1
|
Reyes Y, Larrey EK, Pathak R, Veisaga ML, Barbieri MA, Ward S, Kumar A, Sevilla MD, Adhikary A, Wnuk SF. Azido derivatives of sesquiterpene lactones: Synthesis, anticancer proliferation, and chemistry of nitrogen-centered radicals. RESULTS IN CHEMISTRY 2024; 9:101643. [PMID: 39498431 PMCID: PMC11533910 DOI: 10.1016/j.rechem.2024.101643] [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] [Indexed: 11/07/2024] Open
Abstract
Sesquiterpene lactones (SLs) such as parthenolide (PTL) and dehydroleucodine (DhL) selectively kill cancer cells without exerting normal tissue toxicity, potentially due to presence of α-methylene-γ-lactone (αMγL) fragment. We hypothesize that the addition of an azido group to the αMγL fragment of PTL or DhL further augments their anticancer properties as well as radiation sensitivity of cancer cells. Azido-SLs containing the azido group at the C14 methyl position of PTL (i.e., azido-melampomagnolide B, AzMMB) while preserving the mechanistically crucial exomethylene unit of αMγL fragment were also prepared. Sham-irradiated (i.e., unirradiated control) or irradiated human breast cancer cells (MCF7) were treated with different concentrations of azido-PTL (AzPTL) or azido-DhL (AzDhL) along with parental SLs. Proliferation rate of MCF7 cells were measured by MTT-assay, and their colony forming ability was determined by colony formation assay. Both AzPTL and AzDhL significantly suppress proliferation rate and colony forming ability of MCF-7 cells. AzPTL suppressed colony forming ability, not cellular proliferation, following irradiation to a greater extent than PTL at lower concentrations (5 and 10 μM). Electron spin resonance (ESR) studies were performed employing gamma-irradiated homogeneous supercooled aqueous solutions to investigate radical formation through addition of radiation-mediated prehydrated electrons to the azide group of AzPTL and AzDhL and to follow subsequent reactions of these radicals. In AzPTL, formation of a tertiary carbon-centered radical plausibly via a metastable aminyl radical was observed, whereas AzDhL produced both π-aminyl and α-azidoalkyl radicals. These radicals may contribute to the antitumor activities of AzPTL and AzDhL.
Collapse
Affiliation(s)
- Yahaira Reyes
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, USA
| | - Enoch K. Larrey
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA
| | - Rupak Pathak
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA
| | - Maria L. Veisaga
- Department of Biological Sciences, Florida International University, Miami, Florida 33199, USA
| | - Manuel A. Barbieri
- Department of Biological Sciences, Florida International University, Miami, Florida 33199, USA
| | - Samuel Ward
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, USA
| | - Anil Kumar
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, USA
| | - Michael D. Sevilla
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, USA
| | - Amitava Adhikary
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, USA
| | - Stanislaw F. Wnuk
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, USA
| |
Collapse
|
2
|
Reyes Y, Adhikary A, Wnuk SF. Nitrogen-Centered Radicals Derived from Azidonucleosides. Molecules 2024; 29:2310. [PMID: 38792171 PMCID: PMC11124349 DOI: 10.3390/molecules29102310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Azido-modified nucleosides have been extensively explored as substrates for click chemistry and the metabolic labeling of DNA and RNA. These compounds are also of interest as precursors for further synthetic elaboration and as therapeutic agents. This review discusses the chemistry of azidonucleosides related to the generation of nitrogen-centered radicals (NCRs) from the azido groups that are selectively inserted into the nucleoside frame along with the subsequent chemistry and biological implications of NCRs. For instance, the critical role of the sulfinylimine radical generated during inhibition of ribonucleotide reductases by 2'-azido-2'-deoxy pyrimidine nucleotides as well as the NCRs generated from azidonucleosides by radiation-produced (prehydrated and aqueous) electrons are discussed. Regio and stereoselectivity of incorporation of an azido group ("radical arm") into the frame of nucleoside and selective generation of NCRs under reductive conditions, which often produce the same radical species that are observed upon ionization events due to radiation and/or other oxidative conditions that are emphasized. NCRs generated from nucleoside-modified precursors other than azidonucleosides are also discussed but only with the direct relation to the same/similar NCRs derived from azidonucleosides.
Collapse
Affiliation(s)
- Yahaira Reyes
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL 33199, USA;
| | - Amitava Adhikary
- Department of Chemistry, Oakland University, Rochester, MI 48309, USA;
| | - Stanislaw F. Wnuk
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL 33199, USA;
| |
Collapse
|
3
|
Adjei D, Reyes Y, Kumar A, Ward S, Denisov SA, Alahmadi M, Sevilla MD, Wnuk SF, Mostafavi M, Adhikary A. Pathways of the Dissociative Electron Attachment Observed in 5- and 6-Azidomethyluracil Nucleosides: Nitrogen (N 2) Elimination vs Azide Anion (N 3-) Elimination. J Phys Chem B 2023; 127:1563-1571. [PMID: 36780335 PMCID: PMC9984991 DOI: 10.1021/acs.jpcb.2c08257] [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] [Indexed: 02/14/2023]
Abstract
5-Azidomethyl-2'-deoxyuridine (5-AmdU, 1) has been successfully employed for the metabolic labeling of DNA and fluorescent imaging of live cells. 5-AmdU also demonstrated significant radiosensitization in breast cancer cells via site-specific nitrogen-centered radical (π-aminyl (U-5-CH2-NH•), 2, and σ-iminyl (U-5-CH═N•), 3) formation. This work shows that these nitrogen-centered radicals are not formed via the reduction of the azido group in 6-azidomethyluridine (6-AmU, 4). Radical assignments were performed using electron spin resonance (ESR) in supercooled solutions, pulse radiolysis in aqueous solutions, and theoretical (DFT) calculations. Radiation-produced electron addition to 4 leads to the facile N3- loss, forming a stable neutral C-centered allylic radical (U-6-CH2•, 5) through dissociative electron attachment (DEA) via the transient negative ion, TNI (U-6-CH2-N3•-), in agreement with DFT calculations. In contrast, TNI (U-5-CH2-N3•-) of 1, via facile N2 loss (DEA) and protonation from the surrounding water, forms radical 2. Subsequently, 2 undergoes rapid H-atom abstraction from 1 and produces the metastable intermediate α-azidoalkyl radical (U-5-CH•-N3). U-5-CH•-N3 converts facilely to radical 3. N3- loss from U-6-CH2-N3•- is thermodynamically controlled, whereas N2 loss from U-5-CH2-N3•- is dictated by protonation from the surrounding waters and resonance conjugation of the azidomethyl side chain at C5 with the pyrimidine ring.
Collapse
Affiliation(s)
- Daniel Adjei
- Institut de Chimie Physique, UMR 8000 CNRS, Bât. 349, Université Paris-Saclay; 91405, Orsay, Cedex, France
| | - Yahaira Reyes
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, USA
| | - Anil Kumar
- Department of Chemistry, 146 Library Drive, Oakland University, Rochester, Michigan 48309, USA
| | - Samuel Ward
- Department of Chemistry, 146 Library Drive, Oakland University, Rochester, Michigan 48309, USA
| | - Sergey A. Denisov
- Institut de Chimie Physique, UMR 8000 CNRS, Bât. 349, Université Paris-Saclay; 91405, Orsay, Cedex, France
| | - Moaadh Alahmadi
- Department of Chemistry, 146 Library Drive, Oakland University, Rochester, Michigan 48309, USA
| | - Michael D. Sevilla
- Department of Chemistry, 146 Library Drive, Oakland University, Rochester, Michigan 48309, USA
| | - Stanislaw F. Wnuk
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, USA
| | - Mehran Mostafavi
- Institut de Chimie Physique, UMR 8000 CNRS, Bât. 349, Université Paris-Saclay; 91405, Orsay, Cedex, France
| | - Amitava Adhikary
- Department of Chemistry, 146 Library Drive, Oakland University, Rochester, Michigan 48309, USA
| |
Collapse
|
4
|
Liu Z, Lian X, Zhang X, Zhu Y, Zhang W, Wang J, Wang H, Liu B, Ren Z, Zhang M, Liu M, Gao Y. ESPL1 Is a Novel Prognostic Biomarker Associated With the Malignant Features of Glioma. Front Genet 2021; 12:666106. [PMID: 34512713 PMCID: PMC8428966 DOI: 10.3389/fgene.2021.666106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
Research has confirmed that extra spindle pole bodies-like 1 (ESPL1), an etiological factor, promotes the malignant progression of cancers. However, the relationship between ESPL1 and glioma has not yet been demonstrated. The purpose of this study was to reveal the potential mechanisms of ESPL1-mediated malignant glioma progression. Gene expression data and detailed clinical information of glioma cases were obtained from multiple public databases. Subsequently, a series of bioinformatics analyses were used to elucidate the effects of ESPL1 on glioma. The results demonstrated that the mRNA and protein levels of ESPL1 in glioma were higher than those in normal brain tissues. In addition, ESPL1 expression was considerably associated with the clinical and pathological features of gliomas, such as World Health Organization grade, histology, and 1p19q co-deletion status. Importantly, ESPL1 reduced the overall survival (OS) of glioma patients and had prognostic value for gliomas. Gene set enrichment analysis (GSEA) indirectly revealed that ESPL1 regulates the activation of cancer-related pathways, such as the cell cycle and base excision repair pathways. In addition, we used the Connectivity Map (CMap) database to screen three molecular drugs that inhibit ESPL1: thioguanosine, antimycin A, and zidovudine. Finally, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression levels of ESPL1 in glioma cell lines. This study plays an important role in revealing the etiology of glioma by revealing the function of ESPL1, providing a potential molecular marker for the diagnosis and treatment of glioma, especially low-grade glioma.
