1
|
Janeček M, Kührová P, Mlýnský V, Stadlbauer P, Otyepka M, Bussi G, Šponer J, Banáš P. Computer Folding of Parallel DNA G-Quadruplex: Hitchhiker's Guide to the Conformational Space. J Comput Chem 2025; 46:e27535. [PMID: 39653644 PMCID: PMC11628365 DOI: 10.1002/jcc.27535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 12/12/2024]
Abstract
Guanine quadruplexes (GQs) play crucial roles in various biological processes, and understanding their folding pathways provides insight into their stability, dynamics, and functions. This knowledge aids in designing therapeutic strategies, as GQs are potential targets for anticancer drugs and other therapeutics. Although experimental and theoretical techniques have provided valuable insights into different stages of the GQ folding, the structural complexity of GQs poses significant challenges, and our understanding remains incomplete. This study introduces a novel computational protocol for folding an entire GQ from single-strand conformation to its native state. By combining two complementary enhanced sampling techniques, we were able to model folding pathways, encompassing a diverse range of intermediates. Although our investigation of the GQ free energy surface (FES) is focused solely on the folding of the all-anti parallel GQ topology, this protocol has the potential to be adapted for the folding of systems with more complex folding landscapes.
Collapse
Affiliation(s)
- Michal Janeček
- Department of Physical Chemistry, Faculty of SciencePalacký University OlomoucOlomoucCzech Republic
| | - Petra Kührová
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucOlomoucCzech Republic
- Institute of Biophysics of the Czech Academy of SciencesBrnoCzech Republic
| | - Vojtěch Mlýnský
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucOlomoucCzech Republic
- Institute of Biophysics of the Czech Academy of SciencesBrnoCzech Republic
- IT4InnovationsVŠB—Technical University of OstravaOstravaCzech Republic
| | - Petr Stadlbauer
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucOlomoucCzech Republic
| | - Michal Otyepka
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucOlomoucCzech Republic
- IT4InnovationsVŠB—Technical University of OstravaOstravaCzech Republic
| | - Giovanni Bussi
- Scuola Internazionale Superiore di Studi Avanzati, SISSATriesteItaly
| | - Jiří Šponer
- Institute of Biophysics of the Czech Academy of SciencesBrnoCzech Republic
| | - Pavel Banáš
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN)Palacký University OlomoucOlomoucCzech Republic
- Institute of Biophysics of the Czech Academy of SciencesBrnoCzech Republic
- IT4InnovationsVŠB—Technical University of OstravaOstravaCzech Republic
| |
Collapse
|
2
|
Choudhir G, Kumar S, Kumar A. Targeting telomeric RNA quadruplexes with natural metabolites to prevent cancer. In Silico Pharmacol 2024; 12:112. [PMID: 39611109 PMCID: PMC11599832 DOI: 10.1007/s40203-024-00283-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 11/01/2024] [Indexed: 11/30/2024] Open
Abstract
Cancer is a major global health burden, causing significant economic losses and premature deaths worldwide. Maintenance of telomeric repeats by telomerase makes the cancer cells immortal. Non-nucleoside mushroom metabolites were screened for their ability to stabilize RG4 structures, making telomeres inaccessible to telomerase and inducing telomere shortening in cancer cells. Selected mushroom metabolites, namely, Sterenin M, Melleolide K, and Zhankuic Acid A were docked with RG4 using the AutoDock Vina and evaluated for non-covalent interactions. These compounds were found to have strong binding affinity and manifested a set of molecular interactions with RG4. To assess the stability of complexes, state-of-the-art molecular dynamics simulations were carried out using the GROMACS 2018.7 software suite with the AMBER99SB-ILDN force field on 250 nanoseconds. Molecular docking and MD simulations revealed the strong interaction patterns between RG4 and the selected metabolites at the atomic level followed by binding free energy calculations. The results suggest that all three metabolites have the potential to be developed into therapeutic agents for cancer treatment. Further in vitro and in vivo studies are needed to assess these compounds' toxicity, efficacy, and dosage.
Collapse
Affiliation(s)
- Gourav Choudhir
- Department of Botany, Chaudhary Charan Singh University, Meerut, 250004 India
| | - Sushil Kumar
- Department of Botany, Shaheed Mangal Pandey Govt. Girls PG College, Madhavpuram, Meerut, 250002 India
| | - Anuj Kumar
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada
| |
Collapse
|
3
|
Rocca R, Ascrizzi S, Citriniti EL, Scionti F, Juli G, Di Martino MT, Caracciolo D, Artese A, Tagliaferri P, Tassone P, Grillone K, Alcaro S. TERRA G-quadruplex stabilization behind the anti-multiple myeloma activity: Novel insights about resveratrol pleiotropic effects. Arch Pharm (Weinheim) 2024; 357:e2400269. [PMID: 39365272 DOI: 10.1002/ardp.202400269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 10/05/2024]
Abstract
Resveratrol (RSV) is a nutraceutical compound belonging to the nonflavonoid polyphenol family, whose antioxidants, anti-inflammatory, and antitumoral properties have been widely investigated. The ability of RSV to provide beneficial effects for neurological, cardiovascular, and cancer disorders rekindled the interest to explore the molecular mechanisms behind its pleiotropic effects, which are due to the modulation of coding and noncoding genes involved in many key biological pathways. With a computational approach, including docking studies and thermodynamics calculations followed by 200-ns-long molecular dynamics and a clustering analysis, we hypothesized the stabilizing binding between RSV and G4 structures of telomeric repeat-containing RNA (TERRA), which is a tumor-suppressive long noncoding RNAs (lncRNA) involved in the regulation of telomere maintenance. In vitro studies performed on cellular models of multiple myeloma (MM) strengthened our hypothesis by highlighting that the antiproliferative and apoptotic effect induced by the treatment with RSV is associated with an increase of TERRA transcript and with upregulation of telomeric heterochromatin markers, such as H3K27Me3 and H4K20Me3, and of the hallmark of apoptosis, cleaved-poly(ADP-ribose) polymerase-1. Our results propose innovative insights underlying the multifaceted role of RSV in MM, by pointing out the role of this natural compound in an lncRNA-mediated regulation to counteract cellular immortality.
Collapse
Affiliation(s)
- Roberta Rocca
- Net4Science srl, University Magna Græcia, Catanzaro, Italy
- Associazione CRISEA-Centro di Ricerca e Servizi Avanzati per l'Innovazione Rurale, Località Condoleo di Belcastro, Catanzaro, Italy
- Department of Health Sciences, University Magna Græcia, Catanzaro, Italy
| | - Serena Ascrizzi
- Department of Experimental and Clinical Medicine, University Magna Græcia, Catanzaro, Italy
| | | | - Francesca Scionti
- Department of Medical and Surgery Sciences, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Giada Juli
- Department of Experimental and Clinical Medicine, University Magna Græcia, Catanzaro, Italy
| | | | - Daniele Caracciolo
- Department of Experimental and Clinical Medicine, University Magna Græcia, Catanzaro, Italy
| | - Anna Artese
- Net4Science srl, University Magna Græcia, Catanzaro, Italy
- Department of Health Sciences, University Magna Græcia, Catanzaro, Italy
| | | | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, University Magna Græcia, Catanzaro, Italy
| | - Katia Grillone
- Department of Experimental and Clinical Medicine, University Magna Græcia, Catanzaro, Italy
| | - Stefano Alcaro
- Net4Science srl, University Magna Græcia, Catanzaro, Italy
- Associazione CRISEA-Centro di Ricerca e Servizi Avanzati per l'Innovazione Rurale, Località Condoleo di Belcastro, Catanzaro, Italy
- Department of Health Sciences, University Magna Græcia, Catanzaro, Italy
| |
Collapse
|
4
|
Neidle S. A Phenotypic Approach to the Discovery of Potent G-Quadruplex Targeted Drugs. Molecules 2024; 29:3653. [PMID: 39125057 PMCID: PMC11314571 DOI: 10.3390/molecules29153653] [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: 07/17/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
G-quadruplex (G4) sequences, which can fold into higher-order G4 structures, are abundant in the human genome and are over-represented in the promoter regions of many genes involved in human cancer initiation, progression, and metastasis. They are plausible targets for G4-binding small molecules, which would, in the case of promoter G4s, result in the transcriptional downregulation of these genes. However, structural information is currently available on only a very small number of G4s and their ligand complexes. This limitation, coupled with the currently restricted information on the G4-containing genes involved in most complex human cancers, has led to the development of a phenotypic-led approach to G4 ligand drug discovery. This approach was illustrated by the discovery of several generations of tri- and tetra-substituted naphthalene diimide (ND) ligands that were found to show potent growth inhibition in pancreatic cancer cell lines and are active in in vivo models for this hard-to-treat disease. The cycles of discovery have culminated in a highly potent tetra-substituted ND derivative, QN-302, which is currently being evaluated in a Phase 1 clinical trial. The major genes whose expression has been down-regulated by QN-302 are presented here: all contain G4 propensity and have been found to be up-regulated in human pancreatic cancer. Some of these genes are also upregulated in other human cancers, supporting the hypothesis that QN-302 is a pan-G4 drug of potential utility beyond pancreatic cancer.
