1
|
Komal S, Gao Y, Wang ZM, Yu QW, Wang P, Zhang LR, Han SN. Epigenetic Regulation in Myocardial Fibroblasts and Its Impact on Cardiovascular Diseases. Pharmaceuticals (Basel) 2024; 17:1353. [PMID: 39458994 PMCID: PMC11510975 DOI: 10.3390/ph17101353] [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: 08/26/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Myocardial fibroblasts play a crucial role in heart structure and function. In recent years, significant progress has been made in understanding the epigenetic regulation of myocardial fibroblasts, which is essential for cardiac development, homeostasis, and disease progression. In healthy hearts, cardiac fibroblasts (CFs) play a crucial role in synthesizing the extracellular matrix (ECM) when in a dormant state. However, under pathological and environmental stress, CFs transform into activated fibroblasts known as myofibroblasts. These myofibroblasts produce an excess of ECM, which promotes cardiac fibrosis. Although multiple molecular mechanisms are associated with CF activation and myocardial dysfunction, emerging evidence highlights the significant involvement of epigenetic regulation in this process. Epigenetics refers to the heritable changes in gene expression that occur without altering the DNA sequence. These mechanisms have emerged as key regulators of myocardial fibroblast function. This review focuses on recent advancements in the understanding of the role of epigenetic regulation and emphasizes the impact of epigenetic modifications on CF activation. Furthermore, we present perspectives and prospects for future research on epigenetic modifications and their implications for myocardial fibroblasts.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sheng-Na Han
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (S.K.); (Y.G.); (Z.-M.W.); (Q.-W.Y.); (P.W.); (L.-R.Z.)
| |
Collapse
|
2
|
Fioravanti R, Proia E, Tyurenkov IN, Kurkin DV, Bakulin DA, Kovalev NS, Sheikin DS, Kirillov IA, Nawrozkij MB, Vernigora AA, Brunilina LL, Fiorentino F, Mladenović M, Rotili D, Ragno R. Pyrimidine thioethers: A novel class of antidepressant agents, endowed with anxiolytic, performance enhancing and nootropic activity. Eur J Med Chem 2022; 245:114902. [DOI: 10.1016/j.ejmech.2022.114902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 10/22/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022]
|
3
|
Ragno R, Minarini A, Proia E, Lorenzo A, Milelli A, Tumiatti V, Fiore M, Fino P, Rutigliano L, Fioravanti R, Tahara T, Pacella E, Greco A, Canettieri G, Di Paolo ML, Agostinelli E. Bovine Serum Amine Oxidase and Polyamine Analogues: Chemical Synthesis and Biological Evaluation Integrated with Molecular Docking and 3-D QSAR Studies. J Chem Inf Model 2022; 62:3910-3927. [PMID: 35948439 DOI: 10.1021/acs.jcim.2c00559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Natural polyamines (PAs) are key players in cellular homeostasis by regulating cell growth and proliferation. Several observations highlight that PAs are also implicated in pathways regulating cell death. Indeed, the PA accumulation cytotoxic effect, maximized with the use of bovine serum amine oxidase (BSAO) enzyme, represents a valuable strategy against tumor progression. In the present study, along with the design, synthesis, and biological evaluation of a series of new spermine (Spm) analogues (1-23), a mixed structure-based (SB) and ligand-based (LB) protocol was applied. Binding modes of BSAO-PA modeled complexes led to clarify electrostatic and steric features likely affecting the BSAO-PA biochemical kinetics. LB and SB three-dimensional quantitative structure-activity relationship (Py-CoMFA and Py-ComBinE) models were developed by means of the 3d-qsar.com portal, and their analysis represents a strong basis for future design and synthesis of PA BSAO substrates for potential application in oxidative stress-induced chemotherapy.
