1
|
Bollu VS, Chen YC, Zhang F, Gowda K, Amin S, Sharma AK, Schell TD, Zhu J, Robertson GP. Managing telomerase and telomere dysfunction in acral melanoma. Pharmacol Res 2025; 215:107700. [PMID: 40097124 DOI: 10.1016/j.phrs.2025.107700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Acral Lentiginous Melanoma is a rare and aggressive subtype of melanoma that commonly affects the palms, soles, and nail beds. It is more prevalent in individuals with darker skin tones, including Asian, African, and Hispanic populations. Unlike cutaneous melanomas, acral melanoma is not associated with UV exposure and has a distinct genetic and molecular profile, underscoring the need for tailored research and treatment strategies. Standard treatments, such as surgery, chemotherapy, immunotherapy, and targeted therapies, have shown limited success for this melanoma subtype, highlighting the urgency of developing more effective interventions. Telomerase is an enzyme that extends telomeres and is a key target in acral melanoma which exhibits' high telomerase activity, driven by mutations in the telomerase reverse transcriptase TERT promoter, which contributes to uncontrolled tumor cell proliferation, cancer cell immortality, and resistance to conventional therapies. Therefore, targeting telomerase presents a promising therapeutic avenue for acral melanoma patients who do not respond well to current treatments. Several approaches for targeting telomerase deregulation have been developed, and their potential for the management of acral melanoma is discussed in this review. Specifically, the promise of telomerase-targeted therapies for acral melanoma is emphasized and explores how these strategies could improve outcomes for patients with this challenging skin cancer. By focusing on the role of telomerase in tumorigenesis and treatment resistance, telomerase-targeted strategies hold potential as a foundational component of therapies for acral melanoma, complementing existing approaches.
Collapse
Affiliation(s)
- Vishnu Sravan Bollu
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Yu-Chi Chen
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Fan Zhang
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pharmaceutical Sciences, Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, WA 99202, United States
| | - Krishne Gowda
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Shantu Amin
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Arun K Sharma
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Todd D Schell
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Jiyue Zhu
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pharmaceutical Sciences, Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, WA 99202, United States
| | - Gavin P Robertson
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Dermatology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Surgery, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Foreman Foundation for Melanoma Research, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Melanoma and Skin Cancer Center, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Melanoma Therapeutics Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States.
| |
Collapse
|
2
|
Lin J, Gong Z, Lu Y, Cai J, Zhang J, Tan J, Huang Z, Chen S. Recent Progress and Potential of G4 Ligands in Cancer Immunotherapy. Molecules 2025; 30:1805. [PMID: 40333779 PMCID: PMC12029830 DOI: 10.3390/molecules30081805] [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: 03/24/2025] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
G-quadruplex (G4) structures are non-canonical nucleic acid conformations that play crucial roles in gene regulation, DNA replication, and telomere maintenance. Recent studies have highlighted G4 ligands as promising anticancer agents due to their ability to modulate oncogene expression and induce DNA damage. By stabilizing G4 structures, these ligands affect tumor progression. Additionally, they have been implicated in tumor immunity modulation, particularly through the activation and immunogenic cell death induction of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway. Moreover, their disruption of telomere maintenance and regulation of key oncogenes, such as c-MYC and KRAS, position them as candidates for immune-based therapeutic interventions. Despite their therapeutic potential, challenges remain in optimizing their clinical applications, particularly in patient stratification and elucidating their immunomodulatory effects. This review provides a comprehensive overview of the mechanisms through which G4 ligands influence tumor progression and immune regulation, highlighting their potential role in future cancer immunotherapy strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shuobin Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; (J.L.); (Z.G.); (Y.L.); (J.C.); (J.Z.); (J.T.); (Z.H.)
| |
Collapse
|
3
|
Marques DSC, da Silva Lima L, de Oliveira Moraes Miranda JF, Dos Anjos Santos CÁ, da Cruz Filho IJ, de Lima MDCA. Exploring the therapeutic potential of acridines: Synthesis, structure, and biological applications. Bioorg Chem 2025; 155:108096. [PMID: 39756205 DOI: 10.1016/j.bioorg.2024.108096] [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: 10/28/2024] [Revised: 12/15/2024] [Accepted: 12/22/2024] [Indexed: 01/07/2025]
Abstract
The objective of this review was to explore the trends and chemical characteristics of acridines and their derivatives, analyze their contribution to the scientific literature and international cooperation, identify the most influential authors and articles, and provide an overview of the knowledge produced in elucidating their mechanisms of action. To this end, a bibliometric analysis was performed using RStudio software, along with a systematic review focusing on articles indexed in the "Web of Science" and "Scopus" databases. The keywords used were "acridine$", "Synthesi$", "Structure$", and "Biologic* Application$" for the period from 2020 to 2024. Relevant articles were carefully selected from these databases, and a bibliometric analysis was carried out to comprehensively discuss the most relevant biological activities associated with acridines. The results showed that, during the analyzed period, China and India led in the number of publications, followed by Brazil in third place. However, a decline in the number of publications was observed in the last two years of the period. Keyword analysis revealed that antitumor activity remains the most extensively studied aspect of acridines and their derivatives.
Collapse
Affiliation(s)
- Diego Santa Clara Marques
- Laboratory of Chemistry and Therapeutic Innovation - LQIT, Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, 50740-600 Recife, PE, Brazil
| | - Lisandra da Silva Lima
- Laboratory of Chemistry and Therapeutic Innovation - LQIT, Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, 50740-600 Recife, PE, Brazil
| | - Josué Filipe de Oliveira Moraes Miranda
- Laboratory of Chemistry and Therapeutic Innovation - LQIT, Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, 50740-600 Recife, PE, Brazil
| | - Carolina Ávila Dos Anjos Santos
- Laboratory of Chemistry and Therapeutic Innovation - LQIT, Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, 50740-600 Recife, PE, Brazil
| | - Iranildo José da Cruz Filho
- Laboratory of Chemistry and Therapeutic Innovation - LQIT, Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, 50740-600 Recife, PE, Brazil.
| | - Maria do Carmo Alves de Lima
- Laboratory of Chemistry and Therapeutic Innovation - LQIT, Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, 50740-600 Recife, PE, Brazil
| |
Collapse
|
4
|
Libera V, Fasolato C, Ripanti F, Catalini S, Bertini L, Petrillo C, Schirò G, D'Amico F, Rossi B, Paciaroni A, Comez L. Molecular mechanisms behind BRACO19 and human telomeric G-quadruplex interaction. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 322:124684. [PMID: 38981290 DOI: 10.1016/j.saa.2024.124684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 07/11/2024]
Abstract
Human telomeres (HTs) can form DNA G-quadruplex (G4), an attractive target for anticancer and antiviral drugs. HT-G4s exhibit inherent structural polymorphism, posing challenges for understanding their specific recognition by ligands. Here, we aim to explore the impact of different topologies within a small segment of the HT (Tel22) on its interaction with BRACO19, a rationally designed G4 ligand with high quadruplex affinity, already employed in in-vivo treatments. Our multi-technique approach is based on the combined use of a set of contactless spectroscopic tools. Circular dichroism and UV resonance Raman spectroscopy probe ligand-induced conformational changes in the G4 sequence, while UV-visible absorption, coupled with steady-state fluorescence spectroscopy, provides further insights into the electronic features of the complex, exploiting the photoresponsive properties of BRACO19. Overall, we find that modifying the topology of the unbound Tel22 through cations (K+ or Na+), serves as a critical determinant for ligand interactions and binding modes, thus influencing the HT-G4's assembly capabilities. Furthermore, we show how fluorescence serves as a valuable probe for recognizing cation-driven multimeric structures, which may be present in living organisms, giving rise to pathological forms.
Collapse
Affiliation(s)
- Valeria Libera
- Department of Physics and Geology, University of Perugia, via Alessandro Pascoli, 06123, Perugia, Italy.
| | - Claudia Fasolato
- Institute for Complex System, National Research Council (ISC-CNR), Piazzale Aldo Moro, 5, 00185, Roma, Italy
| | - Francesca Ripanti
- Department of Physics and Geology, University of Perugia, via Alessandro Pascoli, 06123, Perugia, Italy
| | - Sara Catalini
- Department of Physics and Geology, University of Perugia, via Alessandro Pascoli, 06123, Perugia, Italy; European Laboratory for Non-Linear Spectroscopy (LENS), via Nello Carrara 1, 50019, Sesto Fiorentino (FI), Italy; National Research Council-National Institute of Optics (CNR-INO), Largo Fermi 6, 50125, Florence, Italy
| | - Luca Bertini
- Department of Physics and Geology, University of Perugia, via Alessandro Pascoli, 06123, Perugia, Italy
| | - Caterina Petrillo
- Department of Physics and Geology, University of Perugia, via Alessandro Pascoli, 06123, Perugia, Italy
| | - Giorgio Schirò
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale, F-38044, Grenoble, France
| | - Francesco D'Amico
- Elettra - Sincrotrone Trieste S.C.p.A, s.s. 14 km 163, 500 in Area Science Park, 34149, Trieste, Italy
| | - Barbara Rossi
- Elettra - Sincrotrone Trieste S.C.p.A, s.s. 14 km 163, 500 in Area Science Park, 34149, Trieste, Italy
| | - Alessandro Paciaroni
- Department of Physics and Geology, University of Perugia, via Alessandro Pascoli, 06123, Perugia, Italy
| | - Lucia Comez
- CNR-IOM - Istituto Officina dei Materiali, Via Alessandro Pascoli, 06123, Perugia, Italy.
| |
Collapse
|
5
|
Ambrosio MR, Migliaccio T, Napolitano F, Di Somma S, Maneli G, Amato J, Pagano B, Randazzo A, Portella G, Formisano P, Malfitano AM. Targeting G-quadruplex motifs interferes with differentiation of adipose-derived mesenchymal stem cells. Stem Cell Res Ther 2023; 14:98. [PMID: 37076894 PMCID: PMC10116735 DOI: 10.1186/s13287-023-03320-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/29/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND G-quadruplex (G4) motifs are nucleic acid secondary structures observed in mammalian genomes and transcriptomes able to regulate various cellular processes. Several small molecules have been developed so far to modulate G4 stability, frequently associated with anticancer activity. However, how G4 structures are regulated over homeostatic conditions is mostly unexplored. Here, we used human adipose-derived mesenchymal stem cells (ASCs) to address the role of G4 motifs during adipogenic differentiation. METHODS Adipocyte differentiation of ASCs was investigated in the presence or absence of a well-known G4 ligand, Braco-19. Cell viability was determined by sulforhodamine B assay. Cell dimension and granularity, DNA G4 motifs and cell cycle were detected by flow cytometry. Lipid droplet accumulation was assessed by Oil Red O staining. Cell senescence was evaluated by β-galactosidase staining. Gene expression was measured by qPCR. Protein release in the extracellular medium was quantified by ELISA. RESULTS Braco-19 used at non-cytotoxic concentrations induced morphological changes in mature adipocytes partially restoring an undifferentiated-like status. Braco-19 reduced lipid vacuolization and PPARG, AP2, LEP and TNFA mRNA levels in terminally differentiated cells. No effect was observed in cell senescence, fibrotic markers, IL-6 and IL-8 production, while the secretion of VEGF was dose-dependently reduced. Interestingly, G4 structures were increased in differentiated adipocytes compared to their precursors. Braco-19 treatment reduced G4 content in mature adipocytes. CONCLUSIONS Our data highlight a new role of G4 motifs as genomic structural elements related to human ASC differentiation into mature adipocytes, with potential implications in physio-pathological processes.