Collapse
Affiliation(s)
- Stephen Neidle
- The School of Pharmacy, University College London, London WC1N 1AX, UK
| |
Collapse
|
5
|
Saw PE, Song E. Advancements in clinical RNA therapeutics: Present developments and prospective outlooks. Cell Rep Med 2024; 5:101555. [PMID: 38744276 PMCID: PMC11148805 DOI: 10.1016/j.xcrm.2024.101555] [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: 01/16/2024] [Revised: 03/05/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024]
Abstract
RNA molecules have emerged as promising clinical therapeutics due to their ability to target "undruggable" proteins or molecules with high precision and minimal side effects. Nevertheless, the primary challenge in RNA therapeutics lies in rapid degradation and clearance from systemic circulation, the inability to traverse cell membranes, and the efficient intracellular delivery of bioactive RNA molecules. In this review, we explore the implications of RNAs in diseases and provide a chronological overview of the development of RNA therapeutics. Additionally, we summarize the technological advances in RNA-screening design, encompassing various RNA databases and design platforms. The paper then presents an update on FDA-approved RNA therapeutics and those currently undergoing clinical trials for various diseases, with a specific emphasis on RNA medicine and RNA vaccines.
Collapse
Affiliation(s)
- Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Nanhai Clinical Translational Center, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Nanhai Clinical Translational Center, Sun Yat-sen Memorial Hospital, Foshan 528200, China; Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
6
|
Bartas M, Brázda V, Pečinka P. Special Issue "Bioinformatics of Unusual DNA and RNA Structures". Int J Mol Sci 2024; 25:5226. [PMID: 38791265 PMCID: PMC11121459 DOI: 10.3390/ijms25105226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Nucleic acids are not only static carriers of genetic information but also play vital roles in controlling cellular lifecycles through their fascinating structural diversity [...].
Collapse
Affiliation(s)
- Martin Bartas
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, 710 00 Ostrava, Czech Republic;
| | - Václav Brázda
- Institute of Biophysics, Czech Academy of Sciences, Královopolská 135, 612 00 Brno, Czech Republic;
| | - Petr Pečinka
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, 710 00 Ostrava, Czech Republic;
| |
Collapse
|
7
|
Rocca R, Grillone K, Citriniti EL, Gualtieri G, Artese A, Tagliaferri P, Tassone P, Alcaro S. Targeting non-coding RNAs: Perspectives and challenges of in-silico approaches. Eur J Med Chem 2023; 261:115850. [PMID: 37839343 DOI: 10.1016/j.ejmech.2023.115850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/08/2023] [Accepted: 09/29/2023] [Indexed: 10/17/2023]
Abstract
The growing information currently available on the central role of non-coding RNAs (ncRNAs) including microRNAs (miRNAS) and long non-coding RNAs (lncRNAs) for chronic and degenerative human diseases makes them attractive therapeutic targets. RNAs carry out different functional roles in human biology and are deeply deregulated in several diseases. So far, different attempts to therapeutically target the 3D RNA structures with small molecules have been reported. In this scenario, the development of computational tools suitable for describing RNA structures and their potential interactions with small molecules is gaining more and more interest. Here, we describe the most suitable strategies to study ncRNAs through computational tools. We focus on methods capable of predicting 2D and 3D ncRNA structures. Furthermore, we describe computational tools to identify, design and optimize small molecule ncRNA binders. This review aims to outline the state of the art and perspectives of computational methods for ncRNAs over the past decade.
Collapse
Affiliation(s)
- Roberta Rocca
- Department of Health Science, Magna Graecia University, Catanzaro, Italy; Net4Science srl, Academic Spinoff, Magna Græcia University, Catanzaro, Italy
| | - Katia Grillone
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | | | | | - Anna Artese
- Department of Health Science, Magna Graecia University, Catanzaro, Italy; Net4Science srl, Academic Spinoff, Magna Græcia University, Catanzaro, Italy.
| | | | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Stefano Alcaro
- Department of Health Science, Magna Graecia University, Catanzaro, Italy; Net4Science srl, Academic Spinoff, Magna Græcia University, Catanzaro, Italy
| |
Collapse
|
8
|
Wang S, Song Y, He Z, Saneyoshi H, Iwakiri R, Xu P, Zhao C, Qu X, Xu Y. Unusual topological RNA G-quadruplex formed by an RNA duplex: implications for the dimerization of SARS-CoV-2 RNA. Chem Commun (Camb) 2023; 59:12703-12706. [PMID: 37819218 DOI: 10.1039/d3cc03192f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
The infectious disease coronavirus 2019 (SARS-CoV-2) is caused by a virus that has RNA as its genetic material. To understand the detailed structural features of SARS-COV-2 RNA, we probed the RNA structure by NMR. Two RNA sequences form a duplex and self-associate to form a dimeric G-quadruplex. The FrG nucleoside was employed as a 19F sensor to confirm the RNA structure in cells by 19F NMR. A FRET assay further demonstrated that the dimeric G-quadruplex resulted in RNA dimerization in cells. These results provide the basis for the elucidation of SARS-COV-2 RNA function, which provides new insights into developing novel antiviral drugs against SARS-COV-2.
Collapse
Affiliation(s)
- Shiyu Wang
- Division of Chemistry, Department of Medical Sciences, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| | - Yi Song
- Division of Chemistry, Department of Medical Sciences, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| | - Zhiyong He
- Division of Chemistry, Department of Medical Sciences, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| | - Hisao Saneyoshi
- Division of Chemistry, Department of Medical Sciences, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| | - Rie Iwakiri
- Division of Chemistry, Department of Medical Sciences, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| | - Pengyu Xu
- Shonan Laboratory, Corporate R&D Headquarters, Otsuka Chemical Co., Ltd., 2Chome26-1, Muraokahigashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Chuanqi Zhao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, Jilin 130022, P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun, Jilin 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Yan Xu
- Division of Chemistry, Department of Medical Sciences, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| |
Collapse
|
9
|
Rocca R, Polerà N, Juli G, Grillone K, Maruca A, Di Martino MT, Artese A, Amato J, Pagano B, Randazzo A, Tagliaferri P, Tassone P, Alcaro S. Hit identification of novel small molecules interfering with MALAT1 triplex by a structure-based virtual screening. Arch Pharm (Weinheim) 2023; 356:e2300134. [PMID: 37309243 DOI: 10.1002/ardp.202300134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 06/14/2023]
Abstract
Nowadays, RNA is an attractive target for the design of new small molecules with different pharmacological activities. Among several RNA molecules, long noncoding RNAs (lncRNAs) are extensively reported to be involved in cancer pathogenesis. In particular, the overexpression of lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) plays an important role in the development of multiple myeloma (MM). Starting from the crystallographic structure of the triple-helical stability element at the 3'-end of MALAT1, we performed a structure-based virtual screening of a large commercial database, previously filtered according to the drug-like properties. After a thermodynamic analysis, we selected five compounds for the in vitro assays. Compound M5, characterized by a diazaindene scaffold, emerged as the most promising molecule enabling the destabilization of the MALAT1 triplex structure and antiproliferative activity on in vitro models of MM. M5 is proposed as a lead compound to be further optimized for improving its affinity toward MALAT1.
Collapse
Affiliation(s)
- Roberta Rocca
- Department of Experimental and Clinical Medicine, Università degli Studi "Magna Graecia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro, Italy
- Net4science srl, Università degli Studi "Magna Graecia" di Catanzaro, Catanzaro, Italy
| | - Nicoletta Polerà
- Department of Experimental and Clinical Medicine, Università degli Studi "Magna Graecia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Giada Juli
- Department of Experimental and Clinical Medicine, Università degli Studi "Magna Graecia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Katia Grillone
- Department of Experimental and Clinical Medicine, Università degli Studi "Magna Graecia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Annalisa Maruca
- Net4science srl, Università degli Studi "Magna Graecia" di Catanzaro, Catanzaro, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Università degli Studi "Magna Graecia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Anna Artese
- Net4science srl, Università degli Studi "Magna Graecia" di Catanzaro, Catanzaro, Italy
- Dipartimento di Scienze della Salute, Università degli Studi "Magna Graecia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Jussara Amato
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Bruno Pagano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Antonio Randazzo
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Pietrosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Università degli Studi "Magna Graecia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Università degli Studi "Magna Graecia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Stefano Alcaro
- Net4science srl, Università degli Studi "Magna Graecia" di Catanzaro, Catanzaro, Italy
- Dipartimento di Scienze della Salute, Università degli Studi "Magna Graecia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro, Italy
| |
Collapse
|
10
|
Acquah FA, Mooers BHM. Targeting RNA Structure to Inhibit Editing in Trypanosomes. Int J Mol Sci 2023; 24:10110. [PMID: 37373258 PMCID: PMC10298474 DOI: 10.3390/ijms241210110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Mitochondrial RNA editing in trypanosomes represents an attractive target for developing safer and more efficient drugs for treating infections with trypanosomes because this RNA editing pathway is not found in humans. Other workers have targeted several enzymes in this editing system, but not the RNA. Here, we target a universal domain of the RNA editing substrate, which is the U-helix formed between the oligo-U tail of the guide RNA and the target mRNA. We selected a part of the U-helix that is rich in G-U wobble base pairs as the target site for the virtual screening of 262,000 compounds. After chemoinformatic filtering of the top 5000 leads, we subjected 50 representative complexes to 50 nanoseconds of molecular dynamics simulations. We identified 15 compounds that retained stable interactions in the deep groove of the U-helix. The microscale thermophoresis binding experiments on these five compounds show low-micromolar to nanomolar binding affinities. The UV melting studies show an increase in the melting temperatures of the U-helix upon binding by each compound. These five compounds can serve as leads for drug development and as research tools to probe the role of the RNA structure in trypanosomal RNA editing.