Collapse
Affiliation(s)
- Rino Ragno
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Sapienza Università di Roma, P. le A. Moro 5, Roma 00185, Italy
| | - Anna Minarini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Eleonora Proia
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Sapienza Università di Roma, P. le A. Moro 5, Roma 00185, Italy
| | - Antonini Lorenzo
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Sapienza Università di Roma, P. le A. Moro 5, Roma 00185, Italy
| | - Andrea Milelli
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto, 237, Rimini 47921, Italy
| | - Vincenzo Tumiatti
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto, 237, Rimini 47921, Italy
| | - Marco Fiore
- Department Institute of Biochemistry and Cell Biology, IBBC-CNR, Via E. Ramarini, 32, Monterotondo Scalo Rome 00015, Italy
| | - Pasquale Fino
- UOC of Dermatology, Policlinico Umberto I Hospital, Sapienza Medical School of Rome, Viale del Policlinico 155, Rome I-00161, Italy
| | - Lavinia Rutigliano
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico155, Rome I-00161, Italy
| | - Rossella Fioravanti
- Department of Drug Chemistry and Technology, Sapienza Università di Roma, P. le A. Moro 5, Roma 00185, Italy
| | - Tomoaki Tahara
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico155, Rome I-00161, Italy
| | - Elena Pacella
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico155, Rome I-00161, Italy
| | - Antonio Greco
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico155, Rome I-00161, Italy
| | - Gianluca Canettieri
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, Rome 00161, Italy.,Istituto Pasteur, Fondazione Cenci-Bolognetti, Sapienza University of Rome, Viale Regina Elena 291, Rome 00161, Italy
| | - Maria Luisa Di Paolo
- Department of Molecular Medicine, University Padua, Via G. Colombo 3, Padova 35131, Italy
| | - Enzo Agostinelli
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico155, Rome I-00161, Italy.,International Polyamines Foundation 'ETS-ONLUS', Via del Forte Tiburtino 98, Rome I-00159, Italy
| |
Collapse
|
4
|
Fiorentino F, Castiello C, Mai A, Rotili D. Therapeutic Potential and Activity Modulation of the Protein Lysine Deacylase Sirtuin 5. J Med Chem 2022; 65:9580-9606. [PMID: 35802779 PMCID: PMC9340778 DOI: 10.1021/acs.jmedchem.2c00687] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sirtiun 5 (SIRT5) is a NAD+-dependent protein lysine deacylase primarily located in mitochondria. SIRT5 displays an affinity for negatively charged acyl groups and mainly catalyzes lysine deglutarylation, desuccinylation, and demalonylation while possessing weak deacetylase activity. SIRT5 substrates play crucial roles in metabolism and reactive oxygen species (ROS) detoxification, and SIRT5 activity is protective in neuronal and cardiac physiology. Moreover, SIRT5 exhibits a dichotomous role in cancer, acting as context-dependent tumor promoter or suppressor. Given its multifaceted activity, SIRT5 is a promising target in the design of activators or inhibitors that might act as therapeutics in many pathologies, including cancer, cardiovascular disorders, and neurodegeneration. To date, few cellular-active peptide-based SIRT5 inhibitors (SIRT5i) have been described, and potent and selective small-molecule SIRT5i have yet to be discovered. In this perspective, we provide an outline of SIRT5's roles in different biological settings and describe SIRT5 modulators in terms of their mode of action, pharmacological activity, and structure-activity relationships.
