Lihong Guo
- Institute of Digestive Disease, Shengli Oilfield Central Hospital, Dongying 257000, China
| | - Lei Cao
- Department of General Surgery, Tianjin Union Medical Center, Tianjin 300122, China
| | - Yanming Wang
- College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China.
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China; Institute of Digestive Disease, Shengli Oilfield Central Hospital, Dongying 257000, China; Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300122, China.
| | - Liang Yang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin 300071, China; Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300122, China.
| |
Collapse
|
3
|
Geurs S, Clarisse D, De Bosscher K, D'hooghe M. The Zinc-Binding Group Effect: Lessons from Non-Hydroxamic Acid Vorinostat Analogs. J Med Chem 2023. [PMID: 37276138 DOI: 10.1021/acs.jmedchem.3c00226] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Histone deacetylases (HDACs) are enzymes pursued as drug targets in various cancers and several non-oncological conditions, such as inflammation and neurodegenerative disorders. In the past decade, HDAC inhibitors (HDACi) have emerged as relevant pharmaceuticals, with many efforts devoted to the development of new representatives. However, the growing safety concerns regarding the established hydroxamic acid-based HDAC inhibitors tend to drive current research more toward the design of inhibitors bearing alternative zinc-binding groups (ZBGs). This Perspective presents an overview of all non-hydroxamic acid ZBGs that have been incorporated into the clinically approved prototypical HDACi, suberoylanilide hydroxamic acid (vorinostat). This provides the unique opportunity to compare the inhibition potential and biological effects of different ZBGs in a direct way, as the compounds selected for this Perspective differ only in their ZBG. To that end, different strategies used to select a ZBG, its properties, activity, and liabilities are discussed.
Collapse
Affiliation(s)
- Silke Geurs
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium
- Translational Nuclear Receptor Research, VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, B-9052 Ghent, Belgium
| | - Dorien Clarisse
- Translational Nuclear Receptor Research, VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, B-9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, B-9052 Ghent, Belgium
| | - Karolien De Bosscher
- Translational Nuclear Receptor Research, VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, B-9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Technologiepark-Zwijnaarde 75, B-9052 Ghent, Belgium
| | - Matthias D'hooghe
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium
| |
Collapse
|
4
|
Campiani G, Cavella C, Osko JD, Brindisi M, Relitti N, Brogi S, Saraswati AP, Federico S, Chemi G, Maramai S, Carullo G, Jaeger B, Carleo A, Benedetti R, Sarno F, Lamponi S, Rottoli P, Bargagli E, Bertucci C, Tedesco D, Herp D, Senger J, Ruberti G, Saccoccia F, Saponara S, Gorelli B, Valoti M, Kennedy B, Sundaramurthi H, Butini S, Jung M, Roach KM, Altucci L, Bradding P, Christianson DW, Gemma S, Prasse A. Harnessing the Role of HDAC6 in Idiopathic Pulmonary Fibrosis: Design, Synthesis, Structural Analysis, and Biological Evaluation of Potent Inhibitors. J Med Chem 2021; 64:9960-9988. [PMID: 34251197 PMCID: PMC8300879 DOI: 10.1021/acs.jmedchem.1c00184] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterized by a progressive-fibrosing phenotype. IPF has been associated with aberrant HDAC activities confirmed by our immunohistochemistry studies on HDAC6 overexpression in IPF lung tissues. We herein developed a series of novel hHDAC6 inhibitors, having low inhibitory potency over hHDAC1 and hHDAC8, as potential pharmacological tools for IPF treatment. Their inhibitory potency was combined with low in vitro and in vivo toxicity. Structural analysis of 6h and structure-activity relationship studies contributed to the optimization of the binding mode of the new molecules. The best-performing analogues were tested for their efficacy in inhibiting fibrotic sphere formation and cell viability, proving their capability in reverting the IPF phenotype. The efficacy of analogue 6h was also determined in a validated human lung model of TGF-β1-dependent fibrogenesis. The results highlighted in this manuscript may pave the way for the identification of first-in-class molecules for the treatment of IPF.