Collapse
Affiliation(s)
- Francis A. Acquah
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Blaine H. M. Mooers
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Laboratory of Biomolecular Structure and Function, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
11
|
Scionti F, Juli G, Rocca R, Polerà N, Nadai M, Grillone K, Caracciolo D, Riillo C, Altomare E, Ascrizzi S, Caparello B, Cerra M, Arbitrio M, Richter SN, Artese A, Alcaro S, Tagliaferri P, Tassone P, Di Martino MT. TERRA G-quadruplex stabilization as a new therapeutic strategy for multiple myeloma. J Exp Clin Cancer Res 2023; 42:71. [PMID: 36967378 PMCID: PMC10041726 DOI: 10.1186/s13046-023-02633-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 02/28/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a hematologic malignancy characterized by high genomic instability, and telomere dysfunction is an important cause of acquired genomic alterations. Telomeric repeat-containing RNA (TERRA) transcripts are long non-coding RNAs involved in telomere stability through the interaction with shelterin complex. Dysregulation of TERRAs has been reported across several cancer types. We recently identified a small molecule, hit 17, which stabilizes the secondary structure of TERRA. In this study, we investigated in vitro and in vivo anti-MM activities of hit 17. METHODS Anti-proliferative activity of hit 17 was evaluated in different MM cell lines by cell proliferation assay, and the apoptotic process was analyzed by flow cytometry. Gene and protein expressions were detected by RT-qPCR and western blotting, respectively. Microarray analysis was used to analyze the transcriptome profile. The effect of hit 17 on telomeric structure was evaluated by chromatin immunoprecipitation. Further evaluation in vivo was proceeded upon NCI-H929 and AMO-1 xenograft models. RESULTS TERRA G4 stabilization induced in vitro dissociation of telomeric repeat-binding factor 2 (TRF2) from telomeres leading to the activation of ATM-dependent DNA damage response, cell cycle arrest, proliferation block, and apoptotic death in MM cell lines. In addition, up-regulation of TERRA transcription was observed upon DNA damage and TRF2 loss. Transcriptome analysis followed by gene set enrichment analysis (GSEA) confirmed the involvement of the above-mentioned processes and other pathways such as E2F, MYC, oxidative phosphorylation, and DNA repair genes as early events following hit 17-induced TERRA stabilization. Moreover, hit 17 exerted anti-tumor activity against MM xenograft models. CONCLUSION Our findings provide evidence that targeting TERRA by hit 17 could represent a promising strategy for a novel therapeutic approach to MM.
Collapse
Affiliation(s)
- Francesca Scionti
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Giada Juli
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Roberta Rocca
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Net4science Srl, Università degli Studi "Magna Graecia" di Catanzaro, Catanzaro, Italy
| | - Nicoletta Polerà
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Matteo Nadai
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121, Padua, Italy
| | - Katia Grillone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Caterina Riillo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Emanuela Altomare
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Serena Ascrizzi
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Basilio Caparello
- Presidio Ospedaliero "Giovanni Paolo II", Lamezia Terme, Catanzaro, Italy
| | - Maria Cerra
- Presidio Ospedaliero "Giovanni Paolo II", Lamezia Terme, Catanzaro, Italy
| | - Mariamena Arbitrio
- Institute of Research and Biomedical Innovation (IRIB), Italian National Council (CNR), 88100, Catanzaro, Italy
| | - Sara N Richter
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121, Padua, Italy
| | - Anna Artese
- Net4science Srl, Università degli Studi "Magna Graecia" di Catanzaro, Catanzaro, Italy
- Department of Health Sciences, Magna Graecia University of Catanzaro, Campus "Salvatore Venuta", Viale Europa, 88100, Catanzaro, Italy
| | - Stefano Alcaro
- Net4science Srl, Università degli Studi "Magna Graecia" di Catanzaro, Catanzaro, Italy
- Department of Health Sciences, Magna Graecia University of Catanzaro, Campus "Salvatore Venuta", Viale Europa, 88100, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| |
Collapse
|
12
|
Weynand J, Episkopou H, Le Berre G, Gillard M, Dejeu J, Decottignies A, Defrancq E, Elias B. Photo-induced telomeric DNA damage in human cancer cells. RSC Chem Biol 2022; 3:1375-1379. [PMID: 36544575 PMCID: PMC9709782 DOI: 10.1039/d2cb00192f] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/04/2022] [Indexed: 12/05/2022] Open
Abstract
Herein we report on the study of novel dinuclear ruthenium(ii) complexes designed to target and to photo-react with G-quadruplex telomeric DNA. Upon irradiation, complexes efficiently generate guanine radical cation sites as photo-oxidation products. The compounds also display efficient cell penetration with localization to the nucleus and show strong photocytotoxicity toward osteosarcoma cells. Thanks to a microscopic-based telomere dysfunction assay, which allows the direct visualization of DNA damage in cells, we brought the first evidence of forming photo-oxidative damage at telomeres in cellulo. This emphasizes interesting prospects for the development of future cancer phototherapies.
Collapse
Affiliation(s)
- Justin Weynand
- Université catholique de Louvain (UCLouvain), Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST)Place Louis Pasteur 1, bte L4.01.02B-1348 Louvain-la-NeuveBelgium,Université Grenoble-Alpes (UGA), Département de Chimie Moléculaire, UMR CNRS 5250, CS 4070038058 GrenobleFrance
| | - Harikleia Episkopou
- Université catholique de Louvain (UCLouvain), Genetic and Epigenetic Alterations of Genomes, de Duve InstituteAvenue Hippocrate 751200 BrusselsBelgium
| | - Gabriel Le Berre
- Université catholique de Louvain (UCLouvain), Genetic and Epigenetic Alterations of Genomes, de Duve InstituteAvenue Hippocrate 751200 BrusselsBelgium
| | - Martin Gillard
- Université catholique de Louvain (UCLouvain), Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST)Place Louis Pasteur 1, bte L4.01.02B-1348 Louvain-la-NeuveBelgium
| | - Jérôme Dejeu
- Université Grenoble-Alpes (UGA), Département de Chimie Moléculaire, UMR CNRS 5250, CS 4070038058 GrenobleFrance
| | - Anabelle Decottignies
- Université catholique de Louvain (UCLouvain), Genetic and Epigenetic Alterations of Genomes, de Duve InstituteAvenue Hippocrate 751200 BrusselsBelgium
| | - Eric Defrancq
- Université Grenoble-Alpes (UGA), Département de Chimie Moléculaire, UMR CNRS 5250, CS 4070038058 GrenobleFrance
| | - Benjamin Elias
- Université catholique de Louvain (UCLouvain), Institut de la Matière Condensée et des Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST)Place Louis Pasteur 1, bte L4.01.02B-1348 Louvain-la-NeuveBelgium
| |
Collapse
|
13
|
Katsuda Y, Sato SI, Inoue M, Tsugawa H, Kamura T, Kida T, Matsumoto R, Asamitsu S, Shioda N, Shiroto S, Oosawatsu Y, Yatsuzuka K, Kitamura Y, Hagihara M, Ihara T, Uesugi M. Small molecule-based detection of non-canonical RNA G-quadruplex structures that modulate protein translation. Nucleic Acids Res 2022; 50:8143-8153. [PMID: 35801908 PMCID: PMC9371906 DOI: 10.1093/nar/gkac580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/24/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022] Open
Abstract
Tandem repeats of guanine-rich sequences in RNA often form thermodynamically stable four-stranded RNA structures. Such RNA G-quadruplexes have long been considered to be linked to essential biological processes, yet their physiological significance in cells remains unclear. Here, we report a approach that permits the detection of RNA G-quadruplex structures that modulate protein translation in mammalian cells. The approach combines antibody arrays and RGB-1, a small molecule that selectively stabilizes RNA G-quadruplex structures. Analysis of the protein and mRNA products of 84 cancer-related human genes identified Nectin-4 and CapG as G-quadruplex-controlled genes whose mRNAs harbor non-canonical G-quadruplex structures on their 5′UTR region. Further investigations revealed that the RNA G-quadruplex of CapG exhibits a structural polymorphism, suggesting a possible mechanism that ensures the translation repression in a KCl concentration range of 25–100 mM. The approach described in the present study sets the stage for further discoveries of RNA G-quadruplexes.