Collapse
Affiliation(s)
- Maria Rosaria Ambrosio
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - Teresa Migliaccio
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
| | - Fabiana Napolitano
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
| | - Sarah Di Somma
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
| | - Giovanni Maneli
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
| | - Jussara Amato
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Bruno Pagano
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Antonio Randazzo
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Giuseppe Portella
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - Pietro Formisano
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy.
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy.
| | - Anna Maria Malfitano
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy.
| |
Collapse
|
6
|
Telomerase: A prominent oncological target for development of chemotherapeutic agents. Eur J Med Chem 2023; 249:115121. [PMID: 36669398 DOI: 10.1016/j.ejmech.2023.115121] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/26/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
Telomerase is a ribonucleoprotein (RNP) responsible for the maintenance of chromosomal integrity by stabilizing telomere length. Telomerase is a widely expressed hallmark responsible for replicative immortality in 80-90% of malignant tumors. Cancer cells produce telomerase which prevents telomere shortening by adding telomeres sequences beyond Hayflick's limit; which enables them to divide uncontrollably. The activity of telomerase is relatively low in somatic cells and absent in normal cells, but the re-activation of this RNP in normal cells suppresses p53 activity which leads to the avoidance of senescence causing malignancy. Here, we have focused explicitly on various anti-telomerase therapies and telomerase-inhibiting molecules for the treatment of cancer. We have covered molecules that are reported in developmental, preclinical, and clinical trial stages as potent telomerase inhibitors. Apart from chemotherapy, we have also included details of immunotherapy, gene therapy, G-quadruplex stabilizers, and HSP-90 inhibitors. The purpose of this work is to discuss the challenges behind the development of novel telomerase inhibitors and to identify various perspectives for designing anti-telomerase compounds.
Collapse
|
7
|
Thaichana P, Summart R, Dejkriengkraikul P, Meepowpan P, Lee TR, Tuntiwechapikul W. Hydrosoluble Perylene Monoimide-Based Telomerase Inhibitors with Diminished Cytotoxicity. ACS OMEGA 2022; 7:16746-16756. [PMID: 35601338 PMCID: PMC9118414 DOI: 10.1021/acsomega.2c01343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/08/2022] [Indexed: 06/15/2023]
Abstract
Telomerase is essential for the immortality characteristics of most cancers. Telomerase-specific inhibitors should render cancer cells to replicative senescence without acute cytotoxicity. Perylene-based G-quadruplex (G4) ligands are widely studied as telomerase inhibitors. Most reported perylene-based G4 ligands are perylene diimides (PDIs), which often suffer from self-aggregation in aqueous solutions. Previously, we found that PM2, a perylene monoimide (PMI), exhibited better solubility, G4 binding affinity, and telomerase inhibition than PIPER, the prototypic PDI. However, the acute cytotoxicity of PM2 was about 20-30 times more than PIPER in cancer cells. In this report, we replaced the piperazine side chain of PM2 with ethylenediamine to yield PM3 and replaced the N,N-diethylethylenediamine side chain of PM2 with the 1-(2-aminoethyl) piperidine to yield PM5. We found that asymmetric PMIs with two basic side chains (PM2, PM3, and PM5) performed better than PIPER (the prototypic PDI), in terms of hydrosolubility, G4 binding, in vitro telomerase inhibition, and suppression of human telomerase reverse transcriptase (hTERT) expression and telomerase activity in A549 cells. However, PM5 was 7-10 times less toxic than PM2 and PM3 in three cancer cell lines. We conclude that replacing the N,N-diethylethylenediamine side chain with the 2-aminoethylpiperidine on PMIs reduces the cytotoxicity in cancer cells without impacting G4 binding and telomerase inhibition. This study paves the way for synthesizing new PMIs with drug-like properties for selective telomerase inhibition.
Collapse
Affiliation(s)
- Pak Thaichana
- Department
of Biochemistry, Faculty of Medicine, Chiang
Mai University, Chiang
Mai 50200, Thailand
| | - Ratasark Summart
- Department
of Biochemistry, Faculty of Medicine, Chiang
Mai University, Chiang
Mai 50200, Thailand
| | - Pornngarm Dejkriengkraikul
- Department
of Biochemistry, Faculty of Medicine, Chiang
Mai University, Chiang
Mai 50200, Thailand
- Center
for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Puttinan Meepowpan
- Department
of Chemistry, Faculty of Science, Chiang
Mai University, Chiang Mai 50200, Thailand
| | - T. Randall Lee
- Department
of Chemistry and the Texas Center for Superconductivity, University of Houston, Houston, Texas 77204-5003, United States
| | - Wirote Tuntiwechapikul
- Department
of Biochemistry, Faculty of Medicine, Chiang
Mai University, Chiang
Mai 50200, Thailand
- Center
for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
8
|
Jiang J, Teunens T, Tisaun J, Denuit L, Moucheron C. Ruthenium(II) Polypyridyl Complexes and Their Use as Probes and Photoreactive Agents for G-quadruplexes Labelling. Molecules 2022; 27:1541. [PMID: 35268640 PMCID: PMC8912042 DOI: 10.3390/molecules27051541] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 02/01/2023] Open
Abstract
Due to their optical and electrochemical properties, ruthenium(II) polypyridyl complexes have been used in a wide array of applications. Since the discovery of the light-switch ON effect of [Ru(bpy)2dppz]2+ when interacting with DNA, the design of new Ru(II) complexes as light-up probes for specific regions of DNA has been intensively explored. Amongst them, G-quadruplexes (G4s) are of particular interest. These structures formed by guanine-rich parts of DNA and RNA may be associated with a wide range of biological events. However, locating them and understanding their implications in biological pathways has proven challenging. Elegant approaches to tackle this challenge relies on the use of photoprobes capable of marking, reversibly or irreversibly, these G4s. Indeed, Ru(II) complexes containing ancillary π-deficient TAP ligands can create a covalently linked adduct with G4s after a photoinduced electron transfer from a guanine residue to the excited complex. Through careful design of the ligands, high selectivity of interaction with G4 structures can be achieved. This allows the creation of specific Ru(II) light-up probes and photoreactive agents for G4 labelling, which is at the core of this review composed of an introduction dedicated to a brief description of G-quadruplex structures and two main sections. The first one will provide a general picture of ligands and metal complexes interacting with G4s. The second one will focus on an exhaustive and comprehensive overview of the interactions and (photo)reactions of Ru(II) complexes with G4s.
Collapse
Affiliation(s)
- Julie Jiang
- Laboratoire de Chimie Organique et Photochimie, Service de Chimie et PhysicoChimie Organiques, Université Libre de Bruxelles, Avenue F. D. Roosevelt 50-CP 160/08, 1050 Brussels, Belgium; (J.J.); (T.T.); (J.T.); (L.D.)
| | - Titouan Teunens
- Laboratoire de Chimie Organique et Photochimie, Service de Chimie et PhysicoChimie Organiques, Université Libre de Bruxelles, Avenue F. D. Roosevelt 50-CP 160/08, 1050 Brussels, Belgium; (J.J.); (T.T.); (J.T.); (L.D.)
- Laboratoire de Chimie des Matériaux Nouveaux, Université de Mons, Place du Parc 20, 7000 Mons, Belgium
| | - Jérôme Tisaun
- Laboratoire de Chimie Organique et Photochimie, Service de Chimie et PhysicoChimie Organiques, Université Libre de Bruxelles, Avenue F. D. Roosevelt 50-CP 160/08, 1050 Brussels, Belgium; (J.J.); (T.T.); (J.T.); (L.D.)
| | - Laura Denuit
- Laboratoire de Chimie Organique et Photochimie, Service de Chimie et PhysicoChimie Organiques, Université Libre de Bruxelles, Avenue F. D. Roosevelt 50-CP 160/08, 1050 Brussels, Belgium; (J.J.); (T.T.); (J.T.); (L.D.)
| | - Cécile Moucheron
- Laboratoire de Chimie Organique et Photochimie, Service de Chimie et PhysicoChimie Organiques, Université Libre de Bruxelles, Avenue F. D. Roosevelt 50-CP 160/08, 1050 Brussels, Belgium; (J.J.); (T.T.); (J.T.); (L.D.)
| |
Collapse
|
9
|
Wang Y, Li C, Hao X, Wang L, Ma X, Jin R, Kang C, Gao L. A naphthyridine-indole ligand for selective stabilization of G-quadruplexes and conformational conversion of hybrid topology. Bioorg Med Chem 2021; 48:116416. [PMID: 34560615 DOI: 10.1016/j.bmc.2021.116416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/12/2021] [Accepted: 09/13/2021] [Indexed: 11/15/2022]
Abstract
The development of ligands to stabilize G-quadruplexes (G4s) or induce G4s to transition from metastable topology to stable topology is a potential strategy for inhibiting cancer cell proliferation. In this study, a novel G-quadruplex (G4) ligand based on a naphthyridine scaffold with two indole pendants, L5-DA, is reported to convert hybrid to the parallel topology. Circular dichroism (CD) and fluorescence spectroscopies were used to investigate the interactions between L5-DA and G4s. The CD spectra revealed that the L5-DA induced the conformational conversion from hybrid topologies to parallel topologies with a melting temperature increase of more than 30 °C. According to Förster resonance energy transfer assays, the presence of excess duplex competitor had no effect on the ligand-induced stabilization of the hybrid topology, confirming the L5-DA's selectivity for G4s over ds26. With IC50 values of 4.3 μM, the ligand showed significant cytotoxicity against HeLa cells and effectively induced growth inhibition and apoptosis in HeLa cells. Immunofluorescence microscopy revealed an increase in BG4 foci in the presence of the L5-DA, confirming ligand-induced G4s stabilization in HeLa cells. According to these results, the combination of naphthyridine and indole scaffold was an effective design strategy for G4s stabilization and conformational conversion of metastable G4 topology for inhibiting cancer cell growth.