Collapse
Affiliation(s)
- Giuseppe Campiani
- University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, 53100 Siena, Italy
| | - Caterina Cavella
- University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, 53100 Siena, Italy
| | - Jeremy D. Osko
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, United States
| | - Margherita Brindisi
- University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, 53100 Siena, Italy
| | - Nicola Relitti
- University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, 53100 Siena, Italy
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126, Pisa, Italy
| | - A. Prasanth Saraswati
- University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, 53100 Siena, Italy
| | - Stefano Federico
- University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, 53100 Siena, Italy
| | - Giulia Chemi
- University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, 53100 Siena, Italy
| | - Samuele Maramai
- University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, 53100 Siena, Italy
| | - Gabriele Carullo
- University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, 53100 Siena, Italy
| | - Benedikt Jaeger
- Klinik für Pneumologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, Hannover, 30625, Germany
| | - Alfonso Carleo
- Klinik für Pneumologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, Hannover, 30625, Germany
| | - Rosaria Benedetti
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Vico L. de Crecchio 7, 80138, Naples, Italy
| | - Federica Sarno
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Vico L. de Crecchio 7, 80138, Naples, Italy
| | - Stefania Lamponi
- University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, 53100 Siena, Italy
| | - Paola Rottoli
- University of Siena, Specialization School of Respiratory Diseases, Department of Medical Sciences, Surgery and Neurosciences, Centro didattico Le Scotte, , 53100, Siena, Italy
| | - Elena Bargagli
- University of Siena, Department of Medical Sciences, Surgery and Neurosciences, Respiratory Diseases Unit, AOUS, Centro didattico Le Scotte, 53100, Siena, Italy
| | - Carlo Bertucci
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro, 6, Bologna 40126, Italy
| | - Daniele Tedesco
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro, 6, Bologna 40126, Italy
| | - Daniel Herp
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Albertstraße 25, 79104, Freiburg, Germany
| | - Johanna Senger
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Albertstraße 25, 79104, Freiburg, Germany
| | - Giovina Ruberti
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), via E. Ramarini 32, 00015 Monterotondo (Rome), Italy
| | - Fulvio Saccoccia
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), via E. Ramarini 32, 00015 Monterotondo (Rome), Italy
| | - Simona Saponara
- Department of Life Sciences, University of Siena, via Aldo Moro 2, I-53100 Siena, Italy
| | - Beatrice Gorelli
- Department of Life Sciences, University of Siena, via Aldo Moro 2, I-53100 Siena, Italy
| | - Massimo Valoti
- Department of Life Sciences, University of Siena, via Aldo Moro 2, I-53100 Siena, Italy
| | - Breándan Kennedy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, D04 V1W8, Dublin, Ireland
| | - Husvinee Sundaramurthi
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, D04 V1W8, Dublin, Ireland
| | - Stefania Butini
- University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, 53100 Siena, Italy
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Albertstraße 25, 79104, Freiburg, Germany
| | - Katy M. Roach
- Department of Respiratory Sciences, University of Leicester, UK, Institute of Lung Health and NIHR Leicester BRC-Respiratory, LE5 4PW, Leicester, UK
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Vico L. de Crecchio 7, 80138, Naples, Italy
| | - Peter Bradding
- Department of Respiratory Sciences, University of Leicester, UK, Institute of Lung Health and NIHR Leicester BRC-Respiratory, LE5 4PW, Leicester, UK
| | - David W. Christianson
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, United States
| | - Sandra Gemma
- University of Siena, Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, via Aldo Moro 2, 53100 Siena, Italy
| | - Antje Prasse
- Klinik für Pneumologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, Hannover, 30625, Germany
| |
Collapse
|
5
|
Luo Y, Li H. Structure-Based Inhibitor Discovery of Class I Histone Deacetylases (HDACs). Int J Mol Sci 2020; 21:E8828. [PMID: 33266366 PMCID: PMC7700698 DOI: 10.3390/ijms21228828] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 12/17/2022] Open
Abstract
Class I histone deacetylases (HDACs) are promising targets for epigenetic therapies for a range of diseases such as cancers, inflammations, infections and neurological diseases. Although six HDAC inhibitors are now licensed for clinical treatments, they are all pan-inhibitors with little or no HDAC isoform selectivity, exhibiting undesirable side effects. A major issue with the currently available HDAC inhibitors is that they have limited specificity and target multiple deacetylases. Except for HDAC8, Class I HDACs (1, 2 and 3) are recruited to large multiprotein complexes to function. Therefore, there are rising needs to develop new, hopefully, therapeutically efficacious HDAC inhibitors with isoform or complex selectivity. Here, upon the introduction of the structures of Class I HDACs and their complexes, we provide an up-to-date overview of the structure-based discovery of Class I HDAC inhibitors, including pan-, isoform-selective and complex-specific inhibitors, aiming to provide an insight into the discovery of additional HDAC inhibitors with greater selectivity, specificity and therapeutic utility.