Collapse
Affiliation(s)
- Francesco Fiorentino
- Department
of Drug Chemistry and Technologies, Sapienza
University of Rome, Piazzala Aldo Moro 5, Rome 00185, Italy
| | - Carola Castiello
- Department
of Drug Chemistry and Technologies, Sapienza
University of Rome, Piazzala Aldo Moro 5, Rome 00185, Italy
| | - Antonello Mai
- Department
of Drug Chemistry and Technologies, Sapienza
University of Rome, Piazzala Aldo Moro 5, Rome 00185, Italy
- Pasteur
Institute, Cenci-Bolognetti Foundation, Sapienza University of Rome, Piazzala Aldo Moro 5, Rome 00185, Italy
| | - Dante Rotili
- Department
of Drug Chemistry and Technologies, Sapienza
University of Rome, Piazzala Aldo Moro 5, Rome 00185, Italy
| |
Collapse
|
5
|
Proia E, Ragno A, Antonini L, Sabatino M, Mladenovič M, Capobianco R, Ragno R. Ligand-based and structure-based studies to develop predictive models for SARS-CoV-2 main protease inhibitors through the 3d-qsar.com portal. J Comput Aided Mol Des 2022; 36:483-505. [PMID: 35716228 PMCID: PMC9206107 DOI: 10.1007/s10822-022-00460-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/28/2022] [Indexed: 11/05/2022]
Abstract
The main protease (Mpro) of SARS-Cov-2 is the essential enzyme for maturation of functional proteins implicated in viral replication and transcription. The peculiarity of its specific cleavage site joint with its high degree of conservation among all coronaviruses promote it as an attractive target to develop broad-spectrum inhibitors, with high selectivity and tolerable safety profile. Herein is reported a combination of three-dimensional quantitative structure-activity relationships (3-D QSAR) and comparative molecular binding energy (COMBINE) analysis to build robust and predictive ligand-based and structure-based statistical models, respectively. Models were trained on experimental binding poses of co-crystallized Mpro-inhibitors and validated on available literature data. By means of deep optimization both models' goodness and robustness reached final statistical values of r2/q2 values of 0.97/0.79 and 0.93/0.79 for the 3-D QSAR and COMBINE approaches respectively, and an overall predictiveness values of 0.68 and 0.57 for the SDEPPRED and AAEP metrics after application to a test set of 60 compounds covered by the training set applicability domain. Despite the different nature (ligand-based and structure-based) of the employed methods, their outcome fully converged. Furthermore, joint ligand- and structure-based structure-activity relationships were found in good agreement with nirmatrelvir chemical features properties, a novel oral Mpro-inhibitor that has recently received U.S. FDA emergency use authorization (EUA) for the oral treatment of mild-to-moderate COVID-19 infected patients. The obtained results will guide future rational design and/or virtual screening campaigns with the aim of discovering new potential anti-coronavirus lead candidates, minimizing both time and financial resources. Moreover, as most of calculation were performed through the well-established web portal 3d-qsar.com the results confirm the portal as a useful tool for drug design.
Collapse
Affiliation(s)
- Eleonora Proia
- Department of Drug Chemistry and Technology, Rome Center for Molecular Design, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Alessio Ragno
- Department of Computer, Control and Management Engineering "Antonio Ruberti", Sapienza University of Rome, Rome, Italy
| | - Lorenzo Antonini
- Department of Drug Chemistry and Technology, Rome Center for Molecular Design, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Manuela Sabatino
- Department of Drug Chemistry and Technology, Rome Center for Molecular Design, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Milan Mladenovič
- Department of Chemistry, Faculty of Science, Kragujevac Center for Computational Biochemistry, University of Kragujevac, Radoja Domanovića 12, P.O. Box 60, 34000, Kragujevac, Serbia
| | - Roberto Capobianco
- Department of Computer, Control and Management Engineering "Antonio Ruberti", Sapienza University of Rome, Rome, Italy
- Sony AI, Rome, Italy
| | - Rino Ragno
- Department of Drug Chemistry and Technology, Rome Center for Molecular Design, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
6
|
Alexandrova E, Salvati A, Pecoraro G, Lamberti J, Melone V, Sellitto A, Rizzo F, Giurato G, Tarallo R, Nassa G, Weisz A. Histone Methyltransferase DOT1L as a Promising Epigenetic Target for Treatment of Solid Tumors. Front Genet 2022; 13:864612. [PMID: 35495127 PMCID: PMC9043692 DOI: 10.3389/fgene.2022.864612] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
The histone lysine methyltransferase DOT1L (DOT1-like histone lysine methyltransferase) is responsible for the epigenetic regulation of gene expression through specific methylation of lysine79 residue of histone H3 (H3K79) in actively transcribed genes. Its normal activity is crucial for embryonic development and adult tissues functions, whereas its aberrant functioning is known to contribute to leukemogenesis. DOT1L is the only lysine methyltransferase that does not contain a SET domain, which is a feature that allowed the development of selective DOT1L inhibitors that are currently investigated in Phase I clinical trials for cancer treatment. Recently, abnormal expression of this enzyme has been associated with poor survival and increased aggressiveness of several solid tumors. In this review evidences of aberrant DOT1L expression and activity in breast, ovarian, prostate, colon, and other solid tumors, and its relationships with biological and clinical behavior of the disease and response to therapies, are summarized. Current knowledge of the structural basis of DOT1L ability to regulate cell proliferation, invasion, plasticity and stemness, cell cycle progression, cell-to-cell signaling, epithelial-to-mesenchymal transition, and chemoresistance, through cooperation with several molecular partners including noncoding RNAs, is also reviewed. Finally, available options for the treatment of therapeutically challenging solid tumors by targeting DOT1L are discussed.