Collapse
Affiliation(s)
- Zhendong Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China.,Department of Surgery of Spine and Spinal Cord, Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Xiaoyu Lian
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xiuru Zhang
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Yongjie Zhu
- Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Wang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jialin Wang
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Hongbo Wang
- Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Binfeng Liu
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhishuai Ren
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Mengjun Zhang
- Harbin Medical University Cancer Hospital, Heilongjiang Provincial Cancer Hospital, Harbin, China
| | - Mingyang Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China.,Department of Surgery of Spine and Spinal Cord, Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
5
|
García-Trejo JJ, Ortega R, Zarco-Zavala M. Putative Repurposing of Lamivudine, a Nucleoside/Nucleotide Analogue and Antiretroviral to Improve the Outcome of Cancer and COVID-19 Patients. Front Oncol 2021; 11:664794. [PMID: 34367956 PMCID: PMC8335563 DOI: 10.3389/fonc.2021.664794] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022] Open
Abstract
Lamivudine, also widely known as 3TC belongs to a family of nucleotide/nucleoside analogues of cytidine or cytosine that inhibits the Reverse Transcriptase (RT) of retroviruses such as HIV. Lamivudine is currently indicated in combination with other antiretroviral agents for the treatment of HIV-1 infection or for chronic Hepatitis B (HBV) virus infection associated with evidence of hepatitis B viral replication and active liver inflammation. HBV reactivation in patients with HBV infections who receive anticancer chemotherapy can be a life-threatening complication during and after the completion of chemotherapy. Lamivudine is used, as well as other antiretrovirals, to prevent the reactivation of the Hepatitis B virus during and after chemotherapy. In addition, Lamivudine has been shown to sensitize cancer cells to chemotherapy. Lamivudine and other similar analogues also have direct positive effects in the prevention of cancer in hepatitis B or HIV positive patients, independently of chemotherapy or radiotherapy. Recently, it has been proposed that Lamivudine might be also repurposed against SARS-CoV-2 in the context of the COVID-19 pandemic. In this review we first examine recent reports on the re-usage of Lamivudine or 3TC against the SARS-CoV-2, and we present docking evidence carried out in silico suggesting that Lamivudine may bind and possibly work as an inhibitor of the SARS-CoV-2 RdRp RNA polymerase. We also evaluate and propose assessment of repurposing Lamivudine as anti-SARS-CoV-2 and anti-COVID-19 antiviral. Secondly, we summarize the published literature on the use of Lamivudine or (3TC) before or during chemotherapy to prevent reactivation of HBV, and examine reports of enhanced effectiveness of radiotherapy in combination with Lamivudine treatment against the cancerous cells or tissues. We show that the anti-cancer properties of Lamivudine are well established, whereas its putative anti-COVID effect is under investigation. The side effects of lamivudine and the appearance of resistance to 3TC are also discussed.
Collapse
Affiliation(s)
- José J García-Trejo
- Department of Biology, Laboratory of Bioenergetics, Chemistry Faculty and School, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Raquel Ortega
- Department of Biology, Laboratory of Bioenergetics, Chemistry Faculty and School, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Mariel Zarco-Zavala
- Department of Biology, Laboratory of Bioenergetics, Chemistry Faculty and School, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| |
Collapse
|
6
|
Repurposing of Antimicrobial Agents for Cancer Therapy: What Do We Know? Cancers (Basel) 2021; 13:cancers13133193. [PMID: 34206772 PMCID: PMC8269327 DOI: 10.3390/cancers13133193] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
The substantial costs of clinical trials, the lengthy timelines of new drug discovery and development, along the high attrition rates underscore the need for alternative strategies for finding quickly suitable therapeutics agents. Given that most approved drugs possess more than one target tightly linked to other diseases, it encourages promptly testing these drugs in patients. Over the past decades, this has led to considerable attention for drug repurposing, which relies on identifying new uses for approved or investigational drugs outside the scope of the original medical indication. The known safety of approved drugs minimizes the possibility of failure for adverse toxicology, making them attractive de-risked compounds for new applications with potentially lower overall development costs and shorter development timelines. This latter case is an exciting opportunity, specifically in oncology, due to increased resistance towards the current therapies. Indeed, a large body of evidence shows that a wealth of non-cancer drugs has beneficial effects against cancer. Interestingly, 335 drugs are currently being evaluated in different clinical trials for their potential activities against various cancers (Redo database). This review aims to provide an extensive discussion about the anti-cancer activities exerted by antimicrobial agents and presents information about their mechanism(s) of action and stage of development/evaluation.
Collapse
|
7
|
Mudgal M, Dang TP, Sobczak AJ, Lumpuy DA, Dutta P, Ward S, Ward K, Alahmadi M, Kumar A, Sevilla MD, Wnuk SF, Adhikary A. Site of Azido Substitution in the Sugar Moiety of Azidopyrimidine Nucleosides Influences the Reactivity of Aminyl Radicals Formed by Dissociative Electron Attachment. J Phys Chem B 2020; 124:11357-11370. [PMID: 33270461 DOI: 10.1021/acs.jpcb.0c08201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this work, electron-induced site-specific formation of neutral π-type aminyl radicals (RNH·) and their reactions with pyrimidine nucleoside analogs azidolabeled at various positions in the sugar moiety, e.g., at 2'-, 3'-, 4'-, and 5'- sites along with a model compound 3-azido-1-propanol (3AZPrOH), were investigated. Electron paramagnetic resonance (EPR) studies confirmed the site and mechanism of RNH· formation via dissociative electron attachment-mediated loss of N2 and subsequent facile protonation from the solvent employing the 15N-labeled azido group, deuterations at specific sites in the sugar and base, and changing the solvent from H2O to D2O. Reactions of RNH· were investigated employing EPR by warming these samples from 77 K to ca. 170 K. RNH· at a primary carbon site (5'-azido-2',5'-dideoxyuridine, 3AZPrOH) facilely converted to a σ-type iminyl radical (R═N·) via a bimolecular H-atom abstraction forming an α-azidoalkyl radical. RNH· when at a secondary carbon site (e.g., 2'-azido-2'-deoxyuridine) underwent bimolecular electrophilic addition to the C5═C6 double bond of a proximate pyrimidine base. Finally, RNH· at tertiary alkyl carbon (4'-azidocytidine) underwent little reaction. These results show the influence of the stereochemical and electronic environment on RNH· reactivity and allow the selection of those azidonucleosides that would be most effective in augmenting cellular radiation damage.
Collapse
Affiliation(s)
- Mukesh Mudgal
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Thao P Dang
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Adam J Sobczak
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Daniel A Lumpuy
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Priya Dutta
- Department of Chemistry, Oakland University, 146 Library Drive, Rochester, Michigan 48309, United States
| | - Samuel Ward
- Department of Chemistry, Oakland University, 146 Library Drive, Rochester, Michigan 48309, United States
| | - Katherine Ward
- Department of Chemistry, Oakland University, 146 Library Drive, Rochester, Michigan 48309, United States
| | - Moaadh Alahmadi
- Department of Chemistry, Oakland University, 146 Library Drive, Rochester, Michigan 48309, United States
| | - Anil Kumar
- Department of Chemistry, Oakland University, 146 Library Drive, Rochester, Michigan 48309, United States
| | - Michael D Sevilla
- Department of Chemistry, Oakland University, 146 Library Drive, Rochester, Michigan 48309, United States
| | - Stanislaw F Wnuk
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Amitava Adhikary
- Department of Chemistry, Oakland University, 146 Library Drive, Rochester, Michigan 48309, United States
| |
Collapse
|
8
|
Kumar A, Becker D, Adhikary A, Sevilla MD. Reaction of Electrons with DNA: Radiation Damage to Radiosensitization. Int J Mol Sci 2019; 20:E3998. [PMID: 31426385 PMCID: PMC6720166 DOI: 10.3390/ijms20163998] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/01/2019] [Accepted: 08/12/2019] [Indexed: 01/19/2023] Open
Abstract
This review article provides a concise overview of electron involvement in DNA radiation damage. The review begins with the various states of radiation-produced electrons: Secondary electrons (SE), low energy electrons (LEE), electrons at near zero kinetic energy in water (quasi-free electrons, (e-qf)) electrons in the process of solvation in water (presolvated electrons, e-pre), and fully solvated electrons (e-aq). A current summary of the structure of e-aq, and its reactions with DNA-model systems is presented. Theoretical works on reduction potentials of DNA-bases were found to be in agreement with experiments. This review points out the proposed role of LEE-induced frank DNA-strand breaks in ion-beam irradiated DNA. The final section presents radiation-produced electron-mediated site-specific formation of oxidative neutral aminyl radicals from azidonucleosides and the evidence of radiosensitization provided by these aminyl radicals in azidonucleoside-incorporated breast cancer cells.