Collapse
Affiliation(s)
- Yuxiang Luo
- School of Pharmaceutical Sciences, Sun Yat-sen University, No.132 Wai Huan Dong lu, Guangzhou Higher Education Mega Center, Guangzhou 510006, Guangdong, China;
| | - Huilin Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, No.132 Wai Huan Dong lu, Guangzhou Higher Education Mega Center, Guangzhou 510006, Guangdong, China;
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, China
| |
Collapse
|
6
|
Liu S, Hu Z, Zhang Q, Zhu Q, Chen Y, Lu W. Co-Prodrugs of 7-Ethyl-10-hydroxycamptothecin and Vorinostat with in Vitro Hydrolysis and Anticancer Effects. ACS OMEGA 2020; 5:350-357. [PMID: 31956782 PMCID: PMC6964270 DOI: 10.1021/acsomega.9b02786] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/10/2019] [Indexed: 05/17/2023]
Abstract
7-Ethyl-10-hydroxycamptothecin (SN38) and vorinostat (SAHA) are quite promising combination therapy agents applied to the clinical treatment of cancer. In this study, we designed and synthesized a series of novel SN38-SAHA co-prodrugs, which were conjugated by four different amino acids including glycine, alanine, aminobutyric acid, and 6-aminocaproic acid. The hydrolytic reconversion rate to SN38 and SAHA critically depended on the carbon chain length, which were evaluated in PBS (pH 6.0/7.4) and plasma (human/mouse). With decreasing amino acid chain length, the hydrolytic reconversion rate increased gradually. The in vitro cytotoxicity test was evaluated by the sulforhodamine B (SRB) assay on the human lung adenocarcinoma cell line A549 and human colorectal cancer cell line HCT116. With the evaluation of stability and in vitro cytotoxicity, an appropriate linker was found, and the active drug can be released efficiently from compound 3a, which exhibited strong antiproliferative activity in A549 and HCT-116 cell lines correspondingly. These results indicated that the well-designed co-prodrug 3a and this kind of strategy can be a promising approach for anticancer therapy.
Collapse
Affiliation(s)
- Shuangxi Liu
- Shanghai
Engineering Research Center of Molecular Therapeutics and New Drug
Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
| | - Zonglong Hu
- Division
of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy
of Sciences, Shanghai 201203, P. R. China
- University
of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China
| | - Qiumeng Zhang
- Shanghai
Engineering Research Center of Molecular Therapeutics and New Drug
Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
- Division
of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy
of Sciences, Shanghai 201203, P. R. China
- E-mail: . Tel: +86-21- 62238771 (Q.Z.)
| | - Qiwen Zhu
- Shanghai
Engineering Research Center of Molecular Therapeutics and New Drug
Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
| | - Yi Chen
- Division
of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy
of Sciences, Shanghai 201203, P. R. China
- E-mail: . Tel: +86-21- 50801552 (Y.C.)
| | - Wei Lu
- Shanghai
Engineering Research Center of Molecular Therapeutics and New Drug
Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
- E-mail: . Tel: +86-21- 62238771 (W.L.)
| |
Collapse
|
7
|
Liu H, Zhang F, Wang K, Tang X, Wu R. Conformational dynamics and allosteric effect modulated by the unique zinc-binding motif in class IIa HDACs. Phys Chem Chem Phys 2019; 21:12173-12183. [PMID: 31144693 DOI: 10.1039/c9cp02261a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Class IIa histone deacetylases (HDACs) have been considered as potential targets for the treatment of several diseases. Compared to other HDACs, class IIa HDACs have an additional second zinc binding motif. So far, the function of the unique zinc-binding motif is still not very clear. In this work, extensive classical molecular dynamics (MD) simulations were employed to illuminate the conformational change modulated by the unique zinc-binding motif. It has been revealed that the unique zinc-binding motif is a crucial structural stabilization factor in retaining the catalytic activity of the enzyme and the stability of the multi-protein complex, by remotely modulating the active site pocket in a "closed" conformation. Moreover, it is also revealed that the Loop2 motion in HDAC4 is less flexible than that in HDAC7, which opens a new avenue to design selective inhibitors by utilizing the local conformational dynamics difference between the structurally highly similar HDAC4 and HDAC7. Finally, by comparative studies with class I HDACs (HDAC1-3), it is found that the reversible "in-out" conformational transformation of the binding rail (highly conserved both in class I and IIa HDACs) occurs spontaneously in HDAC1-3, whereas the binding rail is trapped in an "in" conformation owing to the strong metal coordination interaction of the unique CCHC zinc-binding motif in class IIa HDACs. Thus, the CCHC zinc-binding motif may be a feasible allosteric site for the development of class IIa-selective inhibitors.