Collapse
Affiliation(s)
- Elena Alexandrova
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Annamaria Salvati
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Medical Genomics Program and Division of Oncology, AOU “S. Giovanni di Dio e Ruggi d’Aragona”, University of Salerno, Salerno, Italy
| | - Giovanni Pecoraro
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Jessica Lamberti
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Viola Melone
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Assunta Sellitto
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Genome Research Center for Health—CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Genome Research Center for Health—CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
| | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Genome Research Center for Health—CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Genome Research Center for Health—CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
- *Correspondence: Giovanni Nassa, ; Alessandro Weisz,
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Medical Genomics Program and Division of Oncology, AOU “S. Giovanni di Dio e Ruggi d’Aragona”, University of Salerno, Salerno, Italy
- Genome Research Center for Health—CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
- *Correspondence: Giovanni Nassa, ; Alessandro Weisz,
| |
Collapse
|
7
|
Yi Y, Ge S. Targeting the histone H3 lysine 79 methyltransferase DOT1L in MLL-rearranged leukemias. J Hematol Oncol 2022; 15:35. [PMID: 35331314 PMCID: PMC8944089 DOI: 10.1186/s13045-022-01251-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/09/2022] [Indexed: 01/28/2023] Open
Abstract
Disrupting the methylation of telomeric silencing 1-like (DOT1L)-mediated histone H3 lysine 79 has been implicated in MLL fusion-mediated leukemogenesis. Recently, DOT1L has become an attractive therapeutic target for MLL-rearranged leukemias. Rigorous studies have been performed, and much progress has been achieved. Moreover, one DOT1L inhibitor, EPZ-5676, has entered clinical trials, but its clinical activity is modest. Here, we review the recent advances and future trends of various therapeutic strategies against DOT1L for MLL-rearranged leukemias, including DOT1L enzymatic activity inhibitors, DOT1L degraders, protein-protein interaction (PPI) inhibitors, and combinatorial interventions. In addition, the limitations, challenges, and prospects of these therapeutic strategies are discussed. In summary, we present a general overview of DOT1L as a target in MLL-rearranged leukemias to provide valuable guidance for DOT1L-associated drug development in the future. Although a variety of DOT1L enzymatic inhibitors have been identified, most of them require further optimization. Recent advances in the development of small molecule degraders, including heterobifunctional degraders and molecular glues, provide valuable insights and references for DOT1L degraders. However, drug R&D strategies and platforms need to be developed and preclinical experiments need to be performed with the purpose of blocking DOT1L-associated PPIs. DOT1L epigenetic-based combination therapy is worth considering and exploring, but the therapy should be based on a thorough understanding of the regulatory mechanism of DOT1L epigenetic modifications.
Collapse
Affiliation(s)
- Yan Yi
- Departments of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Shenglei Ge
- Departments of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Street, Changsha, 410011, Hunan, People's Republic of China.
| |
Collapse
|
8
|
Cao H, Zhou Z, Hu Z, Wei C, Li J, Wang L, Liu G, Zhang J, Wang Y, Wang T, Liang Y. Effect of Enterohepatic Circulation on the Accumulation of Per- and Polyfluoroalkyl Substances: Evidence from Experimental and Computational Studies. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:3214-3224. [PMID: 35138827 DOI: 10.1021/acs.est.1c07176] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The pharmacokinetic characteristics of per- and polyfluoroalkyl substances (PFAS) affect their distribution and bioaccumulation in biological systems. The enterohepatic circulation leads to reabsorption of certain chemicals from bile back into blood and the liver and thus influences their elimination, yet its influence on PFAS bioaccumulation remains unclear. We explored the role of enterohepatic circulation in PFAS bioaccumulation by examining tissue distribution of various PFAS in wild fish and a rat model. Computational models were used to determine the reabsorbed fractions of PFAS by calculating binding affinities of PFAS for key transporter proteins of enterohepatic circulation. The results indicated that higher concentrations were observed in blood, the liver, and bile compared to other tissues for some PFAS in fish. Furthermore, exposure to a PFAS mixture on the rat model showed that the reabsorption phenomenon appeared during 8-12 h for most long-chain PFAS. Molecular docking calculations suggest that PFAS can bind to key transporter proteins via electrostatic and hydrophobic interactions. Further regression analysis adds support to the hypothesis that binding affinity of the apical sodium-dependent bile acid transporter is the most important variable to predict the human half-lives of PFAS. This study demonstrated the critical role of enterohepatic circulation in reabsorption, distribution, and accumulation of PFAS.