Collapse
Affiliation(s)
- Yousuke Katsuda
- Division of Materials Science and Chemistry, Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Shin-Ichi Sato
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Maimi Inoue
- Division of Materials Science and Chemistry, Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Hisashi Tsugawa
- Graduate School of Science and Technology, Hirosaki University, 3 Bunkyo-cho, Hirosaki, Aomori 036-8561, Japan
| | - Takuto Kamura
- Division of Materials Science and Chemistry, Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Tomoki Kida
- Division of Materials Science and Chemistry, Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Rio Matsumoto
- Division of Materials Science and Chemistry, Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Sefan Asamitsu
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Norifumi Shioda
- Department of Genomic Neurology, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.,Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe, Chuo-ku, Kumamoto 862-0973, Japan
| | - Shuhei Shiroto
- Graduate School of Science and Technology, Hirosaki University, 3 Bunkyo-cho, Hirosaki, Aomori 036-8561, Japan
| | - Yoshiki Oosawatsu
- Division of Materials Science and Chemistry, Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Kenji Yatsuzuka
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yusuke Kitamura
- Division of Materials Science and Chemistry, Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Masaki Hagihara
- Graduate School of Science and Technology, Hirosaki University, 3 Bunkyo-cho, Hirosaki, Aomori 036-8561, Japan
| | - Toshihiro Ihara
- Division of Materials Science and Chemistry, Faculty of Advanced Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan
| | - Motonari Uesugi
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan.,School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
14
|
Rocca R, Scionti F, Nadai M, Moraca F, Maruca A, Costa G, Catalano R, Juli G, Di Martino MT, Ortuso F, Alcaro S, Tagliaferri P, Tassone P, Richter SN, Artese A. Chromene Derivatives as Selective TERRA G-Quadruplex RNA Binders with Antiproliferative Properties. Pharmaceuticals (Basel) 2022; 15:ph15050548. [PMID: 35631373 PMCID: PMC9147070 DOI: 10.3390/ph15050548] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 11/30/2022] Open
Abstract
In mammalian cells, telomerase transcribes telomeres in large G-rich non-coding RNA, known as telomeric repeat-containing RNA (TERRA), which folds into noncanonical nucleic acid secondary structures called G-quadruplexes (G4s). Since TERRA G4 has been shown to be involved in telomere length and translation regulation, it could provide valuable insight into fundamental biological processes, such as cancer growth, and TERRA G4 binders could represent an innovative strategy for cancer treatment. In this work, the three best candidates identified in our previous virtual screening campaign on bimolecular DNA/RNA G4s were investigated on the monomolecular Tel DNA and TERRA G4s by means of molecular modelling simulations and in vitro and in cell analysis. The results obtained in this work highlighted the stabilizing power of all the three candidates on TERRA G4. In particular, the two compounds characterized by a chromene scaffold were selective TERRA G4 binders, while the compound with a naphthyridine core acted as a dual Tel/TERRA G4-binder. A biophysical investigation by circular dichroism confirmed the relative stabilization efficiency of the compounds towards TERRA and Tel G4s. The TERRA G4 stabilizing hits showed good antiproliferative activity against colorectal and lung adenocarcinoma cell lines. Lead optimization to increase TERRA G4 stabilization may provide new powerful tools against cancer.
Collapse
Affiliation(s)
- Roberta Rocca
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy; (R.R.); (G.J.); (M.T.D.M.); (P.T.); (P.T.)
- Net4science Srl, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (F.M.); (A.M.); (G.C.); (R.C.); (F.O.); (S.A.)
| | - Francesca Scionti
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Matteo Nadai
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy;
| | - Federica Moraca
- Net4science Srl, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (F.M.); (A.M.); (G.C.); (R.C.); (F.O.); (S.A.)
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
| | - Annalisa Maruca
- Net4science Srl, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (F.M.); (A.M.); (G.C.); (R.C.); (F.O.); (S.A.)
- Department of Health Sciences, Magna Graecia University of Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Giosuè Costa
- Net4science Srl, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (F.M.); (A.M.); (G.C.); (R.C.); (F.O.); (S.A.)
- Department of Health Sciences, Magna Graecia University of Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Raffaella Catalano
- Net4science Srl, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (F.M.); (A.M.); (G.C.); (R.C.); (F.O.); (S.A.)
- Department of Health Sciences, Magna Graecia University of Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Giada Juli
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy; (R.R.); (G.J.); (M.T.D.M.); (P.T.); (P.T.)
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy; (R.R.); (G.J.); (M.T.D.M.); (P.T.); (P.T.)
| | - Francesco Ortuso
- Net4science Srl, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (F.M.); (A.M.); (G.C.); (R.C.); (F.O.); (S.A.)
- Department of Health Sciences, Magna Graecia University of Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Stefano Alcaro
- Net4science Srl, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (F.M.); (A.M.); (G.C.); (R.C.); (F.O.); (S.A.)
- Department of Health Sciences, Magna Graecia University of Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy; (R.R.); (G.J.); (M.T.D.M.); (P.T.); (P.T.)
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy; (R.R.); (G.J.); (M.T.D.M.); (P.T.); (P.T.)
| | - Sara N. Richter
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy;
- Correspondence: (S.N.R.); (A.A.)
| | - Anna Artese
- Net4science Srl, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (F.M.); (A.M.); (G.C.); (R.C.); (F.O.); (S.A.)
- Department of Health Sciences, Magna Graecia University of Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
- Correspondence: (S.N.R.); (A.A.)
| |
Collapse
|
15
|
Galindo-Murillo R, Winkler L, Ma J, Hanelli F, Fleming AM, Burrows CJ, Cheatham TE. Riboflavin Stabilizes Abasic, Oxidized G-Quadruplex Structures. Biochemistry 2022; 61:265-275. [PMID: 35104101 PMCID: PMC8851688 DOI: 10.1021/acs.biochem.1c00598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
![]()
The G-quadruplex
is a noncanonical fold of DNA commonly found at
telomeres and within gene promoter regions of the genome. These guanine-rich
sequences are highly susceptible to damages such as base oxidation
and depurination, leading to abasic sites. In the present work, we
address whether a vacancy, such as an abasic site, in a G-quadruplex
serves as a specific ligand recognition site. When the G-tetrad is
all guanines, the vacant (abasic) site is recognized and bound by
free guanine nucleobase. However, we aim to understand whether the
preference for a specific ligand recognition changes with the presence
of a guanine oxidation product 8-oxo-7,8-dihydroguanine (OG) adjacent
to the vacancy in the tetrad. Using molecular dynamics simulation,
circular dichroism, and nuclear magnetic resonance, we examined the
ability for riboflavin to stabilize abasic site-containing G-quadruplex
structures. Through structural and free energy binding analysis, we
observe riboflavin’s ability to stabilize an abasic site-containing
G-quadruplex only in the presence of an adjacent OG-modified base.
Further, when compared to simulation with the vacancy filled by free
guanine, we observe that the free guanine nucleobase is pushed outside
of the tetrad by OG to interact with other parts of the structure,
including loop residues. These results support the preference of riboflavin
over free guanine to fill an OG-adjacent G-quadruplex abasic vacancy.
Collapse
Affiliation(s)
- Rodrigo Galindo-Murillo
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, 2000 East 30 South Skaggs 306, Salt Lake City, Utah 84112, United States
| | - Lauren Winkler
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, 2000 East 30 South Skaggs 306, Salt Lake City, Utah 84112, United States
| | - Jingwei Ma
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-0850, United States
| | - Fatjon Hanelli
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-0850, United States
| | - Aaron M Fleming
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-0850, United States
| | - Cynthia J Burrows
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-0850, United States
| | - Thomas E Cheatham
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, 2000 East 30 South Skaggs 306, Salt Lake City, Utah 84112, United States
| |
Collapse
|
16
|
Targeting of Telomeric Repeat-Containing RNA G-Quadruplexes: From Screening to Biophysical and Biological Characterization of a New Hit Compound. Int J Mol Sci 2021; 22:ijms221910315. [PMID: 34638655 PMCID: PMC8508872 DOI: 10.3390/ijms221910315] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 12/19/2022] Open
Abstract
DNA G-quadruplex (G4) structures, either within gene promoter sequences or at telomeres, have been extensively investigated as potential small-molecule therapeutic targets. However, although G4s forming at the telomeric DNA have been extensively investigated as anticancer targets, few studies focus on the telomeric repeat-containing RNA (TERRA), transcribed from telomeres, as potential pharmacological targets. Here, a virtual screening approach to identify a library of drug-like putative TERRA G4 binders, in tandem with circular dichroism melting assay to study their TERRA G4-stabilizing properties, led to the identification of a new hit compound. The affinity of this compound for TERRA RNA and some DNA G4s was analyzed through several biophysical techniques and its biological activity investigated in terms of antiproliferative effect, DNA damage response (DDR) activation, and TERRA RNA expression in high vs. low TERRA-expressing human cancer cells. The selected hit showed good affinity for TERRA G4 and no binding to double-stranded DNA. In addition, biological assays showed that this compound is endowed with a preferential cytotoxic effect on high TERRA-expressing cells, where it induces a DDR at telomeres, probably by displacing TERRA from telomeres. Our studies demonstrate that the identification of TERRA G4-targeting drugs with potential pharmacological effects is achievable, shedding light on new perspectives aimed at discovering new anticancer agents targeting these G4 structures.