Collapse
Affiliation(s)
- Anil Kumar
- Department of Chemistry, Oakland University, Rochester, MI 48309, USA
| | - David Becker
- Department of Chemistry, Oakland University, Rochester, MI 48309, USA
| | - Amitava Adhikary
- Department of Chemistry, Oakland University, Rochester, MI 48309, USA
| | - Michael D Sevilla
- Department of Chemistry, Oakland University, Rochester, MI 48309, USA.
| |
Collapse
|
9
|
Savaloni H, Goli-Haghighi S, Babaei R. Application of Mn-Cu Helical Star-Shaped (Pine-Tree-Like) Sculpted Thin Films with Different Symmetries Using Surface-Enhanced Raman Spectroscopy (SERS). APPLIED SPECTROSCOPY 2019; 73:879-892. [PMID: 30990064 DOI: 10.1177/0003702819841913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In this work, the surface engineering method is used to produce Mn helical star-shaped (pine-tree-like) nanosculptured thin films with three-, four-, and fivefold symmetries on Cu substrates using an oblique angle deposition technique together with rotation of the sample holder at certain angles. Nano structure and morphologies of the produced samples were obtained by means of atomic force microscope and field emission scanning electron microscope. Raman spectroscopy of the Mn/Cu samples impregnated by 4,4'-bipyridine (C10H8N2) solution with different concentrations, zidovudine (C10H13N5O4), and L-histidine (C6H9N3O2) was performed using 532 nm laser wavelength. A high degree of enhancement is achieved on Raman spectroscopy of all of these specimens. Comparison of the surface-enhanced Raman spectroscopy (SERS) results for 4,4' bipyridine (bipy) obtained in this work with the published literature using Ag and Au substrates in different shapes showed a significant enhancement improvement by using Mn sculptured structures. Reduction of the bipy concentration changed the enhancement factor. Enhancement factors of 107 and 105 were obtained for threefold symmetry sample using 2.885 × 10-2 and 10-3 mol L-1 bipy concentrations, respectively. Surface-enhanced Raman spectroscopy results of this work show that Mn nanostructures designed and engineered in this work can not only replace Ag and Au materials, but also provide a much higher enhancement factor.
Collapse
Affiliation(s)
- Hadi Savaloni
- 1 School of Physics, College of Science, University of Tehran, Tehran, Iran
| | | | - Reza Babaei
- 2 Plasma Physics Research Center, Faculty of Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
10
|
Wen Z, Peng J, Tuttle PR, Ren Y, Garcia C, Debnath D, Rishi S, Hanson C, Ward S, Kumar A, Liu Y, Zhao W, Glazer PM, Liu Y, Sevilla MD, Adhikary A, Wnuk SF. Electron-Mediated Aminyl and Iminyl Radicals from C5 Azido-Modified Pyrimidine Nucleosides Augment Radiation Damage to Cancer Cells. Org Lett 2018; 20:7400-7404. [PMID: 30457873 PMCID: PMC6465127 DOI: 10.1021/acs.orglett.8b03035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Two classes of azido-modified pyrimidine nucleosides were synthesized as potential radiosensitizers; one class is 5-azidomethyl-2'-deoxyuridine (AmdU) and cytidine (AmdC), while the second class is 5-(1-azidovinyl)-2'-deoxyuridine (AvdU) and cytidine (AvdC). The addition of radiation-produced electrons to C5-azido nucleosides leads to the formation of π-aminyl radicals followed by facile conversion to σ-iminyl radicals either via a bimolecular reaction involving intermediate α-azidoalkyl radicals in AmdU/AmdC or by tautomerization in AvdU/AvdC. AmdU demonstrates effective radiosensitization in EMT6 tumor cells.
Collapse
Affiliation(s)
- Zhiwei Wen
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Jufang Peng
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Paloma R. Tuttle
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Yaou Ren
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Carol Garcia
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Dipra Debnath
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, United States
| | - Sunny Rishi
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, United States
| | - Cameron Hanson
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, United States
| | - Samuel Ward
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, United States
| | - Anil Kumar
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, United States
| | - Yanfeng Liu
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Weixi Zhao
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Peter M. Glazer
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Yuan Liu
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Michael D. Sevilla
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, United States
| | - Amitava Adhikary
- Department of Chemistry, Oakland University, Rochester, Michigan 48309, United States
| | - Stanislaw F. Wnuk
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| |
Collapse
|
11
|
Liu W, Song XL, Zhao SC, He M, Wang H, Chen Z, Xiang W, Yi G, Qi S, Liu Y. Antitumor Activity and Mechanism of a Reverse Transcriptase Inhibitor, Dapivirine, in Glioblastoma. J Cancer 2018; 9:117-128. [PMID: 29290776 PMCID: PMC5743718 DOI: 10.7150/jca.21965] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/18/2017] [Indexed: 01/07/2023] Open
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dapivirine is one of reverse transcriptase inhibitors (RTIs). It is the prototype of diarylpyrimidines (DAPY), formerly known as TMC120 or DAPY R147681 (IUPAC name: 4- [[4-(2, 4, 6-trimethylphenyl) amino]-2-pyrimidinyl] amino]-benzonitrile; CAS no.244767-67-7). AIM The purpose of this study is to investigate the antitumor activity of dapivirine, one of the RTIs, on U87 glioblastoma (GBM) cells in vitro and in vivo. MATERIALS AND METHODS U87 GBM cells were cultured and treated with or without dapivirine. Cell viability was evaluated by CCK-8 (Cell Counting Kit 8, CCK-8) assay; apoptosis was analyzed by flow cytometry; cell migration was evaluated by Boyden Chamber assay; Western blotting was performed to detect proteins related to apoptosis, epithelial-to-mesenchymal transition and autophagy. PathScan intracellular signaling array kit was used to detect important and well-characterized signaling molecules. Tumor xenograft model in nude mice was used to evaluate the antitumorigenic effect in vivo. RESULTS Dapivirine weakened proliferation of glioma cells and induced the apoptosis of U87 glioblastoma cells. Furthermore, dapivirine regulated autophagy and induced Akt, Bad and SAPK/JNK activations. Moreover, the inhibition of glioma cell growth by dapivirine was also observed in nude mice in vivo. CONCLUSION In summary, in our study dapivirine exposure induces stress, resulting in JNK and PI3K/Akt pathway activation through diminished inhibition of the apoptosis and autophagy cascade in U87 GBM cells, which inhibits cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Weiwen Liu
- First College of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xian-Lu Song
- Department of Radiotherapy, Guangzhou Medical University Cancer Institute and Hospital, Guangzhou 510095, China
| | - Shan-Chao Zhao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Minyi He
- Center for Clinical Medical Education, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hai Wang
- Department of Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou 510515, China.,Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou 510515, China
| | - Ziyang Chen
- Department of Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou 510515, China.,Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou 510515, China
| | - Wei Xiang
- Department of Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou 510515, China.,Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou 510515, China
| | - Guozhong Yi
- Department of Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou 510515, China.,Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou 510515, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou 510515, China.,Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou 510515, China
| | - Yawei Liu
- Department of Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou 510515, China.,Laboratory for Precision Neurosurgery, Nanfang hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
12
|
Wang H, Zhou J, He Q, Dong Y, Liu Y. Azidothymidine inhibits cell growth and telomerase activity and induces DNA damage in human esophageal cancer. Mol Med Rep 2017; 15:4055-4060. [PMID: 28487971 PMCID: PMC5436214 DOI: 10.3892/mmr.2017.6549] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 02/27/2017] [Indexed: 12/26/2022] Open
Abstract
Esophageal cancer is one of the most common type of malignancies. Telomerase activity, which is absent or weakly detected in the majority of human somatic cells, is elevated in esophageal cancer. Although azidothymidine (AZT), a reverse transcriptase inhibitor, has been utilized as a treatment for tumors, its role in treating esophageal cancer has not been confirmed. The aim of the present study was to determine the effect of AZT on telomerase activity and the proliferation of the human esophageal cancer cell line TE-11. A telomeric repeat amplification assay was utilized to detect telomerase activity following treatment of TE-11 cells with AZT. The effect of AZT on TE-11 cell cycle distribution was determined by flow cytometry. Cellular DNA damage was evaluated by a comet assay and an MTT assay demonstrated that AZT significantly inhibited the viability of TE-11 cells, in a time-and dose-dependent manner. In addition, TE-11 cells treated with various concentrations of AZT exhibited a significant reduction in telomerase activity and percentage of cells in the G1/G0 phase, and an increase in the percentage of cells in the S phase. High doses of AZT caused DNA damage, and enhanced the expression levels of γ-H2A histone family member X and phosphorylated checkpoint kinase 2 in TE-11 cells. These results demonstrated that AZT effectively inhibits proliferation of the TE-11 human esophageal cancer cell line in vitro. The growth inhibitory effects were associated with a reduction in telomerase activity, S and G2/M phase cell cycle arrest, and enhanced DNA damage, suggesting that AZT may be utilized in the clinic for the treatment of esophageal cancer.