Collapse
Affiliation(s)
- Huiming Cao
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Zhen Zhou
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, China
- Key Laboratory of Optoelectronic Chemical Materials and Devices, Ministry of Education, School of Chemical and Environmental Engineering, Jianghan University, Wuhan 430056, China
| | - Zhe Hu
- College of Resources and Environment, Huazhong Agricultural University, Wuhan 430070, China
| | - Cuiyun Wei
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, China
- College of Resources and Environment, Huazhong Agricultural University, Wuhan 430070, China
| | - Jie Li
- College of Resources and Environment, Huazhong Agricultural University, Wuhan 430070, China
| | - Ling Wang
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Guangliang Liu
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Jie Zhang
- College of Resources and Environment, Huazhong Agricultural University, Wuhan 430070, China
| | - Yawei Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Thanh Wang
- MTM Research Centre, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Yong Liang
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| |
Collapse
|
9
|
Si Y, Bon C, Barbachowska M, Cadet-Daniel V, Jallet C, Soresinetti L, Boullé M, Duchateau M, Matondo M, Agou F, Halby L, Arimondo PB. A novel screening strategy to identify histone methyltransferase inhibitors reveals a crosstalk between DOT1L and CARM1. RSC Chem Biol 2022; 3:456-467. [PMID: 35441144 PMCID: PMC8985137 DOI: 10.1039/d1cb00095k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 02/04/2022] [Indexed: 11/21/2022] Open
Abstract
Epigenetic regulation is a dynamic and reversible process that controls gene expression. Abnormal function results in human diseases such as cancer, thus the enzymes that establish epigenetic marks, such as...
Collapse
Affiliation(s)
- Yang Si
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR3523 CNRS 75015 Paris France
| | - Corentin Bon
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR3523 CNRS 75015 Paris France
- Ecole Doctorale MTCI, Université de Paris, Sorbonne Paris Cité Paris France
| | - Magdalena Barbachowska
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR3523 CNRS 75015 Paris France
- Ecole Doctorale MTCI, Université de Paris, Sorbonne Paris Cité Paris France
| | - Veronique Cadet-Daniel
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR3523 CNRS 75015 Paris France
| | - Corinne Jallet
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR3523 CNRS 75015 Paris France
| | - Laura Soresinetti
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR3523 CNRS 75015 Paris France
| | - Mikaël Boullé
- Chemogenomics and Biology Screening platform, Institut Pasteur 75015 Paris France
| | - Magalie Duchateau
- Proteomic Platform, Mass spectrometry for Biology, CNRS USR 2000, Institut Pasteur 75015 Paris France
| | - Mariette Matondo
- Proteomic Platform, Mass spectrometry for Biology, CNRS USR 2000, Institut Pasteur 75015 Paris France
| | - Fabrice Agou
- Chemogenomics and Biology Screening platform, Institut Pasteur 75015 Paris France
| | - Ludovic Halby
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR3523 CNRS 75015 Paris France
| | - Paola B Arimondo
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR3523 CNRS 75015 Paris France
| |
Collapse
|
10
|
Recent progress on cheminformatics approaches to epigenetic drug discovery. Drug Discov Today 2020; 25:2268-2276. [PMID: 33010481 DOI: 10.1016/j.drudis.2020.09.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 08/31/2020] [Accepted: 09/17/2020] [Indexed: 12/16/2022]
Abstract
The ability of epigenetic markers to affect genome function has enabled transformative changes in drug discovery, especially in cancer and other emerging therapeutic areas. Concordant with the introduction of the term 'epi-informatics', the size of the epigenetically relevant chemical space has grown substantially and so did the number of applications of cheminformatic methods to epigenetics. Recent progress in epi-informatics has improved our understanding of the structure-epigenetic activity relationships and boosted the development of models predicting novel epigenetic agents. Herein, we review the advances in computational approaches to drug discovery of small molecules with epigenetic modulation profiles, summarize the current chemogenomics data available for epigenetic targets, and provide a perspective on the greater utility of biomedical knowledge mining as a means to advance the epigenetic drug discovery.