Collapse
|
17
|
Martin WJ, Grandi P, Marcia M. Screening strategies for identifying RNA- and ribonucleoprotein-targeted compounds. Trends Pharmacol Sci 2021; 42:758-771. [PMID: 34215444 DOI: 10.1016/j.tips.2021.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/16/2021] [Accepted: 06/02/2021] [Indexed: 12/23/2022]
Abstract
The past few years have witnessed important breakthroughs in the identification of compounds that specifically bind and regulate RNAs and in optimizing them for therapeutic use. Here, we review successful and unsuccessful approaches in screening for RNA-targeted small molecules. We discuss advantages and disadvantages of the different screening techniques and variables that affect the outcome of RNA-screening projects. We also highlight key challenges that hamper the development of quality RNA ligands, especially the still-low availability of RNA-specific compound libraries and the poor understanding of RNA structural dynamics. We conclude that the development of new RNA-targeting drugs would greatly benefit from integration of the power of high-throughput screening technologies with improved biochemical, structural, and computational characterization of RNA targets.
Collapse
Affiliation(s)
- William J Martin
- Cellzome GmbH, Functional Genomics R&D, GlaxoSmithKline, 69117 Heidelberg, Germany; European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Paola Grandi
- Cellzome GmbH, Functional Genomics R&D, GlaxoSmithKline, 69117 Heidelberg, Germany
| | - Marco Marcia
- European Molecular Biology Laboratory (EMBL) Grenoble, 71 Avenue des Martyrs, Grenoble 38042, France.
| |
Collapse
|
18
|
Banerjee N, Panda S, Chatterjee S. Frontiers in G-Quadruplex Therapeutics in Cancer: Selection of Small Molecules, Peptides and Aptamers. Chem Biol Drug Des 2021; 99:1-31. [PMID: 34148284 DOI: 10.1111/cbdd.13910] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/27/2021] [Accepted: 06/07/2021] [Indexed: 11/28/2022]
Abstract
G-quadruplex, a unique secondary structure in nucleic acids found throughout human genome, elicited widespread interest in the field of therapeutic research. Being present in key regulatory regions of oncogenes, RNAs and telomere, G-quadruplex structure regulates transcription, translation, splicing etc. Changes in its structure and stability leads to differential expression of oncogenes causing cancer. Thus, targeting G-Quadruplex structures with small molecules/other biologics has shown elevated research interest. Covering previous reports, in this review we try to enlighten the facts on the structural diversity in G-quadruplex ligands aiming to provide newer insights to design first-in-class drugs for the next generation cancer treatment.
Collapse
Affiliation(s)
- Nilanjan Banerjee
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata, 700054, India
| | - Suman Panda
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata, 700054, India
| | - Subhrangsu Chatterjee
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata, 700054, India
| |
Collapse
|
19
|
Mulliri S, Laaksonen A, Spanu P, Farris R, Farci M, Mingoia F, Roviello GN, Mocci F. Spectroscopic and In Silico Studies on the Interaction of Substituted Pyrazolo[1,2-a]benzo[1,2,3,4]tetrazine-3-one Derivatives with c-Myc G4-DNA. Int J Mol Sci 2021; 22:6028. [PMID: 34199659 PMCID: PMC8199725 DOI: 10.3390/ijms22116028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/29/2021] [Accepted: 05/30/2021] [Indexed: 12/14/2022] Open
Abstract
Herein we describe a combined experimental and in silico study of the interaction of a series of pyrazolo[1,2-a]benzo[1,2,3,4]tetrazin-3-one derivatives (PBTs) with parallel G-quadruplex (GQ) DNA aimed at correlating their previously reported anticancer activities and the stabilizing effects observed by us on c-myc oncogene promoter GQ structure. Circular dichroism (CD) melting experiments were performed to characterize the effect of the studied PBTs on the GQ thermal stability. CD measurements indicate that two out of the eight compounds under investigation induced a slight stabilizing effect (2-4 °C) on GQ depending on the nature and position of the substituents. Molecular docking results allowed us to verify the modes of interaction of the ligands with the GQ and estimate the binding affinities. The highest binding affinity was observed for ligands with the experimental melting temperatures (Tms). However, both stabilizing and destabilizing ligands showed similar scores, whilst Molecular Dynamics (MD) simulations, performed across a wide range of temperatures on the GQ in water solution, either unliganded or complexed with two model PBT ligands with the opposite effect on the Tms, consistently confirmed their stabilizing or destabilizing ability ascertained by CD. Clues about a relation between the reported anticancer activity of some PBTs and their ability to stabilize the GQ structure of c-myc emerged from our study. Furthermore, Molecular Dynamics simulations at high temperatures are herein proposed for the first time as a means to verify the stabilizing or destabilizing effect of ligands on the GQ, also disclosing predictive potential in GQ-targeting drug discovery.
Collapse
Affiliation(s)
- Simone Mulliri
- Department of Chemical and Geological Sciences, University of Cagliari, I-09042 Monserrato, Italy; (S.M.); (R.F.); (M.F.)
| | - Aatto Laaksonen
- State Key Laboratory of Materials-Oriented and Chemical Engineering, Nanjing Tech University, Nanjing 210009, China
- Division of Physical Chemistry, Department of Materials and Environmental Chemistry, Arrhenius Laboratory, Stockholm University, 10691 Stockholm, Sweden
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, Petru Poni Institute of Macromolecular Chemistry, 700487 Iasi, Romania
- Department of Engineering Sciences and Mathematics, Division of Energy Science, Luleå University of Technology, SE-97187 Luleå, Sweden
| | - Pietro Spanu
- Istituto di Chimica Biomolecolare, ICB-CNR-Trav. La Crucca 3, 07100 Sassari, Italy;
| | - Riccardo Farris
- Department of Chemical and Geological Sciences, University of Cagliari, I-09042 Monserrato, Italy; (S.M.); (R.F.); (M.F.)
| | - Matteo Farci
- Department of Chemical and Geological Sciences, University of Cagliari, I-09042 Monserrato, Italy; (S.M.); (R.F.); (M.F.)
| | - Francesco Mingoia
- Istituto per lo Studio dei Materiali Nanostrutturati ISMN-CNR, Via U. La Malfa 153, I-90146 Palermo, Italy;
| | - Giovanni N. Roviello
- Istituto di Biostrutture e Bioimmagini, IBB-CNR, Via Mezzocannone 16, I-80134 Naples, Italy
| | - Francesca Mocci
- Department of Chemical and Geological Sciences, University of Cagliari, I-09042 Monserrato, Italy; (S.M.); (R.F.); (M.F.)
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, Petru Poni Institute of Macromolecular Chemistry, 700487 Iasi, Romania
| |
Collapse
|
20
|
Alessandrini I, Recagni M, Zaffaroni N, Folini M. On the Road to Fight Cancer: The Potential of G-quadruplex Ligands as Novel Therapeutic Agents. Int J Mol Sci 2021; 22:5947. [PMID: 34073075 PMCID: PMC8198608 DOI: 10.3390/ijms22115947] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 12/14/2022] Open
Abstract
Nucleic acid sequences able to adopt a G-quadruplex conformation are overrepresented within the human genome. This evidence strongly suggests that these genomic regions have been evolutionary selected to play a pivotal role in several aspects of cell biology. In the present review article, we provide an overview on the biological impact of targeting G-quadruplexes in cancer. A variety of small molecules showing good G-quadruplex stabilizing properties has been reported to exert an antitumor activity in several preclinical models of human cancers. Moreover, promiscuous binders and multiple targeting G-quadruplex ligands, cancer cell defense responses and synthetic lethal interactions of G-quadruplex targeting have been also highlighted. Overall, evidence gathered thus far indicates that targeting G-quadruplex may represent an innovative and fascinating therapeutic approach for cancer. The continued methodological improvements, the development of specific tools and a careful consideration of the experimental settings in living systems will be useful to deepen our knowledge of G-quadruplex biology in cancer, to better define their role as therapeutic targets and to help design and develop novel and reliable G-quadruplex-based anticancer strategies.
Collapse
Affiliation(s)
| | | | | | - Marco Folini
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G.A. Amadeo 42, 20133 Milan, Italy; (I.A.); (M.R.); (N.Z.)
| |
Collapse
|
21
|
TERRA G-quadruplex RNA interaction with TRF2 GAR domain is required for telomere integrity. Sci Rep 2021; 11:3509. [PMID: 33568696 PMCID: PMC7876106 DOI: 10.1038/s41598-021-82406-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 01/13/2021] [Indexed: 12/15/2022] Open
Abstract
Telomere dysfunction causes chromosomal instability which is associated with many cancers and age-related diseases. The non-coding telomeric repeat-containing RNA (TERRA) forms a structural and regulatory component of the telomere that is implicated in telomere maintenance and chromosomal end protection. The basic N-terminal Gly/Arg-rich (GAR) domain of telomeric repeat-binding factor 2 (TRF2) can bind TERRA but the structural basis and significance of this interaction remains poorly understood. Here, we show that TRF2 GAR recognizes G-quadruplex features of TERRA. We show that small molecules that disrupt the TERRA-TRF2 GAR complex, such as N-methyl mesoporphyrin IX (NMM) or genetic deletion of TRF2 GAR domain, result in the loss of TERRA, and the induction of γH2AX-associated telomeric DNA damage associated with decreased telomere length, and increased telomere aberrations, including telomere fragility. Taken together, our data indicates that the G-quadruplex structure of TERRA is an important recognition element for TRF2 GAR domain and this interaction between TRF2 GAR and TERRA is essential to maintain telomere stability.