Collapse
Affiliation(s)
- Haoli Wang
- Department of Pathology, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, Guangdong 510080, P.R. China
| | - Jianwen Zhou
- Department of Pathology, First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Qiong He
- Department of Pathology, First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yu Dong
- Department of Pathology, First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yanhui Liu
- Department of Pathology, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
13
|
Shaimerdenova M, Karapina O, Mektepbayeva D, Alibek K, Akilbekova D. The effects of antiviral treatment on breast cancer cell line. Infect Agent Cancer 2017; 12:18. [PMID: 28344640 PMCID: PMC5364572 DOI: 10.1186/s13027-017-0128-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 03/16/2017] [Indexed: 12/28/2022] Open
Abstract
Background Recent studies have revealed the positive antiproliferative and cytotoxic effects of antiviral agents in cancer treatment. The real effect of adjuvant antiviral therapy is still controversial due to the lack of studies in biochemical mechanisms. Here, we studied the effect of the antiviral agent acyclovir on morphometric and migratory features of the MCF7 breast cancer cell line. Molecular levels of various proteins have also been examined. Methods To evaluate and assess the effect of antiviral treatment on morphometric, migratory and other cellular characteristics of MCF7 breast cancer cells, the following experiments were performed: (i) MTT assay to measure the viability of MCF7 cells; (ii) Colony formation ability by soft agar assay; (iii) Morphometric characterization by immunofluorescent analysis using confocal microscopy; (iv) wound healing and transwell membrane assays to evaluate migration and invasion capacity of the cells; (v) ELISA colorimetric assays to assess expression levels of caspase-3, E-cadherin and enzymatic activity of aldehyde dehydrogenase (ALDH). Results We demonstrate the suppressive effect of acyclovir on breast cancer cells. Acyclovir treatment decreases the growth and the proliferation rate of cells and correlates with the upregulated levels of apoptosis associated cytokine Caspase-3. Moreover, acyclovir inhibits colony formation ability and cell invasion capacity of the cancer cells while enhancing the expression of E-cadherin protein in MCF7 cells. Breast cancer cells are characterized by high ALDH activity and associated with upregulated proliferation and invasion. According to this study, acyclovir downregulates ALDH activity in MCF7 cells. Conclusions These results are encouraging and demonstrate the possibility of partial suppression of cancer cell proliferation using an antiviral agent. Acyclovir antiviral agents have a great potential as an adjuvant therapy in the cancer treatment. However, more research is necessary to identify relevant biochemical mechanisms by which acyclovir induces a potent anti-cancer effect. Electronic supplementary material The online version of this article (doi:10.1186/s13027-017-0128-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Madina Shaimerdenova
- National Laboratory Astana, Nazarbayev University, Qabanbay Batyr Avenue 53, Astana, 010000 Kazakhstan
| | - Orynbassar Karapina
- Nazarbayev University Research and Innovation System, Nazarbayev University, Astana, Kazakhstan
| | - Damel Mektepbayeva
- National Laboratory Astana, Nazarbayev University, Qabanbay Batyr Avenue 53, Astana, 010000 Kazakhstan
| | | | - Dana Akilbekova
- National Laboratory Astana, Nazarbayev University, Qabanbay Batyr Avenue 53, Astana, 010000 Kazakhstan
| |
Collapse
|
14
|
Berardinelli F, Coluzzi E, Sgura A, Antoccia A. Targeting telomerase and telomeres to enhance ionizing radiation effects in in vitro and in vivo cancer models. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2017; 773:204-219. [PMID: 28927529 DOI: 10.1016/j.mrrev.2017.02.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 01/05/2023]
Abstract
One of the hallmarks of cancer consists in the ability of tumor cells to divide indefinitely, and to maintain stable telomere lengths throughout the activation of specific telomere maintenance mechanisms (TMM). Therefore in the last fifteen years, researchers proposed to target telomerase or telomeric structure in order to block limitless replicative potential of cancer cells providing a fascinating strategy for a broad-spectrum cancer therapy. In the present review, we report in vitro and in vivo evidence regarding the use of chemical agents targeting both telomerase or telomere structure and showing promising antitumor effects when used in combination with ionizing radiation (IR). RNA interference, antisense oligonucleotides (e.g., GRN163L), non-nucleoside inhibitors (e.g., BIBR1532) and nucleoside analogs (e.g., AZT) represent some of the most potent strategies to inhibit telomerase activity used in combination with IR. Furthermore, radiosensitizing effects were demonstrated also for agents acting directly on the telomeric structure such as G4-ligands (e.g., RHPS4 and Telomestatin) or telomeric-oligos (T-oligos). To date, some of these compounds are under clinical evaluation (e.g., GRN163L and KML001). Advantages of Telomere/Telomerase Targeting Compounds (T/TTCs) coupled with radiotherapy may be relevant in the treatment of radioresistant tumors and in the development of new optimized treatment plans with reduced dose adsorbed by patients and consequent attenuation of short- end long-term side effects. Pros and cons of possible future applications in cancer therapy based on the combination of T/TCCs and radiation treatment are discussed.
Collapse
Affiliation(s)
- F Berardinelli
- Dipartimento di Scienze, Università Roma Tre, Rome Italy; Istituto Nazionale di Fisica Nucleare, INFN, Sezione di Roma Tre, Rome, Italy.
| | - E Coluzzi
- Dipartimento di Scienze, Università Roma Tre, Rome Italy
| | - A Sgura
- Dipartimento di Scienze, Università Roma Tre, Rome Italy; Istituto Nazionale di Fisica Nucleare, INFN, Sezione di Roma Tre, Rome, Italy
| | - A Antoccia
- Dipartimento di Scienze, Università Roma Tre, Rome Italy; Istituto Nazionale di Fisica Nucleare, INFN, Sezione di Roma Tre, Rome, Italy
| |
Collapse
|
15
|
Yang H, Wu L, Ke S, Wang W, Yang L, Gao X, Fang H, Yu H, Zhong Y, Xie C, Zhou F, Zhou Y. Downregulation of Ubiquitin-conjugating Enzyme UBE2D3 Promotes Telomere Maintenance and Radioresistance of Eca-109 Human Esophageal Carcinoma Cells. J Cancer 2016; 7:1152-62. [PMID: 27326259 PMCID: PMC4911883 DOI: 10.7150/jca.14745] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/26/2016] [Indexed: 12/28/2022] Open
Abstract
Ubiquitin-conjugating enzyme UBE2D3 is an important member of the ubiquitin-proteasome pathways. Our previous study showed that the expression of UBE2D3 was negatively related to human telomerase reverse transcriptase (hTERT) and radioresistance in human breast cancer cells. However, in esophageal carcinoma, the exact effects and mechanisms of UBE2D3 in radioresistance remain unclear. This study shows that UBE2D3 knockdown was associated with significant increases in radioresistance to X-rays, telomerase activity, telomere length, and telomere shelterins. UBE2D3 knockdown-mediated radioresistance was related to a decrease in the spontaneous and ionizing radiation-induced apoptosis, resulting from a decrease in the Bax/Bcl-2 ratio. Furthermore, UBE2D3 downregulation was associated with increased G1-S phase transition and prolonged IR-induced G2/M arrest through over expression of cyclin D1, decrease of CDC25A expression and promotion of the ATM/ATR-Chk1-CDC25C pathway. Moreover, UBE2D3 downregulation reduced spontaneous DNA double-strand breaks and accelerated the repair of DNA damage induced by IR. The current data thus demonstrate that UBE2D3 downregulation enhances radioresistance by increased telomere homeostasis and prolonged IR-induced G2/M arrest, but decreases the IR-induced apoptosis and the number of DNA damage foci. These results suggest that UBE2D3 might be a potential molecular target to improve radiotherapy effects in esophageal carcinoma.