Collapse
Affiliation(s)
- Yu Wang
- Laboratory of Polymer Composite and Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; University of Science and Technology of China, Hefei 230026, China
| | - Chunjie Li
- Laboratory of Polymer Composite and Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xueyu Hao
- Laboratory of Polymer Composite and Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; University of Science and Technology of China, Hefei 230026, China
| | - Liangpeng Wang
- Laboratory of Polymer Composite and Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xiaoye Ma
- Laboratory of Polymer Composite and Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Rizhe Jin
- Laboratory of Polymer Composite and Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Chuanqing Kang
- Laboratory of Polymer Composite and Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; University of Science and Technology of China, Hefei 230026, China.
| | - Lianxun Gao
- Laboratory of Polymer Composite and Engineering, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| |
Collapse
|
10
|
Dwivedi A, Kumari A, Aarthy M, Singh SK, Ojha M, Jha S, Jha SK, Jha NS. Spectroscopic and molecular docking studies for the binding and interaction aspects of curcumin-cysteine conjugate and rosmarinic acid with human telomeric G-quadruplex DNA. Int J Biol Macromol 2021; 182:1463-1472. [PMID: 34015406 DOI: 10.1016/j.ijbiomac.2021.05.089] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/24/2022]
Abstract
The binding and interaction aspects of potential anticancer ligands like: curcumin-cysteine (CC) and rosmarinic acid (RA) with human telomeric G-quadruplex DNA, a novel anticancer target, have been probed by spectroscopic and molecular docking approach. The circular dichroism study unravels the conformational switching from mixed hybrid to parallel structure for the short sequence of human telomeric G-quadruplex structure in the presence of both the ligands. Further a good correlation for binding affinity has been established from the emission and absorption binding spectrum analysis. Further our spectroscopic and molecular docking studies have suggested that the CC having better binding capability than RA to human telomeric G-quadruplex. The presence of L-cysteine moiety in CC ligand is responsible factor for its binding via both minor as well as major groove of human telomeric G-quadruplex DNA where-as RA binds only via minor groove of telomeric G-DNA.
Collapse
Affiliation(s)
- Awadesh Dwivedi
- Department of Chemistry, National Institute of Technology, Patna 800005, India
| | - Arya Kumari
- Department of Chemistry, National Institute of Technology, Patna 800005, India
| | - Murali Aarthy
- Computer Aided Drug Design and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Karaikudi 630003, Tamil Nadu, India
| | - Sanjeev Kumar Singh
- Computer Aided Drug Design and Molecular Modelling Lab, Department of Bioinformatics, Alagappa University, Karaikudi 630003, Tamil Nadu, India
| | - Monalisha Ojha
- Department of Life Science, National Institute of Technology, Rourkela 769008, India
| | - Suman Jha
- Department of Life Science, National Institute of Technology, Rourkela 769008, India
| | | | - Niki Sweta Jha
- Department of Chemistry, National Institute of Technology, Patna 800005, India.
| |
Collapse
|
11
|
Lejault P, Mitteaux J, Sperti FR, Monchaud D. How to untie G-quadruplex knots and why? Cell Chem Biol 2021; 28:436-455. [PMID: 33596431 DOI: 10.1016/j.chembiol.2021.01.015] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/08/2020] [Accepted: 01/20/2021] [Indexed: 12/12/2022]
Abstract
For over two decades, the prime objective of the chemical biology community studying G-quadruplexes (G4s) has been to use chemicals to interact with and stabilize G4s in cells to obtain mechanistic interpretations. This strategy has been undoubtedly successful, as demonstrated by recent advances. However, these insights have also led to a fundamental rethinking of G4-targeting strategies: due to the prevalence of G4s in the human genome, transcriptome, and ncRNAome (collectively referred to as the G4ome), and their involvement in human diseases, should we continue developing G4-stabilizing ligands or should we invest in designing molecular tools to unfold G4s? Here, we first focus on how, when, and where G4s fold in cells; then, we describe the enzymatic systems that have evolved to counteract G4 folding and how they have been used as tools to manipulate G4s in cells; finally, we present strategies currently being implemented to devise new molecular G4 unwinding agents.
Collapse
Affiliation(s)
- Pauline Lejault
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB CNRS UMR 6302, UBFC Dijon, France
| | - Jérémie Mitteaux
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB CNRS UMR 6302, UBFC Dijon, France
| | - Francesco Rota Sperti
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB CNRS UMR 6302, UBFC Dijon, France
| | - David Monchaud
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB CNRS UMR 6302, UBFC Dijon, France.
| |
Collapse
|
12
|
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
|
13
|
Sasaki S, Ma Y, Ishizuka T, Bao HL, Hirokawa T, Xu Y, Tera M, Nagasawa K. Linear consecutive hexaoxazoles as G4 ligands inducing chair-type anti-parallel topology of a telomeric G-quadruplex. RSC Adv 2020; 10:43319-43323. [PMID: 35519695 PMCID: PMC9058415 DOI: 10.1039/d0ra09413g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
G-quadruplex structures (G4s) in guanine-rich regions of DNA play critical roles in various biological phenomena, including replication, translation, and gene expression. There are three types of G4 topology, i.e., parallel, anti-parallel, and hybrid, and ligands that selectively interact with or stabilize a specific topology have been extensively explored to enable studies of topology-related functions. Here, we describe the synthesis of a new series of G4 ligands based on 6LCOs (6-linear consecutive oxazoles), i.e., L2H2-2M2EA-6LCO (2), L2A2-2M2EAc-6LCO (3), and L2G2-2M2EG-6LCO (4), which bear four aminoalkyl, acetamidealkyl, and guanidinylalkyl side chains, respectively. Among them, ligand 2 stabilized telomeric G4 and induced anti-parallel topology independently of the presence of cations. The anti-parallel topology induced by 2 was identified as chair-type by means of 19F NMR spectroscopy and fluorescence experiments with 2-aminopurine-labeled DNA.
Collapse
Affiliation(s)
- Shogo Sasaki
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology 2-24-16 Naka-cho, Koganei Tokyo 184-8588 Japan
| | - Yue Ma
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology 2-24-16 Naka-cho, Koganei Tokyo 184-8588 Japan
| | - Takumi Ishizuka
- Division of Chemistry, Department of Medical Sciences, Faculty of Medicine, University of Miyazak 5200 Kihara, Kiyotake Miyazaki 889-1692 Japan
| | - Hong-Liang Bao
- Division of Chemistry, Department of Medical Sciences, Faculty of Medicine, University of Miyazak 5200 Kihara, Kiyotake Miyazaki 889-1692 Japan
| | - Takatsugu Hirokawa
- Transborder Medical Research Center, University of Tsukuba 1-1-1 Tennodai Tsukuba, 305-8575 Japan
- Division of Biomedical Science, University of Tsukuba 1-1-1 Tennodai Tsukuba, 305-8575 Japan
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology 2-4-7 Aomi, Koto-ward Tokyo 135-0064 Japan
| | - Yan Xu
- Division of Chemistry, Department of Medical Sciences, Faculty of Medicine, University of Miyazak 5200 Kihara, Kiyotake Miyazaki 889-1692 Japan
| | - Masayuki Tera
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology 2-24-16 Naka-cho, Koganei Tokyo 184-8588 Japan
| | - Kazuo Nagasawa
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology 2-24-16 Naka-cho, Koganei Tokyo 184-8588 Japan
| |
Collapse
|
14
|
Synthesis, Biological Evaluation and Stability Studies of Some Novel Aza-Acridine Aminoderivatives. Molecules 2020; 25:molecules25194584. [PMID: 33049986 PMCID: PMC7582268 DOI: 10.3390/molecules25194584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 01/10/2023] Open
Abstract
Several new amino-substituted aza-acridine derivatives bearing a basic side chain have been designed and synthesized. The antiproliferative activity of the target compounds has been evaluated against three cancer cell lines-namely HCT-116 (colorectal), the uterine sarcoma MES-SA, and its doxorubicin-resistant variant MES-SA/Dx5. A limited number of the new acridines showed marginal cytotoxicity against the tested cell lines; nevertheless, these analogues possessed a similar substitution pattern. The moderate biological activity of these derivatives was attributed to their instability in aqueous media, which has been studied by mass spectrometry and computational chemistry experiments at the density functional level of theory (DFT).
Collapse
|
15
|
Berei J, Eckburg A, Miliavski E, Anderson AD, Miller RJ, Dein J, Giuffre AM, Tang D, Deb S, Racherla KS, Patel M, Vela MS, Puri N. Potential Telomere-Related Pharmacological Targets. Curr Top Med Chem 2020; 20:458-484. [DOI: 10.2174/1568026620666200109114339] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/22/2022]
Abstract
Telomeres function as protective caps at the terminal portion of chromosomes, containing
non-coding nucleotide sequence repeats. As part of their protective function, telomeres preserve genomic
integrity and minimize chromosomal exposure, thus limiting DNA damage responses. With
continued mitotic divisions in normal cells, telomeres progressively shorten until they reach a threshold
at a point where they activate senescence or cell death pathways. However, the presence of the enzyme
telomerase can provide functional immortality to the cells that have reached or progressed past
senescence. In senescent cells that amass several oncogenic mutations, cancer formation can occur due
to genomic instability and the induction of telomerase activity. Telomerase has been found to be expressed
in over 85% of human tumors and is labeled as a near-universal marker for cancer. Due to this
feature being present in a majority of tumors but absent in most somatic cells, telomerase and telomeres
have become promising targets for the development of new and effective anticancer therapeutics.
In this review, we evaluate novel anticancer targets in development which aim to alter telomerase
or telomere function. Additionally, we analyze the progress that has been made, including preclinical
studies and clinical trials, with therapeutics directed at telomere-related targets. Furthermore, we review
the potential telomere-related therapeutics that are used in combination therapy with more traditional
cancer treatments. Throughout the review, topics related to medicinal chemistry are discussed,
including drug bioavailability and delivery, chemical structure-activity relationships of select therapies,
and the development of a unique telomere assay to analyze compounds affecting telomere elongation.
Collapse
Affiliation(s)
- Joseph Berei
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Adam Eckburg
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Edward Miliavski
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Austin D. Anderson
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Rachel J. Miller
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Joshua Dein
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Allison M. Giuffre
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Diana Tang
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Shreya Deb
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Kavya Sri Racherla
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Meet Patel
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Monica Saravana Vela
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Neelu Puri
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| |
Collapse
|
16
|
Ma Y, Iida K, Nagasawa K. Topologies of G-quadruplex: Biological functions and regulation by ligands. Biochem Biophys Res Commun 2020; 531:3-17. [PMID: 31948752 DOI: 10.1016/j.bbrc.2019.12.103] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 12/20/2019] [Accepted: 12/28/2019] [Indexed: 01/06/2023]
Abstract
G-Quadruplex (G4) is one of the higher-order structures occurring in guanine-rich sequences of nucleic acids, and plays critical roles in biological processes. The G4-forming sequences can generate three kinds of topologies, i.e., parallel, anti-parallel, and hybrid, and these polymorphic structures have an important influence on G4-related biological functions. In this review, we highlight variety of structures generated by G4s containing various sequences and under diverse conditions. We also discuss the G4 ligands which induce specific topologies and/or conversion between different topologies.