Collapse
|
22
|
Maruca A, Rocca R, Catalano R, Mesiti F, Costa G, Lanzillotta D, Salatino A, Ortuso F, Trapasso F, Alcaro S, Artese A. Natural Products Extracted from Fungal Species as New Potential Anti-Cancer Drugs: A Structure-Based Drug Repurposing Approach Targeting HDAC7. Molecules 2020; 25:E5524. [PMID: 33255661 PMCID: PMC7728054 DOI: 10.3390/molecules25235524] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/16/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023] Open
Abstract
Mushrooms can be considered a valuable source of natural bioactive compounds with potential polypharmacological effects due to their proven antimicrobial, antiviral, antitumor, and antioxidant activities. In order to identify new potential anticancer compounds, an in-house chemical database of molecules extracted from both edible and non-edible fungal species was employed in a virtual screening against the isoform 7 of the Histone deacetylase (HDAC). This target is known to be implicated in different cancer processes, and in particular in both breast and ovarian tumors. In this work, we proposed the ibotenic acid as lead compound for the development of novel HDAC7 inhibitors, due to its antiproliferative activity in human breast cancer cells (MCF-7). These promising results represent the starting point for the discovery and the optimization of new HDAC7 inhibitors and highlight the interesting opportunity to apply the "drug repositioning" paradigm also to natural compounds deriving from mushrooms.
Collapse
Affiliation(s)
- Annalisa Maruca
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (A.M.); (R.C.); (F.M.); (G.C.); (F.O.); (A.A.)
- Net4Science Academic Spin-Off, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy;
| | - Roberta Rocca
- Net4Science Academic Spin-Off, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy;
- Dipartimento di Medicina Sperimentale e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (D.L.); (A.S.); (F.T.)
| | - Raffaella Catalano
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (A.M.); (R.C.); (F.M.); (G.C.); (F.O.); (A.A.)
- Net4Science Academic Spin-Off, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy;
| | - Francesco Mesiti
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (A.M.); (R.C.); (F.M.); (G.C.); (F.O.); (A.A.)
- Net4Science Academic Spin-Off, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy;
| | - Giosuè Costa
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (A.M.); (R.C.); (F.M.); (G.C.); (F.O.); (A.A.)
- Net4Science Academic Spin-Off, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy;
| | - Delia Lanzillotta
- Dipartimento di Medicina Sperimentale e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (D.L.); (A.S.); (F.T.)
| | - Alessandro Salatino
- Dipartimento di Medicina Sperimentale e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (D.L.); (A.S.); (F.T.)
| | - Francesco Ortuso
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (A.M.); (R.C.); (F.M.); (G.C.); (F.O.); (A.A.)
- Net4Science Academic Spin-Off, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy;
| | - Francesco Trapasso
- Dipartimento di Medicina Sperimentale e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (D.L.); (A.S.); (F.T.)
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (A.M.); (R.C.); (F.M.); (G.C.); (F.O.); (A.A.)
- Net4Science Academic Spin-Off, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy;
| | - Anna Artese
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (A.M.); (R.C.); (F.M.); (G.C.); (F.O.); (A.A.)
- Net4Science Academic Spin-Off, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy;
| |
Collapse
|
23
|
In Silico Identification and Biological Evaluation of Antioxidant Food Components Endowed with IX and XII hCA Inhibition. Antioxidants (Basel) 2020; 9:antiox9090775. [PMID: 32825614 PMCID: PMC7555330 DOI: 10.3390/antiox9090775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/30/2022] Open
Abstract
The tumor-associated isoenzymes hCA IX and hCA XII catalyze the hydration of carbon dioxide to bicarbonate and protons. These isoforms are highly overexpressed in many types of cancer, where they contribute to the acidification of the tumor environment, promoting tumor cell invasion and metastasis. In this work, in order to identify novel dual hCA IX and XII inhibitors, virtual screening techniques and biological assays were combined. A structure-based virtual screening towards hCA IX and XII was performed using a database of approximately 26,000 natural compounds. The best shared hits were submitted to a thermodynamic analysis and three promising best hits were identified and evaluated in terms of their hCA IX and XII inhibitor activity. In vitro biological assays were in line with the theoretical studies and revealed that syringin, lithospermic acid, and (-)-dehydrodiconiferyl alcohol behave as good hCA IX and hCA XII dual inhibitors.
Collapse
|
24
|
Grillone K, Riillo C, Scionti F, Rocca R, Tradigo G, Guzzi PH, Alcaro S, Di Martino MT, Tagliaferri P, Tassone P. Non-coding RNAs in cancer: platforms and strategies for investigating the genomic "dark matter". J Exp Clin Cancer Res 2020; 39:117. [PMID: 32563270 PMCID: PMC7305591 DOI: 10.1186/s13046-020-01622-x] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/11/2020] [Indexed: 12/18/2022] Open
Abstract
The discovery of the role of non-coding RNAs (ncRNAs) in the onset and progression of malignancies is a promising frontier of cancer genetics. It is clear that ncRNAs are candidates for therapeutic intervention, since they may act as biomarkers or key regulators of cancer gene network. Recently, profiling and sequencing of ncRNAs disclosed deep deregulation in human cancers mostly due to aberrant mechanisms of ncRNAs biogenesis, such as amplification, deletion, abnormal epigenetic or transcriptional regulation. Although dysregulated ncRNAs may promote hallmarks of cancer as oncogenes or antagonize them as tumor suppressors, the mechanisms behind these events remain to be clarified. The development of new bioinformatic tools as well as novel molecular technologies is a challenging opportunity to disclose the role of the "dark matter" of the genome. In this review, we focus on currently available platforms, computational analyses and experimental strategies to investigate ncRNAs in cancer. We highlight the differences among experimental approaches aimed to dissect miRNAs and lncRNAs, which are the most studied ncRNAs. These two classes indeed need different investigation taking into account their intrinsic characteristics, such as length, structures and also the interacting molecules. Finally, we discuss the relevance of ncRNAs in clinical practice by considering promises and challenges behind the bench to bedside translation.
Collapse
Affiliation(s)
- Katia Grillone
- Laboratory of Translational Medical Oncology, Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, 88100 Catanzaro, Italy
| | - Caterina Riillo
- Laboratory of Translational Medical Oncology, Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, 88100 Catanzaro, Italy
- Medical and Translational Oncology Units, AOU Mater Domini, 88100 Catanzaro, Italy
| | - Francesca Scionti
- Laboratory of Translational Medical Oncology, Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, 88100 Catanzaro, Italy
| | - Roberta Rocca
- Laboratory of Translational Medical Oncology, Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, 88100 Catanzaro, Italy
- Net4science srl, Magna Graecia University, Salvatore Venuta University Campus, 88100 Catanzaro, Italy
| | - Giuseppe Tradigo
- Laboratory of Bioinformatics, Department of Medical and Surgical Sciences, Magna Graecia University, Salvatore Venuta University Campus, 88100 Catanzaro, Italy
| | - Pietro Hiram Guzzi
- Laboratory of Bioinformatics, Department of Medical and Surgical Sciences, Magna Graecia University, Salvatore Venuta University Campus, 88100 Catanzaro, Italy
| | - Stefano Alcaro
- Net4science srl, Magna Graecia University, Salvatore Venuta University Campus, 88100 Catanzaro, Italy
- Department of Health Sciences, Magna Græcia University, Salvatore Venuta University Campus, 88100 Catanzaro, Italy
| | - Maria Teresa Di Martino
- Laboratory of Translational Medical Oncology, Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, 88100 Catanzaro, Italy
- Medical and Translational Oncology Units, AOU Mater Domini, 88100 Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Laboratory of Translational Medical Oncology, Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, 88100 Catanzaro, Italy
- Medical and Translational Oncology Units, AOU Mater Domini, 88100 Catanzaro, Italy
| | - Pierfrancesco Tassone
- Laboratory of Translational Medical Oncology, Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, 88100 Catanzaro, Italy
- Medical and Translational Oncology Units, AOU Mater Domini, 88100 Catanzaro, Italy
| |
Collapse
|
25
|
Maruca A, Lanzillotta D, Rocca R, Lupia A, Costa G, Catalano R, Moraca F, Gaudio E, Ortuso F, Artese A, Trapasso F, Alcaro S. Multi-Targeting Bioactive Compounds Extracted from Essential Oils as Kinase Inhibitors. Molecules 2020; 25:E2174. [PMID: 32384767 PMCID: PMC7249159 DOI: 10.3390/molecules25092174] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 12/12/2022] Open
Abstract
Essential oils (EOs) are popular in aromatherapy, a branch of alternative medicine that claims their curative effects. Moreover, several studies reported EOs as potential anti-cancer agents by inducing apoptosis in different cancer cell models. In this study, we have considered EOs as a potential resource of new kinase inhibitors with a polypharmacological profile. On the other hand, computational methods offer the possibility to predict the theoretical activity profile of ligands, discovering dangerous off-targets and/or synergistic effects due to the potential multi-target action. With this aim, we performed a Structure-Based Virtual Screening (SBVS) against X-ray models of several protein kinases selected from the Protein Data Bank (PDB) by using a chemoinformatics database of EOs. By evaluating theoretical binding affinity, 13 molecules were detected among EOs as new potential kinase inhibitors with a multi-target profile. The two compounds with higher percentages in the EOs were studied more in depth by means Induced Fit Docking (IFD) protocol, in order to better predict their binding modes taking into account also structural changes in the receptor. Finally, given its good binding affinity towards five different kinases, cinnamyl cinnamate was biologically tested on different cell lines with the aim to verify the antiproliferative activity. Thus, this work represents a starting point for the optimization of the most promising EOs structure as kinase inhibitors with multi-target features.