Collapse
Affiliation(s)
- Hui Yang
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Lin Wu
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 3. Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Shaobo Ke
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Wenbo Wang
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Lei Yang
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xiaojia Gao
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Hongyan Fang
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Haijun Yu
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yahua Zhong
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Conghua Xie
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Fuxiang Zhou
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yunfeng Zhou
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Chen X, Wang C, Guan S, Liu Y, Han L, Cheng Y. Zidovudine, abacavir and lamivudine increase the radiosensitivity of human esophageal squamous cancer cell lines. Oncol Rep 2016; 36:239-46. [PMID: 27220342 DOI: 10.3892/or.2016.4819] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/26/2016] [Indexed: 11/06/2022] Open
Abstract
Telomerase is a type of reverse transcriptase that is overexpressed in almost all human tumor cells, but not in normal tissues, which provides an opportunity for radiosensitization targeting telomerase. Zidovudine, abacavir and lamivudine are reverse transcriptase inhibitors that have been applied in clinical practice for several years. We sought to explore the radiosensitization effect of these three drugs on human esophageal cancer cell lines. Eca109 and Eca9706 cells were treated with zidovudine, abacavir and lamivudine for 48 h before irradiation was administered. Samples were collected 1 h after irradiation. Clonal efficiency assay was used to evaluate the effect of the combination of these drugs with radiation doses of 2, 4, 6 and 8 Gy. DNA damage was measured by comet assay. Telomerase activity (TA) and relative telomere length (TL) were detected and evaluated by real-time PCR. Apoptosis rates were assessed by flow cytometric analysis. The results showed that all the drugs tested sensitized the esophageal squamous cell carcinoma (ESCC) cell lines to radiation through an increase in radiation-induced DNA damage and cell apoptosis, deregulation of TA and decreasing the shortened TL caused by radiation. Each of the drugs investigated (zidovudine, abacavir and lamivudine) could be used for sensitizing human esophageal cancer cell lines to radiation. Consequently, the present study supports the potential of these three drugs as therapeutic agents for the radiosensitization of esophageal squamous cell cancer.
Collapse
Affiliation(s)
- Xuan Chen
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Cong Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Shanghui Guan
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yuan Liu
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lihui Han
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
17
|
Yang X, Li Z, Yang L, Lei H, Yu H, Liao Z, Zhou F, Xie C, Zhou Y. Knockdown of telomeric repeat binding factor 2 enhances tumor radiosensitivity regardless of telomerase status. J Cancer Res Clin Oncol 2015; 141:1545-52. [PMID: 25609072 DOI: 10.1007/s00432-015-1911-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/05/2015] [Indexed: 10/24/2022]
Abstract
PURPOSE To investigate the effects of TRF2 depletion on radiosensitivity in both the telomerase-positive cell lines (A549) and alternative lengthening of telomere (ALT) cell lines (U2OS). METHODS X-ray irradiation was used to establish two radioresistant cancer models (A549R and U2OSR) from A549 and U2OS. Colony formation assay was applied to examine the radiosensitivity of radioresistant A549R and U2OSR cells and TRF2 low-expression cells. Real-time PCR and TeloTAGGG Telomerase PCR ELISA Kit were performed to examine telomere length and telomerase activity separately. γ-H2AX was detected by immunofluorescence to assess the radiation-induced DSBs. RESULTS Radioresistant cancer models were established, in which TRF2 was significantly over-expressed. Low expression of TRF2 protein could enhance the radiosensitivity and induce telomere length of A549 and U2OS cell shortening. In A549 cells with TRF2 down-regulated, the telomerase activity was inhibited, too. TRF2 deficiency increases γ-H2AX foci and fails to protect telomere from radiation. CONCLUSION The data suggest that TRF2 is a radioresistant protein in A549 and U2OS cells, and could potentially be a target for radiosensitization of both telomerase-positive and ALT cells in radiotherapy.
Collapse
Affiliation(s)
- Xiaoxi Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China,
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Shoja MH, Reddy ND, Nayak PG, Srinivasan KK, Rao CM. Glycosmis pentaphylla (Retz.) DC arrests cell cycle and induces apoptosis via caspase-3/7 activation in breast cancer cells. JOURNAL OF ETHNOPHARMACOLOGY 2015; 168:50-60. [PMID: 25839119 DOI: 10.1016/j.jep.2015.03.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 03/20/2015] [Accepted: 03/22/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Glycosmis pentaphylla (Retz.) DC belonging to the family Rutaceae has been traditionally used for the treatment of rheumatism, anaemia, jaundice, skin diseases, bronchitis etc. The plant is traditionally considered as anti-cancer medicine and used by the healers of Bangladesh to treat all types of cancers. Perhaps the key to many of its medicinal applications is its inherent anti-inflammatory property. AIM OF THE STUDY The present study is aimed at evaluating the effect of various fractions of G. pentaphylla (Retz.) DC leaves on the cell cycle and apoptosis of breast cancer cells viz. MCF-7 and MDA-MB-231. MATERIALS AND METHODS Various extracts and fractions of the leaves of G. pentaphylla (Retz.) DC were studied for their cytotoxicity with the help of Sulforhodamine B assay, in MCF-7, MDA-MB-231 and Vero cell lines. The most active fractions were studied for their effect on the cell cycle of MCF-7 and MDA-MB-231 cells. Apoptotic studies were done using Hoechst staining, DNA fragmentation, Annexin V staining and caspase-3/7 activation assay in breast cancer cells. HPLC and HPTLC profiling of the active fractions were done. RESULTS HPTLC and HPLC profiling revealed the presence of lupeol, chrysin, quercetin, β-sitosterol and kaempferol as components in active fractions. Lupeol and chrysin are being reported in this plant for the first time. The studies showed that the selected fractions possess cell cycle inhibitory and apoptosis inducing effect on both MCF-7 and MDA-MB-231 cells. Apoptotic effect of the fractions on MCF-7 and MDA-MB-231 cells may be through the mitochondrial pathway by the activation of caspase-3/7.
Collapse
Affiliation(s)
- M H Shoja
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal 576104, Karnataka, India
| | - Neetinkumar D Reddy
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal 576104, Karnataka, India
| | - Pawan G Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal 576104, Karnataka, India
| | - K K Srinivasan
- Department of Chemistry, Shri Madhwa Vadiraja Institute of Technology and Management, Bantakal, Udupi 574115, Karnataka, India
| | - C Mallikarjuna Rao
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal 576104, Karnataka, India.
| |
Collapse
|
19
|
Lei H, Feng D, Zhou F, Xu H, Tang T, Yu H, Xie C, Zhou Y. Expression of human protection of telomere 1 correlates with telomere length and radiosensitivity in the human laryngeal cancer Hep-2 cell line. Oncol Lett 2015; 10:1149-1154. [PMID: 26622642 DOI: 10.3892/ol.2015.3332] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 05/15/2015] [Indexed: 11/06/2022] Open
Abstract
The close association between telomere length and radiosensitivity has been established by several studies. There is also a hypothesis that telomere length may be regulated by human protection of telomere 1 (hPOT1) in human carcinoma cells. In the present study, the hPOT1 level between the radioresistant Hep-2R cells and the wild-type were compared, and the results showed that the hPOT1 gene was upregulated in the radioresistant Hep-2R cell lines compared with the wild-type. This suggested that the expression level of hPOT1 correlates with radiosensitivity. Additionally, an hPOT1-directed short hairpin (sh)RNA plasmid was constructed and transferred into the Hep-2R cells, which lead to telomere shortening, an increase in apoptosis and markedly decreased growth of the RNAi-Hep-2R cell line. These results demonstrate that hPOT1-directed shRNAs are associated with telomere length and radiosensitivity, and maybe a potent sensitizer for laryngeal cancer radiotherapy.
Collapse
Affiliation(s)
- Han Lei
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, P.R. China
| | - Dali Feng
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, P.R. China
| | - Fuxiang Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, P.R. China ; Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, P.R. China
| | - Hui Xu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, P.R. China
| | - Tian Tang
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, P.R. China
| | - Haijun Yu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, P.R. China ; Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, P.R. China
| | - Conghua Xie
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, P.R. China ; Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, P.R. China
| | - Yunfeng Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, P.R. China ; Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, P.R. China
| |
Collapse
|
20
|
Yang L, Wang W, Hu L, Yang X, Zhong J, Li Z, Yang H, Lei H, Yu H, Liao Z, Zhou F, Xie C, Zhou Y. Telomere-binding protein TPP1 modulates telomere homeostasis and confers radioresistance to human colorectal cancer cells. PLoS One 2013; 8:e81034. [PMID: 24260532 PMCID: PMC3834294 DOI: 10.1371/journal.pone.0081034] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/08/2013] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Radiotherapy is one of the major therapeutic strategies in cancer treatment. The telomere-binding protein TPP1 is an important component of the shelterin complex at mammalian telomeres. Our previous reports showed that TPP1 expression was elevated in radioresistant cells, but the exact effects and mechanisms of TPP1 on radiosensitivity is unclear. PRINCIPAL FINDINGS In this study, we found that elevated TPP1 expression significantly correlated with radioresistance and longer telomere length in human colorectal cancer cell lines. Moreover, TPP1 overexpression showed lengthened telomere length and a significant decrease of radiosensitivity to X-rays. TPP1 mediated radioresistance was correlated with a decreased apoptosis rate after IR exposure. Furthermore, TPP1 overexpression showed prolonged G2/M arrest mediated by ATM/ATR-Chk1 signal pathway after IR exposure. Moreover, TPP1 overexpression accelerated the repair kinetics of total DNA damage and telomere dysfunction induced by ionizing radiation. CONCLUSIONS We demonstrated that elevated expressions of TPP1 in human colorectal cancer cells could protect telomere from DNA damage and confer radioresistance. These results suggested that TPP1 may be a potential target in the radiotherapy of colorectal cancer.