Collapse
|
11
|
Zhu T, Yan H, Singh RP, Wang Y, Cheng H. QSPR study on the polyacrylate-water partition coefficients of hydrophobic organic compounds. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:17550-17560. [PMID: 31493082 DOI: 10.1007/s11356-019-06389-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/30/2019] [Indexed: 06/10/2023]
Abstract
The partition coefficient is essential for the analysis of organic chemicals using solid-phase microextraction (SPME) techniques. In this study, a quantitative structure-property relationship (QSPR) model was developed with chemical descriptors for the prediction of the polyacrylate (PA)-water partition coefficient (KPA-w). The major variables influencing KPA-w in the QSPR model were CrippenlogP (crippen octanal-water partition coefficient), RNCG (relative negative charge-most negative charge/total negative charge), VE2_Dzv (average coefficient sum of the last eigenvector from the Barysz matrix/weighted by van der Waals volume), and ATSC4v (centred Broto-Moreau autocorrelation-lag 4/weighted by van der Waals volume). The relative determination coefficient (R2) and cross-validation coefficient (Q2) were 0.898 and 0.858, respectively, which implied that the model had excellent robustness. Mechanistic interpretation suggested that the factors affecting the partitioning process between PA and water are the hydrophobicity, relative negative charge, and van der Waals volume of a chemical. The results of this study provide a good tool for predicting the log KPA-w values of diverse hydrophobic organic compounds (HOCs) within the applicability domain to reduce experimental costs and the time required for innovation.
Collapse
Affiliation(s)
- Tengyi Zhu
- Jiangsu Provincial Laboratory of Water Environmental Protection Engineering, School of Environmental Science and Engineering, Yangzhou University, Yangzhou, 225127, Jiangsu, China.
| | - Heting Yan
- Jiangsu Provincial Laboratory of Water Environmental Protection Engineering, School of Environmental Science and Engineering, Yangzhou University, Yangzhou, 225127, Jiangsu, China
| | | | - Yajun Wang
- School of Civil Engineering, Southeast University, Nanjing, 210096, China
| | - Haomiao Cheng
- Jiangsu Provincial Laboratory of Water Environmental Protection Engineering, School of Environmental Science and Engineering, Yangzhou University, Yangzhou, 225127, Jiangsu, China.
| |
Collapse
|
12
|
Zhu T, Jiang Y, Cheng H, Singh RP, Yan B. Development of pp-LFER and QSPR models for predicting the diffusion coefficients of hydrophobic organic compounds in LDPE. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 190:110179. [PMID: 31927194 DOI: 10.1016/j.ecoenv.2020.110179] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/31/2019] [Accepted: 01/05/2020] [Indexed: 06/10/2023]
Abstract
Diffusion coefficient (D) is important to evaluate the performance of passive samplers and to monitor the concentration of chemicals effectively. Herein, we developed a polyparameter linear free energy relationship (pp-LFER) model and a quantitative structure-property relationship (QSPR) model for the prediction of diffusion coefficients of hydrophobic organic contaminants (HOCs) in low density polyethylene (LDPE). A dataset of 120 various chemicals was used to develop both models. The pp-LFER model was developed with two descriptors (V and E) and the statistical parameters of the model showed satisfactory results. As a further exploration of the diffusion behavior of the compounds, a QSPR model with five descriptors (ETA_Alpha, ASP-6, IC1, TDB6r and ATSC2v) was constructed with adjusted determination coefficient (R2) of 0.949 and cross-validation coefficient (QLoo2) of 0.941. The regression results indicated that both models had satisfactory goodness-of-fit and robustness. This study proves that pp-LFER and QSPR approaches are available for the prediction of log D values for the hydrophobic organic compounds within the applicability domain.