Collapse
Affiliation(s)
- Annalisa Maruca
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (A.M.); (G.C.); (R.C.); (F.O.); (A.A.)
- Net4Science srl, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (R.R.); (A.L.); (F.M.)
| | - Delia Lanzillotta
- Department of Experimental and Clinical Medicine, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (D.L.); (F.T.)
| | - Roberta Rocca
- Net4Science srl, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (R.R.); (A.L.); (F.M.)
- Department of Experimental and Clinical Medicine, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (D.L.); (F.T.)
| | - Antonio Lupia
- Net4Science srl, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (R.R.); (A.L.); (F.M.)
| | - Giosuè Costa
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (A.M.); (G.C.); (R.C.); (F.O.); (A.A.)
- Net4Science srl, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (R.R.); (A.L.); (F.M.)
| | - Raffaella Catalano
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (A.M.); (G.C.); (R.C.); (F.O.); (A.A.)
- Net4Science srl, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (R.R.); (A.L.); (F.M.)
| | - Federica Moraca
- Net4Science srl, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (R.R.); (A.L.); (F.M.)
- Department of Pharmacy, University “Federico II” of Naples, Via D. Montesano 49, 80131 Naples, Italy
| | - Eugenio Gaudio
- Lymphoma and Genomics Research Program, the Institute of Oncology Research, 6500 Bellinzona, Switzerland;
| | - Francesco Ortuso
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (A.M.); (G.C.); (R.C.); (F.O.); (A.A.)
- Net4Science srl, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (R.R.); (A.L.); (F.M.)
| | - Anna Artese
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (A.M.); (G.C.); (R.C.); (F.O.); (A.A.)
- Net4Science srl, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (R.R.); (A.L.); (F.M.)
| | - Francesco Trapasso
- Department of Experimental and Clinical Medicine, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (D.L.); (F.T.)
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (A.M.); (G.C.); (R.C.); (F.O.); (A.A.)
- Net4Science srl, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, Loc. Germaneto, 88100 Catanzaro, Italy; (R.R.); (A.L.); (F.M.)
| |
Collapse
|
26
|
Folding intermediate states of the parallel human telomeric G-quadruplex DNA explored using Well-Tempered Metadynamics. Sci Rep 2020; 10:3176. [PMID: 32081872 PMCID: PMC7035250 DOI: 10.1038/s41598-020-59774-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/30/2020] [Indexed: 11/16/2022] Open
Abstract
An increasingly comprehension of the folding intermediate states of DNA G-quadruplexes (G4s) is currently an important scientific challenge, especially for the human telomeric (h-tel) G4s-forming sequences, characterized by a highly polymorphic nature. Despite the G-triplex conformation was proposed as one of the possible folding intermediates for the antiparallel and hybrid h-tel G4s, for the parallel h-tel topology with an all-anti guanine orientation, a vertical strand-slippage involving the G-triplets was proposed in previous works through microseconds-long standard molecular dynamics simulations (MDs). Here, in order to get further insights into the vertical strand-slippage and the folding intermediate states of the parallel h-tel G4s, we have carried out a Well-Tempered Metadynamics simulation (WT-MetaD), which allowed us to retrieve an ensemble of six G4s having two/G-tetrad conformations derived by the G-triplets vertical slippage. The insights highlighted in this work are aimed at rationalizing the mechanistic characterisation of the parallel h-tel G4 folding process.
Collapse
|
27
|
The Non-Coding RNA Landscape of Plasma Cell Dyscrasias. Cancers (Basel) 2020; 12:cancers12020320. [PMID: 32019064 PMCID: PMC7072200 DOI: 10.3390/cancers12020320] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/14/2022] Open
Abstract
Despite substantial advancements have been done in the understanding of the pathogenesis of plasma cell (PC) disorders, these malignancies remain hard-to-treat. The discovery and subsequent characterization of non-coding transcripts, which include several members with diverse length and mode of action, has unraveled novel mechanisms of gene expression regulation often malfunctioning in cancer. Increasing evidence indicates that such non-coding molecules also feature in the pathobiology of PC dyscrasias, where they are endowed with strong therapeutic and/or prognostic potential. In this review, we aim to summarize the most relevant findings on the biological and clinical features of the non-coding RNA landscape of malignant PCs, with major focus on multiple myeloma. The most relevant classes of non-coding RNAs will be examined, along with the mechanisms accounting for their dysregulation and the recent strategies used for their targeting in PC dyscrasias. It is hoped these insights may lead to clinical applications of non-coding RNA molecules as biomarkers or therapeutic targets/agents in the near future.
Collapse
|
28
|
A drug repurposing screening reveals a novel epigenetic activity of hydroxychloroquine. Eur J Med Chem 2019; 183:111715. [DOI: 10.1016/j.ejmech.2019.111715] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/24/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022]
|
29
|
Catalano R, Moraca F, Amato J, Cristofari C, Rigo R, Via LD, Rocca R, Lupia A, Maruca A, Costa G, Catalanotti B, Artese A, Pagano B, Randazzo A, Sissi C, Novellino E, Alcaro S. Targeting multiple G-quadruplex–forming DNA sequences: Design, biophysical and biological evaluations of indolo-naphthyridine scaffold derivatives. Eur J Med Chem 2019; 182:111627. [DOI: 10.1016/j.ejmech.2019.111627] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/02/2019] [Accepted: 08/14/2019] [Indexed: 11/25/2022]
|
30
|
Estep KN, Butler TJ, Ding J, Brosh RM. G4-Interacting DNA Helicases and Polymerases: Potential Therapeutic Targets. Curr Med Chem 2019; 26:2881-2897. [PMID: 29149833 DOI: 10.2174/0929867324666171116123345] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/16/2017] [Accepted: 10/16/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Guanine-rich DNA can fold into highly stable four-stranded DNA structures called G-quadruplexes (G4). In recent years, the G-quadruplex field has blossomed as new evidence strongly suggests that such alternately folded DNA structures are likely to exist in vivo. G4 DNA presents obstacles for the replication machinery, and both eukaryotic DNA helicases and polymerases have evolved to resolve and copy G4 DNA in vivo. In addition, G4-forming sequences are prevalent in gene promoters, suggesting that G4-resolving helicases act to modulate transcription. METHODS We have searched the PubMed database to compile an up-to-date and comprehensive assessment of the field's current knowledge to provide an overview of the molecular interactions of Gquadruplexes with DNA helicases and polymerases implicated in their resolution. RESULTS Novel computational tools and alternative strategies have emerged to detect G4-forming sequences and assess their biological consequences. Specialized DNA helicases and polymerases catalytically act upon G4-forming sequences to maintain normal replication and genomic stability as well as appropriate gene regulation and cellular homeostasis. G4 helicases also resolve telomeric repeats to maintain chromosomal DNA ends. Bypass of many G4-forming sequences is achieved by the action of translesion DNS polymerases or the PrimPol DNA polymerase. While the collective work has supported a role of G4 in nuclear DNA metabolism, an emerging field centers on G4 abundance in the mitochondrial genome. CONCLUSION Discovery of small molecules that specifically bind and modulate DNA helicases and polymerases or interact with the G4 DNA structure itself may be useful for the development of anticancer regimes.
Collapse
Affiliation(s)
- Katrina N Estep
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, NIH Biomedical Research Center, 251 Bayview Blvd Baltimore, MD 21224, United States
| | - Thomas J Butler
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, NIH Biomedical Research Center, 251 Bayview Blvd Baltimore, MD 21224, United States
| | - Jun Ding
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, NIH Biomedical Research Center, 251 Bayview Blvd Baltimore, MD 21224, United States
| | - Robert M Brosh
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, NIH Biomedical Research Center, 251 Bayview Blvd Baltimore, MD 21224, United States
| |
Collapse
|
31
|
Maruca A, Ambrosio FA, Lupia A, Romeo I, Rocca R, Moraca F, Talarico C, Bagetta D, Catalano R, Costa G, Artese A, Alcaro S. Computer-based techniques for lead identification and optimization I: Basics. PHYSICAL SCIENCES REVIEWS 2019. [DOI: 10.1515/psr-2018-0113] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractThis chapter focuses on computational techniques for identifying and optimizing lead molecules, with a special emphasis on natural compounds. A number of case studies have been specifically discussed, such as the case of the naphthyridine scaffold, discovered through a structure-based virtual screening (SBVS) and proposed as the starting point for further lead optimization process, to enhance its telomeric RNA selectivity. Another example is the case of Liphagal, a tetracyclic meroterpenoid extracted fromAka coralliphaga, known as PI3Kα inhibitor, provide an evidence for the design of new active congeners against PI3Kα using molecular dynamics (MD) simulations. These are only two of the numerous examples of the computational techniques’ powerful in drug design and drug discovery fields. Finally, the design of drugs that can simultaneously interact with multiple targets as a promising approach for treating complicated diseases has been reported. An example of polypharmacological agents are the compounds extracted from mushrooms identified by means of molecular docking experiments. This chapter may be a useful manual of molecular modeling techniques used in the lead-optimization and lead identification processes.