Collapse
Affiliation(s)
- Yue Ma
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Japan.
| | - Keisuke Iida
- Department of Chemistry, Chiba University, Japan
| | - Kazuo Nagasawa
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Japan.
| |
Collapse
|
17
|
Umar MI, Ji D, Chan CY, Kwok CK. G-Quadruplex-Based Fluorescent Turn-On Ligands and Aptamers: From Development to Applications. Molecules 2019; 24:E2416. [PMID: 31262059 PMCID: PMC6650947 DOI: 10.3390/molecules24132416] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 06/17/2019] [Accepted: 06/24/2019] [Indexed: 02/08/2023] Open
Abstract
Guanine (G)-quadruplexes (G4s) are unique nucleic acid structures that are formed by stacked G-tetrads in G-rich DNA or RNA sequences. G4s have been reported to play significant roles in various cellular events in both macro- and micro-organisms. The identification and characterization of G4s can help to understand their different biological roles and potential applications in diagnosis and therapy. In addition to biophysical and biochemical methods to interrogate G4 formation, G4 fluorescent turn-on ligands can be used to target and visualize G4 formation both in vitro and in cells. Here, we review several representative classes of G4 fluorescent turn-on ligands in terms of their interaction mechanism and application perspectives. Interestingly, G4 structures are commonly identified in DNA and RNA aptamers against targets that include proteins and small molecules, which can be utilized as G4 tools for diverse applications. We therefore also summarize the recent development of G4-containing aptamers and highlight their applications in biosensing, bioimaging, and therapy. Moreover, we discuss the current challenges and future perspectives of G4 fluorescent turn-on ligands and G4-containing aptamers.
Collapse
Affiliation(s)
- Mubarak I Umar
- Department of Chemistry, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Danyang Ji
- Department of Chemistry, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Chun-Yin Chan
- Department of Chemistry, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Chun Kit Kwok
- Department of Chemistry, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China.
| |
Collapse
|
18
|
Lee YH, Chen YY, Yeh YL, Wang YJ, Chen RJ. Stilbene Compounds Inhibit Tumor Growth by the Induction of Cellular Senescence and the Inhibition of Telomerase Activity. Int J Mol Sci 2019; 20:ijms20112716. [PMID: 31159515 PMCID: PMC6600253 DOI: 10.3390/ijms20112716] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/17/2022] Open
Abstract
Cellular senescence is a state of cell cycle arrest characterized by a distinct morphology, gene expression pattern, and secretory phenotype. It can be triggered by multiple mechanisms, including those involved in telomere shortening, the accumulation of DNA damage, epigenetic pathways, and the senescence-associated secretory phenotype (SASP), and so on. In current cancer therapy, cellular senescence has emerged as a potent tumor suppression mechanism that restrains proliferation in cells at risk for malignant transformation. Therefore, compounds that stimulate the growth inhibition effects of senescence while limiting its detrimental effects are believed to have great clinical potential. In this review article, we first review the current knowledge of the pro- and antitumorigeneic functions of senescence and summarize the key roles of telomerase in the regulation of senescence in tumors. Second, we review the current literature regarding the anticancer effects of stilbene compounds that are mediated by the targeting of telomerase and cell senescence. Finally, we provide future perspectives on the clinical utilization of stilbene compounds, especially resveratrol and pterostilbene, as novel cancer therapeutic remedies. We conclude and propose that stilbene compounds may induce senescence and may potentially be used as the therapeutic or adjuvant agents for cancers with high telomerase activity.
Collapse
Affiliation(s)
- Yu-Hsuan Lee
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| | - Yu-Ying Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| | - Ya-Ling Yeh
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan.
| | - Rong-Jane Chen
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| |
Collapse
|
19
|
Binding of BRACO19 to a Telomeric G-Quadruplex DNA Probed by All-Atom Molecular Dynamics Simulations with Explicit Solvent. Molecules 2019; 24:molecules24061010. [PMID: 30871220 PMCID: PMC6471034 DOI: 10.3390/molecules24061010] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/01/2019] [Accepted: 03/09/2019] [Indexed: 11/30/2022] Open
Abstract
Although BRACO19 is a potent G-quadruplex binder, its potential for clinical usage is hindered by its low selectivity towards DNA G-quadruplex over duplex. High-resolution structures of BRACO19 in complex with neither single-stranded telomeric DNA G-quadruplexes nor B-DNA duplex are available. In this study, the binding pathway of BRACO19 was probed by 27.5 µs molecular dynamics binding simulations with a free ligand (BRACO19) to a DNA duplex and three different topological folds of the human telomeric DNA G-quadruplex (parallel, anti-parallel and hybrid). The most stable binding modes were identified as end stacking and groove binding for the DNA G-quadruplexes and duplex, respectively. Among the three G-quadruplex topologies, the MM-GBSA binding energy analysis suggested that BRACO19′s binding to the parallel scaffold was most energetically favorable. The two lines of conflicting evidence plus our binding energy data suggest conformation-selection mechanism: the relative population shift of three scaffolds upon BRACO19 binding (i.e., an increase of population of parallel scaffold, a decrease of populations of antiparallel and/or hybrid scaffold). This hypothesis appears to be consistent with the fact that BRACO19 was specifically designed based on the structural requirements of the parallel scaffold and has since proven effective against a variety of cancer cell lines as well as toward a number of scaffolds. In addition, this binding mode is only slightly more favorable than BRACO19s binding to the duplex, explaining the low binding selectivity of BRACO19 to G-quadruplexes over duplex DNA. Our detailed analysis suggests that BRACO19′s groove binding mode may not be stable enough to maintain a prolonged binding event and that the groove binding mode may function as an intermediate state preceding a more energetically favorable end stacking pose; base flipping played an important role in enhancing binding interactions, an integral feature of an induced fit binding mechanism.
Collapse
|
20
|
Developing Novel G-Quadruplex Ligands: from Interaction with Nucleic Acids to Interfering with Nucleic Acid⁻Protein Interaction. Molecules 2019; 24:molecules24030396. [PMID: 30678288 PMCID: PMC6384609 DOI: 10.3390/molecules24030396] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/10/2019] [Accepted: 01/22/2019] [Indexed: 12/20/2022] Open
Abstract
G-quadruplex is a special secondary structure of nucleic acids in guanine-rich sequences of genome. G-quadruplexes have been proved to be involved in the regulation of replication, DNA damage repair, and transcription and translation of oncogenes or other cancer-related genes. Therefore, targeting G-quadruplexes has become a novel promising anti-tumor strategy. Different kinds of small molecules targeting the G-quadruplexes have been designed, synthesized, and identified as potential anti-tumor agents, including molecules directly bind to the G-quadruplex and molecules interfering with the binding between the G-quadruplex structures and related binding proteins. This review will explore the feasibility of G-quadruplex ligands acting as anti-tumor drugs, from basis to application. Meanwhile, since helicase is the most well-defined G-quadruplex-related protein, the most extensive research on the relationship between helicase and G-quadruplexes, and its meaning in drug design, is emphasized.
Collapse
|
21
|
Sullivan HJ, Readmond C, Radicella C, Persad V, Fasano TJ, Wu C. Binding of Telomestatin, TMPyP4, BSU6037, and BRACO19 to a Telomeric G-Quadruplex-Duplex Hybrid Probed by All-Atom Molecular Dynamics Simulations with Explicit Solvent. ACS OMEGA 2018; 3:14788-14806. [PMID: 30555989 PMCID: PMC6289566 DOI: 10.1021/acsomega.8b01574] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 09/11/2018] [Indexed: 06/09/2023]
Abstract
A promising anticancer therapeutic strategy is the stabilization of telomeric G-quadruplexes using G-quadruplex-binding small molecules. Although many G-quadruplex-specific ligands have been developed, their low potency and selectivity to G-quadruplexes over duplex remains unsolved. Recently, a crystal structure of a telomeric 3' quadruplex-duplex hybrid was reported and the quadruplex-duplex interface was suggested to a good target to address the issues. However, there are no high-resolution complex structures reported for G-quadruplex ligands except for a docked BSU6037. In this study, molecular dynamic (MD) binding simulations with a free ligand were used to study binding poses and dynamics of four representative ligands: telomestatin, TMPyP4, BSU6037, and BRACO19. The MD data showed that BSU6037 was able to fully intercalate into the interface whereas TMPyP4 and BRACO19 could only maintain partial intercalation into the interface and telomestatin only binds at the quadruplex and duplex ends. Both linear ligands, BSU6037 and BRACO19, were able to interact with the interface, yet they were not selective over duplex DNA. The DNA geometry, binding modes, and binding pathways were systematically characterized, and the binding energy was calculated and compared for each system. The interaction of the ligands to the interface was by the means of an induced-fit binding mechanism rather than a lock-key mechanism, consisting of the DNA unfolding at the interface to allow entrance of the drug and then the refolding and repacking of the DNA and the ligand to further stabilize the G-quadruplex. On the basis of the findings in this study, modifications were suggested to optimize the interface binding for TMPyp4 and telomestatin.