Collapse
Affiliation(s)
- Tengyi Zhu
- Jiangsu Provincial Laboratory of Water Environmental Protection Engineering, School of Environmental Science and Engineering, Yangzhou University, Yangzhou, 225127, Jiangsu, China
| | - Yue Jiang
- Jiangsu Provincial Laboratory of Water Environmental Protection Engineering, School of Environmental Science and Engineering, Yangzhou University, Yangzhou, 225127, Jiangsu, China
| | - Haomiao Cheng
- Jiangsu Provincial Laboratory of Water Environmental Protection Engineering, School of Environmental Science and Engineering, Yangzhou University, Yangzhou, 225127, Jiangsu, China
| | | | - Bipeng Yan
- Jiangsu Provincial Laboratory of Water Environmental Protection Engineering, School of Environmental Science and Engineering, Yangzhou University, Yangzhou, 225127, Jiangsu, China.
| |
Collapse
|
13
|
Gibbons GS, Chakraborty A, Grigsby SM, Umeano AC, Liao C, Moukha-Chafiq O, Pathak V, Mathew B, Lee YT, Dou Y, Schürer SC, Reynolds RC, Snowden TS, Nikolovska-Coleska Z. Identification of DOT1L inhibitors by structure-based virtual screening adapted from a nucleoside-focused library. Eur J Med Chem 2020; 189:112023. [PMID: 31978781 DOI: 10.1016/j.ejmech.2019.112023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/27/2019] [Accepted: 12/29/2019] [Indexed: 02/07/2023]
Abstract
Disruptor of Telomeric Silencing 1-Like (DOT1L), the sole histone H3 lysine 79 (H3K79) methyltransferase, is required for leukemogenic transformation in a subset of leukemias bearing chromosomal translocations of the Mixed Lineage Leukemia (MLL) gene, as well as other cancers. Thus, DOT1L is an attractive therapeutic target and discovery of small molecule inhibitors remain of high interest. Herein, we are presenting screening results for a unique focused library of 1200 nucleoside analogs originally produced under the aegis of the NIH Pilot Scale Library Program. The complete nucleoside set was screened virtually against DOT1L, resulting in 210 putative hits. In vitro screening of the virtual hits resulted in validation of 11 compounds as DOT1L inhibitors clustered into two distinct chemical classes, adenosine-based inhibitors and a new chemotype that lacks adenosine. Based on the developed DOT1L ligand binding model, a structure-based design strategy was applied and a second-generation of non-nucleoside DOT1L inhibitors was developed. Newly synthesized compound 25 was the most potent DOT1L inhibitor in the new series with an IC50 of 1.0 μM, showing 40-fold improvement in comparison with hit 9 and exhibiting reasonable on target effects in a DOT1L dependent murine cell line. These compounds represent novel chemical probes with a unique non-nucleoside scaffold that bind and compete with the SAM binding site of DOT1L, thus providing foundation for further medicinal chemistry efforts to develop more potent compounds.
Collapse
Affiliation(s)
- Garrett S Gibbons
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA; Molecular and Cellular Pathology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Amarraj Chakraborty
- Department of Chemistry and Biochemistry, The University of Alabama, 250 Hackberry Lane, Tuscaloosa, AL, 35487, USA
| | - Sierrah M Grigsby
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA; Molecular and Cellular Pathology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Afoma C Umeano
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Chenzhong Liao
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Omar Moukha-Chafiq
- Southern Research Institute, Drug Discovery Division, Birmingham, AL, 35205, USA
| | - Vibha Pathak
- Southern Research Institute, Drug Discovery Division, Birmingham, AL, 35205, USA
| | - Bini Mathew
- Southern Research Institute, Drug Discovery Division, Birmingham, AL, 35205, USA
| | - Young-Tae Lee
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Yali Dou
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Stephan C Schürer
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA; Center for Computational Science, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Robert C Reynolds
- Division of Hematology and Oncology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Timothy S Snowden
- Department of Chemistry and Biochemistry, The University of Alabama, 250 Hackberry Lane, Tuscaloosa, AL, 35487, USA.