Collapse
|
32
|
Meleddu R, Petrikaite V, Distinto S, Arridu A, Angius R, Serusi L, Škarnulytė L, Endriulaitytė U, Paškevičiu̅tė M, Cottiglia F, Gaspari M, Taverna D, Deplano S, Fois B, Maccioni E. Investigating the Anticancer Activity of Isatin/Dihydropyrazole Hybrids. ACS Med Chem Lett 2019; 10:571-576. [PMID: 30996798 DOI: 10.1021/acsmedchemlett.8b00596] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 12/17/2018] [Indexed: 12/26/2022] Open
Abstract
A series of isatin-dihydropyrazole hybrids have been synthesized in order to assess their potential as anticancer agents. In particular, 12 compounds were evaluated for their antiproliferative activity toward A549, IGR39, U87, MDA-MB-231, MCF-7, BT474, BxPC-3, SKOV-3, and H1299 cell lines, and human foreskin fibroblasts. Four compounds exhibited interesting antiproliferative activity and were further examined to determine their EC50 values toward a panel of selected tumor cell lines. The best compounds were then investigated for their induced mechanism of cell death. Preliminary structure-activity relationship indicates that the presence of a substituent such as a chlorine atom or a methyl moiety in position 5 of the isatin nucleus is beneficial for the antitumor activity. EMAC4001 proved the most promising compound within the studied series with EC50 values ranging from 0.01 to 0.38 μM.
Collapse
Affiliation(s)
- Rita Meleddu
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Vilma Petrikaite
- Department of Drug Chemistry, Faculty of Pharmacy, Lithuanian University of Health Sciences, 50162 Kaunas, Lithuania
- Institute of Biotechnology, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Simona Distinto
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Antonella Arridu
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Rossella Angius
- Laboratorio NMR e Tecnologie Bioanalitiche, Sardegna Ricerche, Pula, 09010 Cagliari, Italy
| | - Lorenzo Serusi
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Laura Škarnulytė
- Department of Drug Chemistry, Faculty of Pharmacy, Lithuanian University of Health Sciences, 50162 Kaunas, Lithuania
| | - Ugnė Endriulaitytė
- Department of Drug Chemistry, Faculty of Pharmacy, Lithuanian University of Health Sciences, 50162 Kaunas, Lithuania
| | - Miglė Paškevičiu̅tė
- Department of Drug Chemistry, Faculty of Pharmacy, Lithuanian University of Health Sciences, 50162 Kaunas, Lithuania
| | - Filippo Cottiglia
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Marco Gaspari
- Department of Experimental and Clinical Medicine, “Magna Græcia” University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Domenico Taverna
- Department of Experimental and Clinical Medicine, “Magna Græcia” University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Serenella Deplano
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Benedetta Fois
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Elias Maccioni
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| |
Collapse
|
33
|
Tikhomirov AS, Tsvetkov VB, Kaluzhny DN, Volodina YL, Zatonsky GV, Schols D, Shchekotikhin AE. Tri-armed ligands of G-quadruplex on heteroarene-fused anthraquinone scaffolds: Design, synthesis and pre-screening of biological properties. Eur J Med Chem 2018; 159:59-73. [DOI: 10.1016/j.ejmech.2018.09.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 01/30/2023]
|
34
|
Monsen RC, Trent JO. G-quadruplex virtual drug screening: A review. Biochimie 2018; 152:134-148. [PMID: 29966734 PMCID: PMC6134840 DOI: 10.1016/j.biochi.2018.06.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 06/28/2018] [Indexed: 12/18/2022]
Abstract
Over the past two decades biologists and bioinformaticians have unearthed substantial evidence supporting a role for G-quadruplexes as important mediators of biological processes. This includes telomere damage signaling, transcriptional activity, and splicing. Both their structural heterogeneity and their abundance in oncogene promoters makes them ideal targets for drug discovery. Currently, there are hundreds of deposited DNA and RNA quadruplex atomic structures which have allowed researchers to begin using in silico drug screening approaches to develop novel stabilizing ligands. Here we provide a review of the past decade of G-quadruplex virtual drug discovery approaches and campaigns. With this we introduce relevant virtual screening platforms followed by a discussion of best practices to assist future G4 VS campaigns.
Collapse
Affiliation(s)
- Robert C Monsen
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY, 40206, USA
| | - John O Trent
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40206, USA; Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY, 40206, USA; Department of Medicine, University of Louisville, Louisville, KY, 40206, USA.
| |
Collapse
|
35
|
Rocca R, Moraca F, Costa G, Talarico C, Ortuso F, Da Ros S, Nicoletto G, Sissi C, Alcaro S, Artese A. In Silico Identification of Piperidinyl-amine Derivatives as Novel Dual Binders of Oncogene c-myc/c-Kit G-quadruplexes. ACS Med Chem Lett 2018; 9:848-853. [PMID: 30128079 DOI: 10.1021/acsmedchemlett.8b00275] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 07/10/2018] [Indexed: 12/15/2022] Open
Abstract
In the last years, it has been shown that the DNA secondary structure known as G-quadruplex is also involved in the regulation of oncogenes transcription, such as c-myc, c-Kit, KRAS, Bcl-2, VEGF, and PDGF. DNA G-quadruplexes, formed in the promoter region of these proto-oncogenes, are considered alternative anticancer targets since their stabilization causes a reduction of the related oncoprotein overexpression. In this study, a structure-based virtual screening toward the experimental DNA G-quadruplex structures of c-myc and c-Kit was performed by using Glide for the docking analysis of a commercial library of approximately 693 000 compounds. The best hits were submitted to thermodynamic and biophysical studies, highlighting the effective stabilization of both G-quadruplex oncogene promoter structures for three N-(4-piperidinylmethyl)amine derivatives, thus proposed as a new class of dual G-quadruplex binders.
Collapse
Affiliation(s)
- Roberta Rocca
- Dipartimento di Scienze della Salute, Università “Magna Graecia” di Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Federica Moraca
- Dipartimento di Scienze della Salute, Università “Magna Graecia” di Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Giosuè Costa
- Dipartimento di Scienze della Salute, Università “Magna Graecia” di Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Carmine Talarico
- Dipartimento di Scienze della Salute, Università “Magna Graecia” di Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Francesco Ortuso
- Dipartimento di Scienze della Salute, Università “Magna Graecia” di Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Silvia Da Ros
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Giulia Nicoletto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Claudia Sissi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università “Magna Graecia” di Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Anna Artese
- Dipartimento di Scienze della Salute, Università “Magna Graecia” di Catanzaro, Campus “Salvatore Venuta”, Viale Europa, 88100 Catanzaro, Italy
| |
Collapse
|
36
|
Myoblast Myogenic Differentiation but Not Fusion Process Is Inhibited via MyoD Tetraplex Interaction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7640272. [PMID: 29854094 PMCID: PMC5964432 DOI: 10.1155/2018/7640272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 03/17/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022]
Abstract
The presence of tetraplex structures in the promoter region of the myogenic differentiation 1 gene (MyoD1) was investigated with a specific tetraplex-binding porphyrin (TMPyP4), to test its influence on the expression of MyoD1 itself and downstream-regulated genes during myogenic differentiation. TMPyP4-exposed C2C12 myoblasts, blocking MyoD1 transcription, proliferated reaching confluence and fused forming elongated structures, resembling myotubes, devoid of myosin heavy chain 3 (MHC) expression. Besides lack of MHC, upon MyoD1 inhibition, other myogenic gene expressions were also affected in treated cells, while untreated control cell culture showed normal myotube formation expressing MyoD1, Myog, MRF4, Myf5, and MHC. Unexpectedly, the myomaker (Mymk) gene expression was not affected upon TMPyP4 exposure during C2C12 myogenic differentiation. At the genomic level, the bioinformatic comparison of putative tetraplex sites found that three tetraplexes in MyoD1 and Myog are highly conserved in mammals, while Mymk and MHC did not show any conserved tetraplexes in the analysed regions. Thus, here, we report for the first time that the inhibition of the MyoD1 promoter function, stabilizing the tetraplex region, affects downstream myogenic genes by blocking their expression, while leaving the expression of Mymk unaltered. These results reveal the existence of two distinct pathways: one leading to cell fusion and one guaranteeing correct myotube differentiation.
Collapse
|
37
|
Giancola C, Montesarchio D. Not unusual, just different! Chemistry, biology and applications of G-quadruplex nucleic acids. Biochim Biophys Acta Gen Subj 2017; 1861:1201-1204. [PMID: 28578868 DOI: 10.1016/j.bbagen.2017.03.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/19/2017] [Accepted: 04/23/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Concetta Giancola
- Department of Pharmacy, University of Naples Federico II, via D. Montesano 49, I-80131 Naples Italy
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, via Cintia 21,I-80126 Naples Italy
| |
Collapse
|