Collapse
Affiliation(s)
- Huawei Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China.
| | - Fan Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China.
| | - Kai Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China.
| | - Xiaowen Tang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China.
| | - Ruibo Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China.
| |
Collapse
|
8
|
Liao W, Sun J, Liu W, Li W, Jia J, Ou F, Su K, Zheng Y, Zhang Z, Sun Y. HDAC10 upregulation contributes to interleukin 1β‐mediated inflammatory activation of synovium‐derived mesenchymal stem cells in temporomandibular joint. J Cell Physiol 2018; 234:12646-12662. [PMID: 30515817 DOI: 10.1002/jcp.27873] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/15/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Wenting Liao
- Department of Oral and Maxillofacial Surgery Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat‐sen University Guangzhou Guangdong People's Republic of China
| | - Jiadong Sun
- Department of Oral and Maxillofacial Surgery Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat‐sen University Guangzhou Guangdong People's Republic of China
| | - Wenjing Liu
- Department of Prosthodontics Stomatological Hospital, Southern Medical University Guangzhou People's Republic of China
| | - Wenyu Li
- Department of Oncology The First Affiliated Hospital, Sun Yat‐sen University Guangzhou Guangdong People's Republic of China
| | - Jiaxin Jia
- Department of Oral and Maxillofacial Surgery Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat‐sen University Guangzhou Guangdong People's Republic of China
| | - Farong Ou
- Department of Oral and Maxillofacial Surgery Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat‐sen University Guangzhou Guangdong People's Republic of China
| | - Kai Su
- Department of Oral and Maxillofacial Surgery Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat‐sen University Guangzhou Guangdong People's Republic of China
| | - Youhua Zheng
- Department of Oral and Maxillofacial Surgery Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat‐sen University Guangzhou Guangdong People's Republic of China
| | - Zhiguang Zhang
- Department of Oral and Maxillofacial Surgery Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat‐sen University Guangzhou Guangdong People's Republic of China
| | - Yangpeng Sun
- Department of Oral and Maxillofacial Surgery Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat‐sen University Guangzhou Guangdong People's Republic of China
| |
Collapse
|
9
|
Novel spiroindoline HDAC inhibitors: Synthesis, molecular modelling and biological studies. Eur J Med Chem 2018; 157:127-138. [DOI: 10.1016/j.ejmech.2018.07.069] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/27/2018] [Accepted: 07/29/2018] [Indexed: 02/08/2023]
|
10
|
Cheshmedzhieva D, Toshev N, Gerova M, Petrov O, Dudev T. Hydroxamic acid derivatives as histone deacetylase inhibitors: a DFT study of their tautomerism and metal affinities/selectivities. J Mol Model 2018; 24:114. [PMID: 29691666 DOI: 10.1007/s00894-018-3651-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 04/09/2018] [Indexed: 01/28/2023]
Abstract
Hydroxamic acids are regarded as potent inhibitors of histone deacetylases (HDAC), and can therefore be used to reduce malignancy growth and size in affected organisms. Although there is a substantial body of information on the structures, syntheses, and biological activities of HDAC inhibitors, several important questions regarding their physicochemical properties and metal affinities/selectivities remain answered. First, how do the conformation and ionization of the hydroxamic group depend on its chemical composition and the dielectric properties of the medium? Second, how do these factors affect the affinities and selectivities of HDAC inhibitors for essential biogenic metal cations? Third, what is the preferred deprotonation site of the hydroxamic moiety and its mode of binding to the metal cation? The present work addressed these questions by performing density functional calculations combined with polarizable continuum model computations. The geometry, deprotonation pattern, metal-binding mode, and metal affinity/selectivity of SAHA, a typical HDAC inhibitor, were examined, and key factors affecting its ligation properties were elucidated. Sulfur- and selenium-containing analogs of SAHA were also modeled for the first time, and their potential as efficient metal-binding entities (to Mg2+, Fe2+, and Zn2+ cations) was assessed. The present calculations shed light on the thermodynamics of the binding of HDAC inhibitors to metal ions, and suggest techniques for enhancing their metal-ligating properties.