Collapse
Affiliation(s)
- Lei Yang
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Wenbo Wang
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Liu Hu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xiaoxi Yang
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Juan Zhong
- Department of Oncology, the Fifth Hospital of Wuhan, Wuhan, China
| | - Zheng Li
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Hui Yang
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Han Lei
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Haijun Yu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - ZhengKai Liao
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Fuxiang Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Conghua Xie
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yunfeng Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
- * E-mail:
| |
Collapse
|
21
|
Wang W, Yang L, Hu L, Li F, Ren L, Yu H, Liu Y, Xia L, Lei H, Liao Z, Zhou F, Xie C, Zhou Y. Inhibition of UBE2D3 expression attenuates radiosensitivity of MCF-7 human breast cancer cells by increasing hTERT expression and activity. PLoS One 2013; 8:e64660. [PMID: 23741361 PMCID: PMC3669415 DOI: 10.1371/journal.pone.0064660] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 04/17/2013] [Indexed: 12/25/2022] Open
Abstract
The known functions of telomerase in tumor cells include replenishing telomeric DNA and maintaining cell immortality. We have previously shown the existence of a negative correlation between human telomerase reverse transcriptase (hTERT) and radiosensitivity in tumor cells. Here we set out to elucidate the molecular mechanisms underlying regulation by telomerase of radiosensitivity in MCF-7 cells. Toward this aim, yeast two-hybrid (Y2H) screening of a human laryngeal squamous cell carcinoma radioresistant (Hep2R) cDNA library was first performed to search for potential hTERT interacting proteins. We identified ubiquitin-conjugating enzyme E2D3 (UBE2D3) as a principle hTERT-interacting protein and validated this association biochemically. ShRNA-mediated inhibition of UBE2D3 expression attenuated MCF-7 radiosensitivity, and induced the accumulation of hTERT and cyclin D1 in these cells. Moreover, down-regulation of UBE2D3 increased hTERT activity and cell proliferation, accelerating G1 to S phase transition in MCF-7 cells. Collectively these findings suggest that UBE2D3 participates in the process of hTERT-mediated radiosensitivity in human breast cancer MCF-7 cells by regulating hTERT and cyclin D1.
Collapse
Affiliation(s)
- Wenbo Wang
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Lei Yang
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Liu Hu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Fen Li
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Li Ren
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Haijun Yu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yu Liu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Ling Xia
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Han Lei
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Zhengkai Liao
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Fuxiang Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Conghua Xie
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yunfeng Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
22
|
Millet P, Granotier C, Etienne O, Boussin FD. Radiation-induced upregulation of telomerase activity escapes PI3-kinase inhibition in two malignant glioma cell lines. Int J Oncol 2013; 43:375-82. [PMID: 23727752 PMCID: PMC3775596 DOI: 10.3892/ijo.2013.1970] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Accepted: 04/19/2013] [Indexed: 02/07/2023] Open
Abstract
Tumor relapse after radiotherapy is a great concern in the treatment of high-grade gliomas. Inhibition of the PI3-kinase/AKT pathway is known to radiosensitize cancer cells and to delay their DNA repair after irradiation. In this study, we show that the radiosensitization of CB193 and T98G, two high-grade glioma cell lines, by the PI3K inhibitor LY294002, correlates with the induction of G1 and G2/M arrest, but is inconsistently linked to a delayed DNA double-strand break (DSBs) repair. The PI3K/AKT pathway has been shown to activate radioprotective factors such as telomerase, whose inhibition may contribute to the radiosensitization of cancer cells. However, we show that radiation upregulates telomerase activity in LY-294002-treated glioma cells as well as untreated controls, demonstrating a PI3K/AKT-independent pathway of telomerase activation. Our study suggests that radiosensitizing strategies based on PI3-kinase inhibition in high-grade gliomas may be optimized by additional treatments targeting either telomerase activity or telomere maintenance.
Collapse
Affiliation(s)
- P Millet
- CEA, DSV-IRCM-SCSR, Laboratory of Radiopathology, UMR 967, F-92260 Fontenay‑aux‑Roses, France.
| | | | | | | |
Collapse
|
23
|
Ramkumaar GR, Srinivasan S, Bhoopathy TJ, Gunasekaran S. Molecular geometry, vibrational spectra, atomic charges, frontier molecular orbital and Fukui function analysis of antiviral drug zidovudine. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2012; 99:189-195. [PMID: 23064549 DOI: 10.1016/j.saa.2012.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 09/09/2012] [Indexed: 06/01/2023]
Abstract
The solid phase FT-IR and FT-Raman spectra of zidovudine (AZT) were recorded in the regions 4000-400 and 3500-100 cm(-1), respectively. The optimized geometry, frequency and intensity of the vibrational bands of zidovudine were obtained by the Restricted Hartree-Fock (RHF) density functional theory (DFT) with complete relaxation in the potential energy surface using 6-31G(d,p) basis set. The harmonic vibrational frequencies for zidovudine were calculated and the scaled values have been compared with experimental values of FTIR and FT-Raman spectra. The observed and the calculated frequencies are found to be in good agreement. The harmonic vibrational wave numbers and intensities of vibrational bands of zidovudine with its cation and anion were calculated and compared with the neutral AZT. The DFT calculated HOMO and LUMO energies shows that charge transfer occurs within the molecule. The electron density-based local reactivity descriptors such as Fukui functions were calculated to explain the chemical selectivity or reactivity site in AZT.
Collapse
Affiliation(s)
- G R Ramkumaar
- PG and Research Department of Physics, Pachaiyappa's College, Chennai 600 030, TN, India
| | | | | | | |
Collapse
|
24
|
Alibek K, Bekmurzayeva A, Mussabekova A, Sultankulov B. Using antimicrobial adjuvant therapy in cancer treatment: a review. Infect Agent Cancer 2012; 7:33. [PMID: 23164412 PMCID: PMC3637577 DOI: 10.1186/1750-9378-7-33] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 11/05/2012] [Indexed: 12/29/2022] Open
Abstract
Recent clinical and pre-clinical data demonstrate that adjuvant antimicrobial therapy is beneficial in cancer treatment. There could be several reasons for this effect, which include treating cancer associated bacteria and viruses, prophylaxis of post-chemotherapy infections due to immunosuppression, and antiproliferative effect of certain antimicrobials. Targeting cancer associated viruses and bacteria with antimicrobial agents is currently used for gastric, cervical, hematopoietic, liver and brain cancer. However this treatment is effective only in combination with conventional therapies. Antimicrobials can also have a direct antiproliferative and cytotoxic effect, and can cause apoptosis. Moreover, some antimicrobials are known to be helpful in overcoming side effects of drugs commonly used in cancer treatment. Chemotherapy related bacteremia and neutropenia can be overcome by the appropriately timed use of antimicrobials. This review summarizes the data on the effects of antivirals and antibiotics on cancer treatment and describes their mechanisms.
Collapse
Affiliation(s)
- Kenneth Alibek
- Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan
- Republican Scientific Center for Emergency Care, 3 Kerey and Zhanibek Khanov Street, Astana 010000, Kazakhstan
| | - Aliya Bekmurzayeva
- “Nazarbayev University Research and Innovation System” private institution, Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan
| | - Assel Mussabekova
- “Nazarbayev University Research and Innovation System” private institution, Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan
| | - Bolat Sultankulov
- “Nazarbayev University Research and Innovation System” private institution, Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan
| |
Collapse
|
25
|
Ivanov S. Biochemical markers predicting response to radiation- and radiochemo-therapy in cancer patients. ACTA ACUST UNITED AC 2012; 58:635-50. [DOI: 10.18097/pbmc20125806635] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In last years there is increasing interest in radiogenomics and the characterization of DNA array molecular profiles that can predict tumor and no tumor tissues radioresponse. Ongoing studies carried out worldwide in the banking of tumor and no tumor samples give evidence that perspective markers for response prediction in individual patient to intended radiation therapy can be some apoptotic indexes, spectrum a number of specific proteins, and DNA-based microarray molecular profiling analysis as well determination of single nucleotide polymorphisms in genome of the patients. So far there are only a few robust reports of molecular markers predicting tumor and no tumor tissues response to radiation. The results of new studies, which in future should be validated in larger definitive trials, are likely to see in nearest years. It is needed to determine technologies of methods and to define more precisely areas of its applications.