Collapse
Affiliation(s)
- Holli-Joi Sullivan
- Chemistry
& Biochemistry and Department of Molecular & Cellular Biosciences, College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| | - Carolyn Readmond
- Chemistry
& Biochemistry and Department of Molecular & Cellular Biosciences, College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| | - Christina Radicella
- Chemistry
& Biochemistry and Department of Molecular & Cellular Biosciences, College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| | - Victoria Persad
- Chemistry
& Biochemistry and Department of Molecular & Cellular Biosciences, College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| | - Thomas J. Fasano
- Chemistry
& Biochemistry and Department of Molecular & Cellular Biosciences, College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| | - Chun Wu
- Chemistry
& Biochemistry and Department of Molecular & Cellular Biosciences, College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| |
Collapse
|
22
|
Rajasekhar B, Kumar C, Premkumar G, Riyaz MAB, Lakshmi PTV, Swu T. Computational studies on G-quadruplex DNA-stabilizing property of novel Wittig-based Schiff-Base ligands and their copper(II) complexes. Struct Chem 2018. [DOI: 10.1007/s11224-018-1229-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
23
|
Cichorek M, Ronowska A, Gensicka-Kowalewska M, Deptula M, Pelikant-Malecka I, Dzierzbicka K. Novel therapeutic compound acridine-retrotuftsin action on biological forms of melanoma and neuroblastoma. J Cancer Res Clin Oncol 2018; 145:165-179. [PMID: 30367436 PMCID: PMC6326014 DOI: 10.1007/s00432-018-2776-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/19/2018] [Indexed: 12/29/2022]
Abstract
PURPOSE As a continuation of our search for anticancer agents, we have synthesized a new acridine-retrotuftsin analog HClx9-[Arg(NO2)-Pro-Lys-Thr-OCH3]-1-nitroacridine (named ART) and have evaluated its activity against melanoma and neuroblastoma lines. Both tumors develop from cells (melanocytes, neurons) of neuroectodermal origin, and both are tumors with high heterogeneity and unsatisfactory susceptibility to chemotherapies. Thus, we analyzed the action of ART on pairs of biological forms of melanoma (amelanotic and melanotic) and neuroblastoma (dopaminergic and cholinergic) with regard to proliferation, mechanism of cell death, and effect on the activity of tricarboxylic acid cycle (TAC) enzymes. METHODS The cytotoxicity of ART was evaluated by XTT and trypan blue tests. Cell death was estimated by plasma membrane structure changes (phosphatidylserine and calreticulin externalization), caspase activation, presence of ROS (reactive oxygen species), activity of tricarboxylic acid cycle enzymes (pyruvate dehydrogenase complex, aconitase, and isocitrate dehydrogenase), NAD level, and ATP level. RESULTS ART influences the biological forms of melanoma and neuroblastoma in different ways. Amelanotic (Ab) melanoma (with the inhibited melanogenesis, higher malignancy) and SHSY5Y neuroblastoma (with cholinergic DC cells) were especially sensitive to ART action. The Ab melanoma cells died through apoptosis, while, with SH-SY5Y-DC neuroblastoma, the number of cells decreased but not as a result of apoptosis. With Ab melanoma and SH-SY5Y-DC cells, a diminished activity of TAC enzymes was noticed, along with ATP/NAD depletion. CONCLUSION Our data show that the biological forms of certain tumors responded in different ways to the action of ART. As a combination of retrotuftsin and acridine, the compound can be an inducer of apoptotic cell death of melanoma, especially the amelanotic form. Although the mechanism of the interrelationships between energy metabolism and cell death is not fully understood, interference of ART with TAC enzymes could encourage the further investigation of its anticancer action.
Collapse
Affiliation(s)
- Miroslawa Cichorek
- Department of Embryology, Medical University of Gdansk, Debinki 1 St, 80-210, Gdansk, PL, Poland.
| | - Anna Ronowska
- Department of Laboratory Medicine, Medical University of Gdansk, Debinki 7 St, 80-211, Gdansk, PL, Poland
| | - Monika Gensicka-Kowalewska
- Department of Organic Chemistry, Gdansk University of Technology, Narutowicza St 11/12, 80-233, Gdansk, PL, Poland
| | - Milena Deptula
- Department of Embryology, Medical University of Gdansk, Debinki 1 St, 80-210, Gdansk, PL, Poland
| | - Iwona Pelikant-Malecka
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 St, 80-210, Gdansk, PL, Poland
| | - Krystyna Dzierzbicka
- Department of Organic Chemistry, Gdansk University of Technology, Narutowicza St 11/12, 80-233, Gdansk, PL, Poland
| |
Collapse
|
24
|
Machireddy B, Kalra G, Jonnalagadda S, Ramanujachary K, Wu C. Probing the Binding Pathway of BRACO19 to a Parallel-Stranded Human Telomeric G-Quadruplex Using Molecular Dynamics Binding Simulation with AMBER DNA OL15 and Ligand GAFF2 Force Fields. J Chem Inf Model 2017; 57:2846-2864. [PMID: 29028340 DOI: 10.1021/acs.jcim.7b00287] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human telomeric DNA G-quadruplex has been identified as a good therapeutic target in cancer treatment. G-quadruplex-specific ligands that stabilize the G-quadruplex have great potential to be developed as anticancer agents. Two crystal structures (an apo form of parallel stranded human telomeric G-quadruplex and its holo form in complex with BRACO19, a potent G-quadruplex ligand) have been solved, yet the binding mechanism and pathway remain elusive. In this study, we simulated the binding of a free BRACO19 molecule to the apo form of the G-quadruplex using the latest AMBER DNA (OL15) and ligand (GAFF2) force fields. Three binding modes have been identified: top stacking, bottom intercalation, and groove binding. Bottom intercalation (51% of the population) resembles the bottom binding pose in the complex crystal structure very well. The groove binding mode is less stable than the bottom binding mode and is likely to be an intermediate state leading to the bottom binding mode. A flip-insertion mechanism was observed in the bottom intercalation mode, during which flipping of the bases outward makes space for ligand insertion, after which the bases flip back to increase the stability of the complex. In addition to reproducing the base-flipping behavior for some loop residues upon ligand binding, the direct alignment type of the ATAT-tetrad was observed in our simulations for the first time. These successes provide initial support for using this combination of the OL15 and GAFF2 force fields to study quadruplex-ligand interactions.
Collapse
Affiliation(s)
- Babitha Machireddy
- College of Science and Mathematics, Rowan University , Glassboro, New Jersey 08028, United States
| | - Gurmannat Kalra
- College of Science and Mathematics, Rowan University , Glassboro, New Jersey 08028, United States
| | - Subash Jonnalagadda
- College of Science and Mathematics, Rowan University , Glassboro, New Jersey 08028, United States
| | - Kandalam Ramanujachary
- College of Science and Mathematics, Rowan University , Glassboro, New Jersey 08028, United States
| | - Chun Wu
- College of Science and Mathematics, Rowan University , Glassboro, New Jersey 08028, United States
| |
Collapse
|
25
|
Treating Cancer by Targeting Telomeres and Telomerase. Antioxidants (Basel) 2017; 6:antiox6010015. [PMID: 28218725 PMCID: PMC5384178 DOI: 10.3390/antiox6010015] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/15/2017] [Accepted: 02/16/2017] [Indexed: 12/22/2022] Open
Abstract
Telomerase is expressed in more than 85% of cancer cells. Tumor cells with metastatic potential may have a high telomerase activity, allowing cells to escape from the inhibition of cell proliferation due to shortened telomeres. Human telomerase primarily consists of two main components: hTERT, a catalytic subunit, and hTR, an RNA template whose sequence is complimentary to the telomeric 5′-dTTAGGG-3′ repeat. In humans, telomerase activity is typically restricted to renewing tissues, such as germ cells and stem cells, and is generally absent in normal cells. While hTR is constitutively expressed in most tissue types, hTERT expression levels are low enough that telomere length cannot be maintained, which sets a proliferative lifespan on normal cells. However, in the majority of cancers, telomerase maintains stable telomere length, thereby conferring cell immortality. Levels of hTERT mRNA are directly related to telomerase activity, thereby making it a more suitable therapeutic target than hTR. Recent data suggests that stabilization of telomeric G-quadruplexes may act to indirectly inhibit telomerase action by blocking hTR binding. Telomeric DNA has the propensity to spontaneously form intramolecular G-quadruplexes, four-stranded DNA secondary structures that are stabilized by the stacking of guanine residues in a planar arrangement. The functional roles of telomeric G-quadruplexes are not completely understood, but recent evidence suggests that they can stall the replication fork during DNA synthesis and inhibit telomere replication by preventing telomerase and related proteins from binding to the telomere. Long-term treatment with G-quadruplex stabilizers induces a gradual reduction in the length of the G-rich 3’ end of the telomere without a reduction of the total telomere length, suggesting that telomerase activity is inhibited. However, inhibition of telomerase, either directly or indirectly, has shown only moderate success in cancer patients. Another promising approach of targeting the telomere is the use of guanine-rich oligonucleotides (GROs) homologous to the 3’ telomere overhang sequence (T-oligos). T-oligos, particularly a specific 11-base oligonucleotide (5’-dGTTAGGGTTAG-3’) called T11, have been shown to induce DNA damage responses (DDRs) such as senescence, apoptosis, and cell cycle arrest in numerous cancer cell types with minimal or no cytostatic effects in normal, non-transformed cells. As a result, T-oligos and other GROs are being investigated as prospective anticancer therapeutics. Interestingly, the DDRs induced by T-oligos in cancer cells are similar to the effects seen after progressive telomere degradation in normal cells. The loss of telomeres is an important tumor suppressor mechanism that is commonly absent in transformed malignant cells, and hence, T-oligos have garnered significant interest as a novel strategy to combat cancer. However, little is known about their mechanism of action. In this review, we discuss the current understanding of how T-oligos exert their antiproliferative effects in cancer cells and their role in inhibition of telomerase. We also discuss the current understanding of telomerase in cancer and various therapeutic targets related to the telomeres and telomerase.
Collapse
|
26
|
Sabolová D, Vilková M, Imrich J, Potočňák I. New spiroacridine derivatives with DNA-binding and topoisomerase I inhibition activity. Tetrahedron Lett 2016. [DOI: 10.1016/j.tetlet.2016.10.108] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Kang HJ, Cui Y, Yin H, Scheid A, Hendricks WPD, Schmidt J, Sekulic A, Kong D, Trent JM, Gokhale V, Mao H, Hurley LH. A Pharmacological Chaperone Molecule Induces Cancer Cell Death by Restoring Tertiary DNA Structures in Mutant hTERT Promoters. J Am Chem Soc 2016; 138:13673-13692. [PMID: 27643954 DOI: 10.1021/jacs.6b07598] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Activation of human telomerase reverse transcriptase (hTERT) is necessary for limitless replication in tumorigenesis. Whereas hTERT is transcriptionally silenced in normal cells, most tumor cells reactivate hTERT expression by alleviating transcriptional repression through diverse genetic and epigenetic mechanisms. Transcription-activating hTERT promoter mutations have been found to occur at high frequencies in multiple cancer types. These mutations have been shown to form new transcription factor binding sites that drive hTERT expression, but this model cannot fully account for differences in wild-type (WT) and mutant promoter activation and has not yet enabled a selective therapeutic strategy. Here, we demonstrate a novel mechanism by which promoter mutations activate hTERT transcription, which also sheds light on a unique therapeutic opportunity. Promoter mutations occur in a core promoter region that forms tertiary structures consisting of a pair of G-quadruplexes involved in transcriptional silencing. We show that promoter mutations exert a detrimental effect on the folding of one of these G-quadruplexes, resulting in a nonfunctional silencer element that alleviates transcriptional repression. We have also identified a small drug-like pharmacological chaperone (pharmacoperone) molecule, GTC365, that acts at an early step in the G-quadruplex folding pathway to redirect mutant promoter G-quadruplex misfolding, partially reinstate the correct folding pathway, and reduce hTERT activity through transcriptional repression. This transcription-mediated repression produces cancer cell death through multiple routes including both induction of apoptosis through inhibition of hTERT's role in regulating apoptosis-related proteins and induction of senescence by decreasing telomerase activity and telomere length. We demonstrate the selective therapeutic potential of this strategy in melanoma cells that overexpress hTERT.