Collapse
Affiliation(s)
- D Cheshmedzhieva
- Faculty of Chemistry and Pharmacy, Sofia University "St. Kliment Ohridski", 1164, Sofia, Bulgaria
| | - N Toshev
- Faculty of Chemistry and Pharmacy, Sofia University "St. Kliment Ohridski", 1164, Sofia, Bulgaria
| | - M Gerova
- Faculty of Chemistry and Pharmacy, Sofia University "St. Kliment Ohridski", 1164, Sofia, Bulgaria
| | - O Petrov
- Faculty of Chemistry and Pharmacy, Sofia University "St. Kliment Ohridski", 1164, Sofia, Bulgaria
| | - T Dudev
- Faculty of Chemistry and Pharmacy, Sofia University "St. Kliment Ohridski", 1164, Sofia, Bulgaria.
| |
Collapse
|
11
|
Zhang Q, Li Y, Zhang B, Lu B, Li J. Design, synthesis and biological evaluation of novel histone deacetylase inhibitors incorporating 4-aminoquinazolinyl systems as capping groups. Bioorg Med Chem Lett 2017; 27:4885-4888. [PMID: 28947154 DOI: 10.1016/j.bmcl.2017.09.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 09/13/2017] [Accepted: 09/15/2017] [Indexed: 12/24/2022]
Abstract
A series of hydroxamic acid-based HDACIs with 4-aminoquinazolinyl moieties as capping groups was profiled. Most compounds showed more potent HDACs inhibition activity than clinically used drug SAHA. Among them, compounds 5f and 5h selectively inhibited HDAC 1,2 over HDAC8, and showed strong activity in several cellular assays, not possessing significant toxicity to primary human cells and hERG inhibition. Strikingly, 5f possessed acceptable pharmacokinetic characteristics and exhibited significant antitumor activity in an A549 xenograft model study at well tolerated doses.
Collapse
Affiliation(s)
- Qingwei Zhang
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 201203, PR China.
| | - Yang Li
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 201203, PR China
| | - Baoyin Zhang
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 201203, PR China
| | - Bingliu Lu
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 201203, PR China
| | - Jianqi Li
- Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai 201203, PR China.
| |
Collapse
|
12
|
Zhou J, Huang Y, Cheng C, Wang K, Wu R. Intrinsic Dynamics of the Binding Rail and Its Allosteric Effect in the Class I Histone Deacetylases. J Chem Inf Model 2017; 57:2309-2320. [PMID: 28805377 DOI: 10.1021/acs.jcim.7b00251] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The development of novel isoform/class-selective inhibitors is still of great biological and medical significance to conquer the continuously reported side effects for the histone deacetylase (HDAC) drugs. The first potent HDAC allosteric inhibitor was discovered last year, and this allosteric inhibitor design is thought to be a promising strategy to overcome the current challenges in HDAC inhibitor design. However, the detailed allosteric mechanism and its remote regulatory effects on the catalytic/inhibitor activity of HDAC are still unclear. In this work, on the basis of microsecond-time-scale all-atom molecular dynamics (MD) simulations and picosecond-time-scale density functional theory/molecular mechanics MD simulations on HDAC8, we propose that the allostery is achieved by the intrinsic conformational flexibility of the binding rail (constituted by a highly conserved X-D residue dyad), which steers the loop-loop motion and creates the diverse shapes of the allosteric sites in different HDAC isoforms. Additionally, the rotatability of the binding rail is an inherent structural feature that regulates the hydrophobicity of the linker binding channel and thus further affects the HDAC enzyme inhibitory/catalytic activity by utilizing the promiscuity of X-D dyad. Since the plastic X residue is different among class I HDACs, these new findings provide a deeper understanding of the allostery, which is guidable for the design of new allosteric inhibitors toward the allosteric site and structure modifications on the conventional inhibitors binding into the active pocket by exploiting the intrinsic dynamic features of the conserved X-D dyad.
Collapse
Affiliation(s)
- Jingwei Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University , Guangzhou 510006, P. R. China
| | - Yue Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University , Guangzhou 510006, P. R. China
| | - Chunyan Cheng
- School of Pharmaceutical Sciences, Sun Yat-sen University , Guangzhou 510006, P. R. China
| | - Kai Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University , Guangzhou 510006, P. R. China
| | - Ruibo Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University , Guangzhou 510006, P. R. China
| |
Collapse
|