Collapse
Affiliation(s)
- S.D. Ivanov
- Russian Research Center for Radiology and Surgical Technologies
| |
Collapse
|
26
|
Paul P, Bansal P, Nayak PG, Pannakal ST, Priyadarsini KI, Unnikrishnan MK. Polyphenolic fraction of Pilea microphylla (L.) protects Chinese hamster lung fibroblasts against γ-radiation-induced cytotoxicity and genotoxicity. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2012; 33:107-119. [PMID: 22196050 DOI: 10.1016/j.etap.2011.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 09/15/2011] [Accepted: 09/16/2011] [Indexed: 05/31/2023]
Abstract
Present study was designed to compare cytoprotective and antigenotoxic activity of the polyphenolic fraction of Pilea microphylla (PM1) with that of its active polyphenolic constituents against γ-radiation in V79 cells. PM1 was standardized with respect to the polyphenols present by RP-HPLC. It was evaluated for its free radical scavenging potential using Fenton reaction-induced DNA damage and lipid peroxidation. Further, PM1 was subjected against γ-radiation-induced cytotoxicity and genotoxicity in V79 cells. PM1 significantly reduced free radical-mediated calf thymus DNA damage and lipid peroxidation. Among the concentrations tested (12.5, 25 and 50 μg/ml) for radioprotection, PM1 at 25 μg/ml exhibited maximum protection. Further, when compared with constituent polyphenols viz., rutin, quercetin and chlorogenic acid (concentrations equivalent to that present in PM1-25 μg/ml), a combination of polyphenols was found most effective in preventing γ-radiation-induced cytotoxicity and genotoxicity. To conclude, radioprotection is possibly a synergistic effect of the phytochemicals present in the herbal extract, rather than any single component.
Collapse
Affiliation(s)
- Piya Paul
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal 576104, Karnataka, India
| | | | | | | | | | | |
Collapse
|
27
|
Wu Q, Beland FA, Chang CW, Fang JL. XPC is essential for nucleotide excision repair of zidovudine-induced DNA damage in human hepatoma cells. Toxicol Appl Pharmacol 2010; 251:155-62. [PMID: 21192964 DOI: 10.1016/j.taap.2010.12.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 12/14/2010] [Accepted: 12/16/2010] [Indexed: 12/18/2022]
Abstract
Zidovudine (3'-azido-3'-dexoythymidine, AZT), a nucleoside reverse transcriptase inhibitor, can be incorporated into DNA and cause DNA damage. The mechanisms underlying the repair of AZT-induced DNA damage are unknown. To investigate the pathways involved in the recognition and repair of AZT-induced DNA damage, human hepatoma HepG2 cells were incubated with AZT for 2 weeks and the expression of DNA damage signaling pathways was determined using a pathway-based real-time PCR array. Compared to control cultures, damaged DNA binding and nucleotide excision repair (NER) pathways showed significantly increased gene expression. Further analysis indicated that AZT treatment increased the expression of genes associated with NER, including XPC, XPA, RPA1, GTF2H1, and ERCC1. Western blot analysis demonstrated that the protein levels of XPC and GTF2H1 were also significantly up-regulated. To explore further the function of XPC in the repair of AZT-induced DNA damage, XPC expression was stably knocked down by 71% using short hairpin RNA interference. In the XPC knocked-down cells, 100 μM AZT treatment significantly increased [³H]AZT incorporation into DNA, decreased the total number of viable cells, increased the release of lactate dehydrogenase, induced apoptosis, and caused a more extensive G2/M cell cycle arrest when compared to non-transfected HepG2 cells or HepG2 cells transfected with a scrambled short hairpin RNA sequence. Overall, these data indicate that XPC plays an essential role in the NER repair of AZT-induced DNA damage.
Collapse
Affiliation(s)
- Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR 72079, USA
| | | | | | | |
Collapse
|
28
|
Zhou FX, Xiong J, Luo ZG, Dai J, Yu HJ, Liao ZK, Lei H, Xie CH, Zhou YF. cDNA Expression Analysis of a Human Radiosensitive-Radioresistant Cell Line Model Identifies Telomere Function as a Hallmark of Radioresistance. Radiat Res 2010; 174:550-7. [DOI: 10.1667/rr1657.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
29
|
Adhikary A, Khanduri D, Pottiboyina V, Rice CT, Sevilla MD. Formation of aminyl radicals on electron attachment to AZT: abstraction from the sugar phosphate backbone versus one-electron oxidation of guanine. J Phys Chem B 2010; 114:9289-99. [PMID: 20575557 PMCID: PMC2912441 DOI: 10.1021/jp103403p] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Employing electron spin resonance (ESR) spectroscopy, we have characterized the radicals formed in 3'-azido-3'-deoxythymidine (3'-AZT) and in its 5'-analog 5'-azido-5'-deoxythymidine (5'-AZT) after electron attachment in gamma-irradiated aqueous (H(2)O or D(2)O) glassy (7.5 M LiCl) systems. ESR spectral studies and theoretical calculations show that the predominant site of electron capture in 3'-AZT and in 5'-AZT is at the azide group and not at the thymine moiety. The azide group in AZT is therefore more electron affinic than the most electron affinic DNA base, thymine. Electron attachment to 3'-AZT and 5'-AZT results in an unstable azide anion radical intermediate (RN(3)*(-)) that is too short-lived to be observed in our work even at 77 K. At 77 K, we observe the neutral aminyl radical (RNH*) after loss of N(2) from RN(3)*(-) followed by protonation of nitrene anion radical (RN*(-)) to give RNH*. The expected RN*(-) intermediate is not observed as protonation from water is complete at 77 K even under highly basic conditions. Formation of RND* in D(2)O solutions confirms water as the source of the NH proton in the RNH*. Our assignments to these radicals are aided by DFT calculations for hyperfine coupling constants that closely match the experimental values. On annealing to higher temperatures (ca. 160-170 K), RNH* undergoes bimolecular hydrogen abstraction reactions from the thymine methyl group and the sugar moiety resulting in the formation of the thymine allyl radical (UCH(2)*) and two sugar radicals, C3'* and C5'*. RNH* also results in one-electron oxidation of the guanine base in 3'-AZG. This work provides a potential mechanism for the reported radiosensitization effects of AZT.
Collapse
Affiliation(s)
- Amitava Adhikary
- Department of Chemistry, Oakland University, Rochester, MI 48309
| | - Deepti Khanduri
- Department of Chemistry, Oakland University, Rochester, MI 48309
| | | | - Cory T. Rice
- Department of Chemistry, Oakland University, Rochester, MI 48309
| | | |
Collapse
|
30
|
Chen CM, Johnson M, Smith BJ, Dornfeld K. Azidothymidine enhances fluorodeoxyuridine-mediated radiosensitization. Int J Radiat Oncol Biol Phys 2010; 76:905-13. [PMID: 20159365 PMCID: PMC2900777 DOI: 10.1016/j.ijrobp.2009.09.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Revised: 09/11/2009] [Accepted: 09/14/2009] [Indexed: 11/24/2022]
Abstract
PURPOSE To examine the role of DNA repair and altered thymidine analogues in altering the response to radiation during thymidine deprivation. METHODS AND MATERIALS Mismatch repair-deficient and -proficient cell lines HEC59 and HC-2.4 were treated with fluorodeoxyuridine (FUdR), azidothymidine (AZT), and irradiation either alone or in combination, and outcomes of clonogenic survival and cell-cycle distributions were determined. RESULTS Survival outcomes for all treatments were similar for both cell lines, suggesting that hMSH2 does not significantly influence thymidine deprivation toxicity or radiosensitization. The chain-terminating thymidine analogue AZT increased the toxicity of FUdR and increased DNA fragmentation. The combination of FUdR and AZT afforded greater radiosensitization than either drug alone. Drug enhancement ratios, the degree of excess radiation-induced cell death in drug-treated cultures compared with radiation alone for HEC59, were 1.2, 1.4, and 1.8 for AZT, FUdR, and the combination, respectively. Enhancement ratios for HC-2.4 were 1.3, 1.5, and 1.8 for AZT, FUdR, and the combination, respectively. CONCLUSION Azidothymidine, a chain-terminating thymidine analogue, can enhance the radiosensitizing affects of thymidine deprivation. Deoxyribonucleic acid strand breaks may play an important role in the mechanism of thymidine deprivation-induced radiosensitization.