Collapse
Affiliation(s)
- Hyun-Jin Kang
- University of Arizona , College of Pharmacy, 1703 East Mabel Street, Tucson, Arizona 85721, United States
| | - Yunxi Cui
- Department of Chemistry and Biochemistry and School of Biomedical Sciences, Kent State University , Kent, Ohio 44242, United States
| | - Holly Yin
- Translational Genomics Research Institute , 445 North Fifth Street, Phoenix, Arizona 85004, United States
| | - Amy Scheid
- College of Science, University of Arizona , 1040 East Fourth Street, Tucson, Arizona 85721, United States
| | - William P D Hendricks
- Translational Genomics Research Institute , 445 North Fifth Street, Phoenix, Arizona 85004, United States
| | - Jessica Schmidt
- Department of Dermatology, Mayo Clinic , 13400 East Shea Boulevard, Scottsdale, Arizona 85259, United States
| | - Aleksandar Sekulic
- Department of Dermatology, Mayo Clinic , 13400 East Shea Boulevard, Scottsdale, Arizona 85259, United States
| | - Deming Kong
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University , Tianjin 300071, People's Republic of China
| | - Jeffrey M Trent
- Translational Genomics Research Institute , 445 North Fifth Street, Phoenix, Arizona 85004, United States
| | - Vijay Gokhale
- BIO5 Institute , 1657 East Helen Street, Tucson, Arizona 85721, United States
| | - Hanbin Mao
- Department of Chemistry and Biochemistry and School of Biomedical Sciences, Kent State University , Kent, Ohio 44242, United States
| | - Laurence H Hurley
- University of Arizona , College of Pharmacy, 1703 East Mabel Street, Tucson, Arizona 85721, United States.,BIO5 Institute , 1657 East Helen Street, Tucson, Arizona 85721, United States.,Arizona Cancer Center , 1515 North Campbell Avenue, Tucson, Arizona 85724, United States
| |
Collapse
|
28
|
Mulholland K, Wu C. Binding of Telomestatin to a Telomeric G-Quadruplex DNA Probed by All-Atom Molecular Dynamics Simulations with Explicit Solvent. J Chem Inf Model 2016; 56:2093-2102. [DOI: 10.1021/acs.jcim.6b00473] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Kelly Mulholland
- College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| | - Chun Wu
- College of Science and Mathematics, Rowan University, Glassboro, New Jersey 08028, United States
| |
Collapse
|
29
|
Chattoraj S, Bhattacharyya K. Biological oscillations: Fluorescence monitoring by confocal microscopy. Chem Phys Lett 2016. [DOI: 10.1016/j.cplett.2016.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
30
|
Recent advances in targeting the telomeric G-quadruplex DNA sequence with small molecules as a strategy for anticancer therapies. Future Med Chem 2016; 8:1259-90. [PMID: 27442231 DOI: 10.4155/fmc-2015-0017] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Human telomeric DNA (hTelo), present at the ends of chromosomes to protect their integrity during cell division, comprises tandem repeats of the sequence d(TTAGGG) which is known to form a G-quadruplex secondary structure. This unique structural formation of DNA is distinct from the well-known helical structure that most genomic DNA is thought to adopt, and has recently gained prominence as a molecular target for new types of anticancer agents. In particular, compounds that can stabilize the intramolecular G-quadruplex formed within the human telomeric DNA sequence can inhibit the activity of the enzyme telomerase which is known to be upregulated in tumor cells and is a major contributor to their immortality. This provides the basis for the discovery and development of small molecules with the potential for selective toxicity toward tumor cells. This review summarizes the various families of small molecules reported in the literature that have telomeric quadruplex stabilizing properties, and assesses the potential for compounds of this type to be developed as novel anticancer therapies. A future perspective is also presented, emphasizing the need for researchers to adopt approaches that will allow the discovery of molecules with more drug-like properties in order to improve the chances of lead molecules reaching the clinic in the next decade.
Collapse
|
31
|
Abstract
Quadruplex-forming sequences are widely prevalent in human and other genomes, including bacterial ones. These sequences are over-represented in eukaryotic telomeres, promoters, and 5' untranslated regions. They can form quadruplex structures, which may be transient in many situations in normal cells since they can be effectively resolved by helicase action. Mutated helicases in cancer cells are unable to unwind quadruplexes, which are impediments to transcription, translation, or replication, depending on their location within a particular gene. Small molecules that can stabilize quadruplex structures augment these effects and produce cell and proliferation growth inhibition. This article surveys the chemical biology of quadruplexes. It critically examines the major classes of quadruplex-binding small molecules that have been developed to date and the various approaches to discovering selective agents. The challenges of requiring (and achieving) small-molecule targeted selectivity for a particular quadruplex are discussed in relation to the potential of these small molecules as clinically useful therapeutic agents.
Collapse
Affiliation(s)
- Stephen Neidle
- UCL School of Pharmacy, University College London , 29-39 Brunswick Square, London WC1N 1AX, U.K
| |
Collapse
|
32
|
Russo Krauss I, Ramaswamy S, Neidle S, Haider S, Parkinson GN. Structural Insights into the Quadruplex-Duplex 3' Interface Formed from a Telomeric Repeat: A Potential Molecular Target. J Am Chem Soc 2016; 138:1226-33. [PMID: 26730610 DOI: 10.1021/jacs.5b10492] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We report here on an X-ray crystallographic and molecular modeling investigation into the complex 3' interface formed between putative parallel stranded G-quadruplexes and a duplex DNA sequence constructed from the human telomeric repeat sequence TTAGGG. Our crystallographic approach provides a detailed snapshot of a telomeric 3' quadruplex-duplex junction: a junction that appears to have the potential to form a unique molecular target for small molecule binding and interference with telomere-related functions. This unique target is particularly relevant as current high-affinity compounds that bind putative G-quadruplex forming sequences only rarely have a high degree of selectivity for a particular quadruplex. Here DNA junctions were assembled using different putative quadruplex-forming scaffolds linked at the 3' end to a telomeric duplex sequence and annealed to a complementary strand. We successfully generated a series of G-quadruplex-duplex containing crystals, both alone and in the presence of ligands. The structures demonstrate the formation of a parallel folded G-quadruplex and a B-form duplex DNA stacked coaxially. Most strikingly, structural data reveals the consistent formation of a TAT triad platform between the two motifs. This triad allows for a continuous stack of bases to link the quadruplex motif with the duplex region. For these crystal structures formed in the absence of ligands, the TAT triad interface occludes ligand binding at the 3' quadruplex-duplex interface, in agreement with in silico docking predictions. However, with the rearrangement of a single nucleotide, a stable pocket can be produced, thus providing an opportunity for the binding of selective molecules at the interface.
Collapse
Affiliation(s)
- Irene Russo Krauss
- Department of Chemical Sciences, University of Naples Federico II , I-80126 Napoli, Italy
| | - Sneha Ramaswamy
- UCL School of Pharmacy, University College London , London WC1N 1AX, United Kingdom
| | - Stephen Neidle
- UCL School of Pharmacy, University College London , London WC1N 1AX, United Kingdom
| | - Shozeb Haider
- UCL School of Pharmacy, University College London , London WC1N 1AX, United Kingdom
| | - Gary N Parkinson
- UCL School of Pharmacy, University College London , London WC1N 1AX, United Kingdom
| |
Collapse
|
33
|
Castillo-González D, Mergny JL, De Rache A, Pérez-Machado G, Cabrera-Pérez MA, Nicolotti O, Introcaso A, Mangiatordi GF, Guédin A, Bourdoncle A, Garrigues T, Pallardó F, Cordeiro MNDS, Paz-y-Miño C, Tejera E, Borges F, Cruz-Monteagudo M. Harmonization of QSAR Best Practices and Molecular Docking Provides an Efficient Virtual Screening Tool for Discovering New G-Quadruplex Ligands. J Chem Inf Model 2015; 55:2094-110. [DOI: 10.1021/acs.jcim.5b00415] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Daimel Castillo-González
- ARNA Laboratory, IECB, University of Bordeaux, F-33600 Pessac, France
- ARNA Laboratory,
INSERM, U869, F-33000 Bordeaux, France
| | - Jean-Louis Mergny
- ARNA Laboratory, IECB, University of Bordeaux, F-33600 Pessac, France
- ARNA Laboratory,
INSERM, U869, F-33000 Bordeaux, France
| | - Aurore De Rache
- ARNA Laboratory, IECB, University of Bordeaux, F-33600 Pessac, France
- ARNA Laboratory,
INSERM, U869, F-33000 Bordeaux, France
| | - Gisselle Pérez-Machado
- Molecular Simulation and
Drug Design Group, Centro de Bioactivos Químicos (CBQ), Central University of Las Villas, Santa Clara, Villa Clara 54830, Cuba
- Department of Physiology,
Faculty of Medicine, University of Valencia, Valencia 46010, Valencia, Spain
- Department
of Pharmacy and Pharmaceutical Technology, University of Valencia, Burjassot 46100, Valencia, Spain
| | - Miguel Angel Cabrera-Pérez
- Molecular Simulation and
Drug Design Group, Centro de Bioactivos Químicos (CBQ), Central University of Las Villas, Santa Clara, Villa Clara 54830, Cuba
- Department
of Pharmacy and Pharmaceutical Technology, University of Valencia, Burjassot 46100, Valencia, Spain
- Department of Engineering, Area of Pharmacy and Pharmaceutical
Technology, Miguel Hernández University, 03550 Sant Joan d’Alacant, Alicante, Alicante, Spain
| | - Orazio Nicolotti
- Dipartimento
di Farmacia-Scienze, Università degli Studi di Bari “Aldo Moro″, Via Orabona 4, 70125 Bari, Bari, Italy
| | - Antonellina Introcaso
- Dipartimento
di Farmacia-Scienze, Università degli Studi di Bari “Aldo Moro″, Via Orabona 4, 70125 Bari, Bari, Italy
| | - Giuseppe Felice Mangiatordi
- Dipartimento
di Farmacia-Scienze, Università degli Studi di Bari “Aldo Moro″, Via Orabona 4, 70125 Bari, Bari, Italy
| | - Aurore Guédin
- ARNA Laboratory, IECB, University of Bordeaux, F-33600 Pessac, France
- ARNA Laboratory,
INSERM, U869, F-33000 Bordeaux, France
| | - Anne Bourdoncle
- ARNA Laboratory, IECB, University of Bordeaux, F-33600 Pessac, France
- ARNA Laboratory,
INSERM, U869, F-33000 Bordeaux, France
| | - Teresa Garrigues
- Department
of Pharmacy and Pharmaceutical Technology, University of Valencia, Burjassot 46100, Valencia, Spain
| | - Federico Pallardó
- Department of Physiology,
Faculty of Medicine, University of Valencia, Valencia 46010, Valencia, Spain
| | | | - Cesar Paz-y-Miño
- Instituto de Investigaciones
Biomédicas (IIB), Universidad de Las Américas, 170513 Quito, Pichincha, Ecuador
| | - Eduardo Tejera
- Instituto de Investigaciones
Biomédicas (IIB), Universidad de Las Américas, 170513 Quito, Pichincha, Ecuador
| | | | - Maykel Cruz-Monteagudo
- Instituto de Investigaciones
Biomédicas (IIB), Universidad de Las Américas, 170513 Quito, Pichincha, Ecuador
| |
Collapse
|
34
|
Chiorcea-Paquim AM, Rodrigues Pontinha AD, Oliveira-Brett AM. Quadruplex-targeting anticancer drug BRACO-19 voltammetric and AFM characterization. Electrochim Acta 2015. [DOI: 10.1016/j.electacta.2015.05.146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
35
|
Chiorcea-Paquim AM, Pontinha ADR, Eritja R, Lucarelli G, Sparapani S, Neidle S, Oliveira-Brett AM. Atomic Force Microscopy and Voltammetric Investigation of Quadruplex Formation between a Triazole-Acridine Conjugate and Guanine-Containing Repeat DNA Sequences. Anal Chem 2015; 87:6141-9. [DOI: 10.1021/acs.analchem.5b00743] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
| | | | - Ramon Eritja
- Institute for Research in Biomedicine, IQAC−CSIC, CIBER-BBN
Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Barcelona, Spain
| | - Genny Lucarelli
- UCL
School of Pharmacy, University College London, London WC1N 1AX, U.K
| | - Silvia Sparapani
- UCL
School of Pharmacy, University College London, London WC1N 1AX, U.K
| | - Stephen Neidle
- UCL
School of Pharmacy, University College London, London WC1N 1AX, U.K
| | | |
Collapse
|
36
|
Collie GW, Campbell NH, Neidle S. Loop flexibility in human telomeric quadruplex small-molecule complexes. Nucleic Acids Res 2015; 43:4785-99. [PMID: 25940631 PMCID: PMC4446451 DOI: 10.1093/nar/gkv427] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 04/20/2015] [Indexed: 01/25/2023] Open
Abstract
Quadruplex nucleic acids can be formed at the ends of eukaryotic chromosomes. Their formation and stabilisation by appropriate small molecules can be used as a means of inhibiting the telomere maintenance functions of telomerase in human cancer cells. The crystal structures have been determined for a number of complexes between these small molecules and human telomeric DNA and RNA quadruplexes. The detailed structural characteristics of these complexes have been surveyed here and the variations in conformation for the TTA and UUA loops have been explored. Loop conformations have been classified in terms of a number of discrete types and their distribution among the crystal structures. Sugar conformation and backbone angles have also been examined and trends highlighted. One particular loop class has been found to be most prevalent. Implications for in particular, rational drug design, are discussed.