| | - Zaneta Nikolovska-Coleska
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA; Molecular and Cellular Pathology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA; Rogel Cancer Center at University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
14
|
Stazi G, Taglieri L, Nicolai A, Romanelli A, Fioravanti R, Morrone S, Sabatino M, Ragno R, Taurone S, Nebbioso M, Carletti R, Artico M, Valente S, Scarpa S, Mai A. Dissecting the role of novel EZH2 inhibitors in primary glioblastoma cell cultures: effects on proliferation, epithelial-mesenchymal transition, migration, and on the pro-inflammatory phenotype. Clin Epigenetics 2019; 11:173. [PMID: 31791385 PMCID: PMC6889222 DOI: 10.1186/s13148-019-0763-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most lethal and aggressive malignant primary brain tumor in adults. After surgical resection of the tumor, the patient typically should be subjected to chemotherapy (temozolomide, TMZ) and concomitant radiotherapy. Since the TMZ treatment does not lead to complete remission and often develops resistance, the identification of efficacious therapeutics is strongly to pursue. Among the epigenetic players, the H3K27 methyltransferase (MT) EZH2 (enhancer of zeste homologue 2) has been found overexpressed or mutated in several human cancers including gliomas, and its overexpression is associated with poor outcome in GBM. Two EZH2 inhibitors (EZH2i), UNC1999 and GSK343, suppressed GBM growth in vitro and in vivo indicating that EZH2i can be potential drugs against GBM. RESULTS Two new EZH2i, MC4040 and MC4041, were designed, prepared, and tested by us to determine their effects in primary GBM cell cultures. MC4040 and MC4041 displayed single-digit micromolar inhibition of EZH2, 10-fold less potency against EZH1, and no activity towards other MTs. In primary GBM cells as well as in U-87 GBM cells, the two compounds reduced H3K27me3 levels, and dose- and time-dependently impaired GBM cell viability without inducing apoptosis and arresting the cell cycle in the G0/G1 phase, with increased p21 and p27 levels. In combination with TMZ, MC4040 and MC4041 displayed stronger, but not additive, effects on cell viability. The potent clinical candidate as EZH2i tazemetostat, alone or in combination with TMZ, exhibited a similar potency of inhibition of GBM cell growth when compared to MC4040 and MC4041. At the molecular level, MC4040 and MC4041 reduced the VEGFR1/VEGF expression, reversed the epithelial-mesenchymal transition (EMT), and hampered cell migration and invasion attenuating the cancer malignant phenotype. Treatment of GBM cells with MC4040 and MC4041 also impaired the GBM pro-inflammatory phenotype, with a significant decrease of TGF-β, TNF-α, and IL-6, joined to an increase of the anti-inflammatory cytokine IL-10. CONCLUSIONS The two novel EZH2i MC4040 and MC4041 impaired primary GBM cell viability, showing even stronger effects in combination with TMZ. They also weakened the aggressive malignant phenotype by reducing angiogenesis, EMT, cell migration/invasion and inflammation, thus they may be considered potential candidates against GBM also for combination therapies.
Collapse
Affiliation(s)
- Giulia Stazi
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Ludovica Taglieri
- Department of Experimental Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Alice Nicolai
- Department of Experimental Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Annalisa Romanelli
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Rossella Fioravanti
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Stefania Morrone
- Department of Experimental Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | | | - Rino Ragno
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
- Alchemical Dynamics s.r.l, 00125 Roma, Italy
| | - Samanta Taurone
- Department of Sense Organs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Marcella Nebbioso
- Department of Sense Organs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Raffaella Carletti
- Department of Radiologic, Oncological, and Anatomical and Pathological Sciences, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Marco Artico
- Department of Sense Organs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Sergio Valente
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Susanna Scarpa
- Department of Experimental Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Antonello Mai
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| |
Collapse
|
15
|
Cacabelos R, Carril JC, Sanmartín A, Cacabelos P. Pharmacoepigenetic Processors: Epigenetic Drugs, Drug Resistance, Toxicoepigenetics, and Nutriepigenetics. PHARMACOEPIGENETICS 2019:191-424. [DOI: 10.1016/b978-0-12-813939-4.00006-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|