Collapse
Affiliation(s)
- Chang-Ming Chen
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA 52242
| | - Monika Johnson
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA 52242
| | - Brian J. Smith
- College of Public Health, University of Iowa, Iowa City, IA, USA 52242
| | - Ken Dornfeld
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA 52242
| |
Collapse
|
31
|
Liao Z, Huang C, Zhou F, Xiong J, Bao J, Zhang H, Sun W, Xie C, Zhou Y. Radiation enhances suicide gene therapy in radioresistant laryngeal squamous cell carcinoma via activation of a tumor-specific promoter. Cancer Lett 2009; 283:20-8. [PMID: 19375219 DOI: 10.1016/j.canlet.2009.03.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2009] [Revised: 03/11/2009] [Accepted: 03/15/2009] [Indexed: 10/20/2022]
Abstract
Radioresistant cells have been shown to correlate with poor outcome after radiotherapy. Here, we found that human telomerase reverse transcriptase promoter (hTERTp) had lower activity in laryngeal squamous carcinomas cells than in radioresistant variant cells. Combining radiotherapy with plasmid phTERTp-HRP, in which expression of enzyme horseradish peroxidase (HRP) controlled by hTERTp, resulted in increased apoptosis and necrosis of tumor cells after prodrug indole-3-acetic acid (IAA; converted by HRP into a cytotoxin) incubation. Volume and wet weight of xenograft tumor were reduced more in the combination groups. These data suggest that hTERTp has potential use in targeted cancer gene therapy, especially for radioresistant tumors. Combining radiotherapy with hTERTp-HRP/IAA may overcome radioresistance in laryngeal squamous carcinomas cells and amplify the killing effect in targeted cancer cells.
Collapse
Affiliation(s)
- Zhengkai Liao
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Department of Radiochemotherapy, Zhongnan Hospital, Wuhan University, Hubei Province, Wuhan 430071, China
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Scruggs ER, Dirks Naylor AJ. Mechanisms of zidovudine-induced mitochondrial toxicity and myopathy. Pharmacology 2008; 82:83-88. [PMID: 18504416 DOI: 10.1159/000134943] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Accepted: 02/22/2008] [Indexed: 01/06/2023]
Abstract
Zidovudine (3-azido-3'-deoxythymidine), also referred to as azidothymidine (AZT), has become an integral component in highly active antiretroviral therapy, and has also been used in the treatment of cancer. The clinical effectiveness of AZT is constrained due to its association with increased adverse effects, such as myopathy. There are numerous potential mechanisms that may contribute to AZT-induced myopathy. The first hypothesized mechanism to explain AZT-induced toxicity was mtDNA depletion due to inhibition of DNA polymerase gamma. Although mtDNA depletion is present in patients with myopathy, current data suggests that alternative mechanisms may play a more direct role in the myotoxicity. These mechanisms include AZT-induced oxidative stress, direct inhibition of mitochondrial bioenergetic machinery, and mitochondrial depletion of L-carnitine. Furthermore, we hypothesize that apoptosis may play a role in AZT-induced myopathy.
Collapse
Affiliation(s)
- Erin R Scruggs
- Wingate University School of Pharmacy, Wingate, NC 28174, USA
| | | |
Collapse
|
33
|
Zhong YH, Liao ZK, Zhou FX, Xie CH, Xiao CY, Pan DF, Luo ZG, Liu SQ, Zhou YF. Telomere length inversely correlates with radiosensitivity in human carcinoma cells with the same tissue background. Biochem Biophys Res Commun 2008; 367:84-9. [DOI: 10.1016/j.bbrc.2007.12.078] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2007] [Accepted: 12/11/2007] [Indexed: 10/22/2022]
|
34
|
Tárkányi I, Aradi J. Pharmacological intervention strategies for affecting telomerase activity: future prospects to treat cancer and degenerative disease. Biochimie 2007; 90:156-72. [PMID: 17945408 DOI: 10.1016/j.biochi.2007.09.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Accepted: 09/04/2007] [Indexed: 12/20/2022]
Abstract
Telomerase enzyme is a ribonucleoprotein maintaining the length of the telomeres by adding G-rich repeats to the end of the eukaryotic chromosomes. Normal human somatic cells, cultured in vitro, have a strictly limited proliferative potential undergoing senescence after about 50-70 population doublings. In contrast, most of the tumor cells have unlimited replicative potential. Although the mechanisms of immortalization are not understood completely at a genetic level, the key role of the telomere/telomerase system in the process is clear. The DNA replication machinery is not able to replicate fully the DNA at the very end of the chromosomes; therefore, about 50-200 nucleotides are lost during each of the replication cycles resulting in a gradual decrease of telomere length. Critically short telomere induces senescence, subsequent crisis and cell death. In tumor cells, however, the telomerase enzyme prevents the formation of critically short telomeres, adding GGTTAG repeats to the 3' end of the chromosomes immortalizing the cells. Immortality is one of the hallmarks of cancer. Besides the catalytic activity dependent telomere maintenance, catalytic activity-independent effects of telomerase may also be involved in the regulation of cell cycle. The telomere/telomerase system offers two possibilities to intervene the proliferative activity of the cell: (1) inhibition the telomere maintenance by inhibiting the telomerase activity; (2) activating the residual telomerase enzyme or inducing telomerase expression. Whilst the former approach could abolish the limitless replicative potential of malignant cells, the activation of telomerase might be utilized for treating degenerative diseases. Here, we review the current status of telomerase therapeutics, summarizing the activities of those pharmacological agents which either inhibit or activate the enzyme. We also discuss the future opportunities and challenges of research on pharmacological intervention of telomerase activity.
Collapse
Affiliation(s)
- I Tárkányi
- 3rd Department of Internal Medicine, University of Debrecen, 22 Moricz Zsigmond Krt., Debrecen 4004, Hungary
| | | |
Collapse
|
35
|
Sun YQ, Guo TK, Xi YM, Chen C, Wang J, Wang ZR. Effects of AZT and RNA-protein complex (FA-2-b-β) extracted from Liang Jin mushroom on apoptosis of gastric cancer cells. World J Gastroenterol 2007; 13:4185-91. [PMID: 17696246 PMCID: PMC4250616 DOI: 10.3748/wjg.v13.i31.4185] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the synergistic effects of 3'-azido-3'-deoxythymidine (AZT) and FA-2-b-β extracted from Ling Jin mushroom on apoptosis of gastric cancer cells MKN45 in vitro.
METHODS: MTT analysis was made to examine the inhibition rate of MKN45 cells treated with AZT (2.5, 5, 10 and 20 mg/L) and FA-2-b-β (5, 10, 20 and 40 mg/L) singly and combinatively for 24, 48 and 72 h. Apoptotic effects were evaluated by morphological methods, DNA agarose gel electrophoresis and flow cytometry, respectively. Telomerase activity was estimated by TRAP-ELISA. The mRNA expression of caspase-3 and Bcl-2 were detected by RT-PCR.
RESULTS: AZT and FA-2-b-β could significantly inhibit MKN45 cell proliferation and induce its apoptosis. MKN45 cells were inhibited in dose- and time- dependent manner. The inhibition effect of AZT combined with FA-2-b-β was obviously better than that used singly (0.469 ±0.022 vs 1.075 ± 0.055, P < 0.05, 0.325 ± 0.029 vs 0.469± 0.022 P < 0.01). AZT used singly and combination of FA-2-b-β could decrease the activity of tumor cell telomerase, and AZT has synergistic function with FA-2-b-β. A certain concentration of AZT could up-regulate the expression of caspase-3 mRNA (r = 0.9969, P < 0.01), which was positively related to apoptosis rate, and could down-regulate the expression of Bcl-2 mRNA, which was negatively related to apoptosis rate (r = 0.926, P < 0.01). Furthermore, the effect of AZT combined with FA-2-b-β was significantly higher than that used singly.
CONCLUSION: Combination of AZT and FA-2-b-β has an obviously synergetic effect in the gastric cancer cells MKN45, which has provided a new approach to the treatment of gastric cancer clinically.
Collapse
Affiliation(s)
- Yan-Qing Sun
- School of Life Science, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | | | | | | | | | | |
Collapse
|
36
|
De Cian A, Lacroix L, Douarre C, Temime-Smaali N, Trentesaux C, Riou JF, Mergny JL. Targeting telomeres and telomerase. Biochimie 2007; 90:131-55. [PMID: 17822826 DOI: 10.1016/j.biochi.2007.07.011] [Citation(s) in RCA: 484] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Accepted: 07/16/2007] [Indexed: 01/06/2023]
Abstract
Telomeres and telomerase represent, at least in theory, an extremely attractive target for cancer therapy. The objective of this review is to present the latest view on the mechanism(s) of action of telomerase inhibitors, with an emphasis on a specific class of telomere ligands called G-quadruplex ligands, and to discuss their potential use in oncology.
Collapse
Affiliation(s)
- Anne De Cian
- INSERM, U565, Acides nucléiques: dynamique, ciblage et fonctions biologiques, 43 rue Cuvier, CP26, Paris Cedex 05, F-75231, France
| | | | | | | | | | | | | |
Collapse
|