Collapse
Affiliation(s)
- Gavin W Collie
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Nancy H Campbell
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Stephen Neidle
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| |
Collapse
|
37
|
Sabater L, Nicolau-Travers ML, De Rache A, Prado E, Dejeu J, Bombarde O, Lacroix J, Calsou P, Defrancq E, Mergny JL, Gomez D, Pratviel G. The nickel(II) complex of guanidinium phenyl porphyrin, a specific G-quadruplex ligand, targets telomeres and leads to POT1 mislocalization in culture cells. J Biol Inorg Chem 2015; 20:729-38. [DOI: 10.1007/s00775-015-1260-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 03/22/2015] [Indexed: 10/23/2022]
|
38
|
|
39
|
Siters KE, Fountain MA, Morrow JR. Selective binding of Zn2+ complexes to human telomeric G-quadruplex DNA. Inorg Chem 2014; 53:11540-51. [PMID: 25310175 DOI: 10.1021/ic501484p] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Zn(2+) complex of 5-(1,4,7,10-tetraazacyclododecan-1-ylsulfonyl)-N,N-dimethylnaphthalen-1-amine, Zn(DSC), binds selectively to the biologically relevant human telomeric (H-Telo) G-quadruplex. An increase in the Zn(DSC) dansyl group fluorescence with a simultaneous shift in emission is consistent with the complex binding to H-Telo. The H-Telo G-quadruplex has two binding sites for Zn(DSC) with binding constants in the low micromolar range (2.5 μM). Isothermal calorimetric titrations confirm low micromolar dissociation constants with a 2:1 stoichiometry. The interaction between H-Telo and Zn(DSC) is highly pH-dependent, consistent with binding to the unpaired thymines in the G-quadruplex loops. As a result, Zn(DSC) selectively binds to H-Telo over duplex DNA. In contrast to Zn(2+), Fe(2+) and Co(2+) do not complex to the DSC macrocycle appreciably under the conditions of the experiment. The Cu(2+) complex of DSC does not interact measurably with the H-Telo G-quadruplex. Interestingly, the H-Telo-Zn(DSC) adduct self-assembles from its individual components at physiological pH and 100 mM KCl. The self-assembly feature, which is specific for the Zn(2+) ion, suggests that this system may be viable as a Zn(2+) sensor. Pentanucleotides were studied in order to better describe the binding of Zn(DSC) to thymine sequences. NMR studies were consistent with the binding of Zn(DSC) to thymine-containing oligonucleotides including CCTCC, CTTCC, and CTCTC. Studies showed that the dansyl group of Zn(DSC) interacts with thymines in CTTCC. Fluorescence spectroscopy and ITC data indicate that Zn(DSC) forms 2:1 adducts with thymines that are spaced (CTCTC) but not tandem thymines (CTTCC). These data are consistent with one Zn(DSC) complex binding to two separate loops in the G-quadruplex. A second Zn(2+) complex containing an acridine pendent, Zn(ACR), binds tightly to pentanucleotides with both tandem and spaced thymines. Zn(ACR) indiscriminately binds to both H-Telo and duplex DNA.
Collapse
Affiliation(s)
- Kevin E Siters
- Department of Chemistry, University at Buffalo, State University of New York , Buffalo, New York 14260, United States
| | | | | |
Collapse
|
40
|
Pontinha ADR, Lombardo CM, Neidle S, Oliveira-Brett AM. Triazole-linked phenyl derivatives: redox mechanisms and in situ electrochemical evaluation of interaction with dsDNA. Bioelectrochemistry 2014; 101:97-105. [PMID: 25194950 DOI: 10.1016/j.bioelechem.2014.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/08/2014] [Accepted: 08/13/2014] [Indexed: 11/28/2022]
Abstract
The redox mechanism of two trisubstituted triazole-linked phenyl derivatives (CL41 and CL42) and a disubstituted triazole-linked phenyl derivative (CL2r50) were studied using cyclic, differential pulse and square wave voltammetry at a glassy carbon electrode. The CL41, CL42 and CL2r50 oxidation is a complex, pH-dependent irreversible process involving the formation of electroactive products that undergo two consecutive reversible oxidation reactions. The DNA interaction with CL41, CL42 and CL2r50 was investigated by differential pulse voltammetry using the dsDNA-electrochemical biosensor and in DNA/trisubstituted triazole incubated solutions. All three trisubstituted triazole-linked phenyl derivatives interacted with dsDNA causing morphological and oxidative damage to the dsDNA structure in a time-dependent manner. The DNA-electrochemical biosensor enabled the detection of oxidative damage to DNA following the occurrence of the 8-oxoGua and/or 2,8-oxoAde oxidation peaks.
Collapse
Affiliation(s)
- A Dora R Pontinha
- Department of Chemistry, University of Coimbra, Coimbra 3004-535, Portugal
| | | | - Stephen Neidle
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK
| | | |
Collapse
|
41
|
Ungvarsky J, Plsikova J, Janovec L, Koval J, Mikes J, Mikesová L, Harvanova D, Fedorocko P, Kristian P, Kasparkova J, Brabec V, Vojtickova M, Sabolova D, Stramova Z, Rosocha J, Imrich J, Kozurkova M. Novel trisubstituted acridines as human telomeric quadruplex binding ligands. Bioorg Chem 2014; 57:13-29. [PMID: 25171773 DOI: 10.1016/j.bioorg.2014.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/25/2014] [Accepted: 07/27/2014] [Indexed: 11/24/2022]
Abstract
A novel series of trisubstituted acridines were synthesized with the aim of mimicking the effects of BRACO19. These compounds were synthesized by modifying the molecular structure of BRACO19 at positions 3 and 6 with heteroacyclic moieties. All of the derivatives presented in the study exhibited stabilizing effects on the human telomeric DNA quadruplex. UV-vis spectroscopy, circular dichroism, linear dichroism and viscosimetry were used in order to study the nature of the DNA binding in more detail. The results show that all of the novel derivatives were able to fold the single-stranded DNA sequences into antiparallel G-quadruplex structures, with derivative 15 exhibiting the highest stabilizing capability. Cell cycle analysis revealed that a primary trend of the "braco"-like derivatives was to arrest the cells in the S- and G2M-phases of the cell cycle within the first 72h, with derivative 13 and BRACO19 proving particularly effective in suppressing cell proliferation. All studies derivatives were less toxic to human fibroblast cell line in comparison with HT 29 cancer cell line.
Collapse
Affiliation(s)
- Jan Ungvarsky
- Department of Organic Chemistry, Moyzesova 11, 04001 Kosice, Slovak Republic
| | - Jana Plsikova
- Department of Biochemistry, Moyzesova 11, 04001 Kosice, Slovak Republic; Associated Tissue Bank of Faculty of Medicine, L. Pasteur University Hospital, Trieda SNP 1, 04166 Kosice, Slovak Republic
| | - Ladislav Janovec
- Department of Organic Chemistry, Moyzesova 11, 04001 Kosice, Slovak Republic
| | - Jan Koval
- Department of Cellular Biology, Moyzesova 11, 04001 Kosice, Slovak Republic
| | - Jaromir Mikes
- Department of Cellular Biology, Moyzesova 11, 04001 Kosice, Slovak Republic
| | - Lucia Mikesová
- Department of Cellular Biology, Moyzesova 11, 04001 Kosice, Slovak Republic
| | - Denisa Harvanova
- Associated Tissue Bank of Faculty of Medicine, L. Pasteur University Hospital, Trieda SNP 1, 04166 Kosice, Slovak Republic
| | - Peter Fedorocko
- Department of Cellular Biology, Moyzesova 11, 04001 Kosice, Slovak Republic
| | - Pavol Kristian
- Department of Organic Chemistry, Moyzesova 11, 04001 Kosice, Slovak Republic
| | - Jana Kasparkova
- Institute of Biophysics, Department of Molecular Biophysics and Pharmacology, Academy of Sciences of the Czech Republic, Brno, Czech Republic
| | - Viktor Brabec
- Institute of Biophysics, Department of Molecular Biophysics and Pharmacology, Academy of Sciences of the Czech Republic, Brno, Czech Republic
| | - Maria Vojtickova
- Department of Organic Chemistry, Moyzesova 11, 04001 Kosice, Slovak Republic
| | - Danica Sabolova
- Department of Biochemistry, Moyzesova 11, 04001 Kosice, Slovak Republic
| | - Zuzana Stramova
- Department of Biochemistry, Moyzesova 11, 04001 Kosice, Slovak Republic
| | - Jan Rosocha
- Associated Tissue Bank of Faculty of Medicine, L. Pasteur University Hospital, Trieda SNP 1, 04166 Kosice, Slovak Republic
| | - Jan Imrich
- Department of Organic Chemistry, Moyzesova 11, 04001 Kosice, Slovak Republic
| | - Maria Kozurkova
- Department of Biochemistry, Moyzesova 11, 04001 Kosice, Slovak Republic.
| |
Collapse
|
42
|
Perrone R, Butovskaya E, Daelemans D, Palù G, Pannecouque C, Richter SN. Anti-HIV-1 activity of the G-quadruplex ligand BRACO-19. J Antimicrob Chemother 2014; 69:3248-58. [PMID: 25103489 DOI: 10.1093/jac/dku280] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES A dynamic G-quadruplex region has been previously shown to form in the long terminal repeat (LTR) promoter of the HIV-1 integrated DNA genome. Inhibition of promoter activity and antiviral effects have been observed when this region was stabilized by BRACO-19, a trisubstituted acridine derivative that binds G-quadruplexes. Here, we aimed at characterizing the antiviral mechanism of action of BRACO-19 by analysing its activity towards a broad range of HIV-1 strains, host cells and infection modes. METHODS The antiviral activity of BRACO-19 in cell lines and primary cells infected or persistently infected by HIV-1 strains was evaluated at different times post-infection. Virucidal, viral binding, time-dependent and drug-dependent assays were performed to identify the viral target step. Circular dichroism, UV spectroscopy and a reverse transcriptase (RT) stop assay were used to assess RNA G-quadruplex folding and inhibition of RT processing. RESULTS Thorough virological assays demonstrated that BRACO-19 acts both at the reverse transcription and the post-integration level during the virus life cycle. This behaviour was rationalized by the observation that a G-quadruplex-forming sequence identical to that of the LTR DNA is present at the 3'-end of the virus RNA genome. Biophysics and biomolecular testing showed that this region has the ability to fold into very stable G-quadruplex structures that are even more stabilized by BRACO-19, therefore inhibiting the reverse transcription process at the template level. CONCLUSIONS Our findings strongly support the activity of BRACO-19 at the viral G-quadruplex level and therefore strengthen the use of viral G-quadruplexes as new anti-HIV-1 targets.
Collapse
Affiliation(s)
- Rosalba Perrone
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Elena Butovskaya
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Dirk Daelemans
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Sara N Richter
- Department of Molecular Medicine, University of Padua, Padua, Italy
| |
Collapse
|
43
|
Müller S, Rodriguez R. G-quadruplex interacting small molecules and drugs: from bench toward bedside. Expert Rev Clin Pharmacol 2014; 7:663-79. [DOI: 10.1586/17512433.2014.945909] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
44
|
Small-molecule quadruplex-targeted drug discovery. Bioorg Med Chem Lett 2014; 24:2602-12. [DOI: 10.1016/j.bmcl.2014.04.029] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/06/2014] [Accepted: 04/08/2014] [Indexed: 01/24/2023]
|
45
|
|
46
|
Shalaby T, Fiaschetti G, Nagasawa K, Shin-ya K, Baumgartner M, Grotzer M. G-quadruplexes as potential therapeutic targets for embryonal tumors. Molecules 2013; 18:12500-37. [PMID: 24152672 PMCID: PMC6269990 DOI: 10.3390/molecules181012500] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 09/18/2013] [Accepted: 09/25/2013] [Indexed: 12/27/2022] Open
Abstract
Embryonal tumors include a heterogeneous group of highly malignant neoplasms that primarily affect infants and children and are characterized by a high rate of mortality and treatment-related morbidity, hence improved therapies are clearly needed. G-quadruplexes are special secondary structures adopted in guanine (G)-rich DNA sequences that are often present in biologically important regions, e.g. at the end of telomeres and in the regulatory regions of oncogenes such as MYC. Owing to the significant roles that both telomeres and MYC play in cancer cell biology, G-quadruplexes have been viewed as emerging therapeutic targets in oncology and as tools for novel anticancer drug design. Several compounds that target these structures have shown promising anticancer activity in tumor xenograft models and some of them have entered Phase II clinical trials. In this review we examine approaches to DNA targeted cancer therapy, summarize the recent developments of G-quadruplex ligands as anticancer drugs and speculate on the future direction of such structures as a potential novel therapeutic strategy for embryonal tumors of the nervous system.
Collapse
Affiliation(s)
- Tarek Shalaby
- Division of Oncology, University Children's Hospital of Zurich, Zurich 8032, Switzerland.
| | | | | | | | | | | |
Collapse
|
47
|
Molecular basis of recognition of quadruplexes human telomere and c-myc promoter by the putative anticancer agent sanguinarine. Biochim Biophys Acta Gen Subj 2013; 1830:4189-201. [DOI: 10.1016/j.bbagen.2013.03.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/19/2013] [Accepted: 03/26/2013] [Indexed: 01/24/2023]
|
48
|
Ghosh S, Kar A, Chowdhury S, Dasgupta D. Ellipticine binds to a human telomere sequence: an additional mode of action as a putative anticancer agent? Biochemistry 2013; 52:4127-37. [PMID: 23697684 DOI: 10.1021/bi400080t] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Polyguanine sequences fold into G-quadruplex structures in the presence of monovalent cations. It is accepted that the telomeric DNA region consists of G-quadruplex structure. There are reports that potential G-quadruplex forming motifs are also present in the promoter region of some proto-oncogenes such as c-myc, c-kit, KRAS, etc. Small molecules with the potential to stabilize the telomeric DNA quadruplex have emerged as potential anticancer agents. We have studied the interaction of ellipticine, a putative anticancer agent from a plant source, with a human telomeric DNA sequence (H24). Spectroscopic and calorimetric studies indicate that the association of ellipticine with H24 is an entropically driven phenomenon with a 2:3 (H24:ellipticine) stoichiometry. Though ellipticine binding does not induce any major structural perturbation in H24, the association leads to formation of a complex with enhanced thermal stability. An assay with the telomerase repeat amplification protocol shows that ellipticine inhibits telomerase activity in MDAMB-231 breast cancer cell line extracts. This is the first report of the quadruplex binding ability of ellipticine. Using the results, we propose that along with DNA intercalation and/or topoisomerase II inhibition, interaction with the telomeric DNA region and the resultant inhibition of telomerase activity might be an additional mode of action for its anticancer property.
Collapse
Affiliation(s)
- Saptaparni Ghosh
- Biophysics Division, Saha Institute of Nuclear Physics , Block-AF, Sector- I, Bidhan Nagar, Kolkata 700064, India
| | | | | | | |
Collapse
|
49
|
Hirt BV, Wattis JAD, Preston SP. Modelling the regulation of telomere length: the effects of telomerase and G-quadruplex stabilising drugs. J Math Biol 2013; 68:1521-52. [PMID: 23620229 PMCID: PMC3975128 DOI: 10.1007/s00285-013-0678-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 03/20/2013] [Indexed: 12/13/2022]
Abstract
Telomeres are guanine-rich sequences at the end of chromosomes which shorten during each replication event and trigger cell cycle arrest and/or controlled death (apoptosis) when reaching a threshold length. The enzyme telomerase replenishes the ends of telomeres and thus prolongs the life span of cells, but also causes cellular immortalisation in human cancer. G-quadruplex (G4) stabilising drugs are a potential anticancer treatment which work by changing the molecular structure of telomeres to inhibit the activity of telomerase. We investigate the dynamics of telomere length in different conformational states, namely t-loops, G-quadruplex structures and those being elongated by telomerase. By formulating deterministic differential equation models we study the effects of various levels of both telomerase and concentrations of a G4-stabilising drug on the distribution of telomere lengths, and analyse how these effects evolve over large numbers of cell generations. As well as calculating numerical solutions, we use quasicontinuum methods to approximate the behaviour of the system over time, and predict the shape of the telomere length distribution. We find those telomerase and G4-concentrations where telomere length maintenance is successfully regulated. Excessively high levels of telomerase lead to continuous telomere lengthening, whereas large concentrations of the drug lead to progressive telomere erosion. Furthermore, our models predict a positively skewed distribution of telomere lengths, that is, telomeres accumulate over lengths shorter than the mean telomere length at equilibrium. Our model results for telomere length distributions of telomerase-positive cells in drug-free assays are in good agreement with the limited amount of experimental data available.
Collapse
Affiliation(s)
- Bartholomäus V Hirt
- School of Mathematical Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK,
| | | | | |
Collapse
|
50
|
Micco M, Collie GW, Dale AG, Ohnmacht SA, Pazitna I, Gunaratnam M, Reszka AP, Neidle S. Structure-based design and evaluation of naphthalene diimide G-quadruplex ligands as telomere targeting agents in pancreatic cancer cells. J Med Chem 2013; 56:2959-74. [PMID: 23514618 DOI: 10.1021/jm301899y] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tetra-substituted naphthalene diimide (ND) derivatives with positively charged termini are potent stabilizers of human telomeric and gene promoter DNA quadruplexes and inhibit the growth of human cancer cells in vitro and in vivo. The present study reports the enhancement of the pharmacological properties of earlier ND compounds using structure-based design. Crystal structures of three complexes with human telomeric intramolecular quadruplexes demonstrate that two of the four strongly basic N-methyl-piperazine groups can be replaced by less basic morpholine groups with no loss of intermolecular interactions in the grooves of the quadruplex. The new compounds retain high affinity to human telomeric quadruplex DNA but are 10-fold more potent against the MIA PaCa-2 pancreatic cancer cell line, with IC50 values of ~10 nM. The lead compound induces cellular senescence but does not inhibit telomerase activity at the nanomolar dosage levels required for inhibition of cellular proliferation. Gene array qPCR analysis of MIA PaCa-2 cells treated with the lead compound revealed significant dose-dependent modulation of a distinct subset of genes, including strong induction of DNA damage responsive genes CDKN1A, DDIT3, GADD45A/G, and PPM1D, and repression of genes involved in telomere maintenance, including hPOT1 and PARP1.
Collapse
Affiliation(s)
- Marialuisa Micco
- The School of Pharmacy, University College London, London WC1N 1AX, UK
| | | | | | | | | | | | | | | |
Collapse
|