1
|
Penabeï S, Sepulveda E, Zakaria AM, Meesungnoen J, Jay-Gerin JP. Effect of Linear Energy Transfer on Cystamine's Radioprotective Activity: A Study Using the Fricke Dosimeter with 6-500 MeV per Nucleon Carbon Ions-Implication for Carbon Ion Hadrontherapy. Molecules 2023; 28:8144. [PMID: 38138632 PMCID: PMC10746108 DOI: 10.3390/molecules28248144] [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: 10/23/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
(1) Background: Radioprotective agents have garnered considerable interest due to their prospective applications in radiotherapy, public health medicine, and situations of large-scale accidental radiation exposure or impending radiological emergencies. Cystamine, an organic diamino-disulfide compound, is recognized for its radiation-protective and antioxidant properties. This study aims to utilize the aqueous ferrous sulfate (Fricke) dosimeter to measure the free-radical scavenging capabilities of cystamine during irradiation by fast carbon ions. This analysis spans an energy range from 6 to 500 MeV per nucleon, which correlates with "linear energy transfer" (LET) values ranging from approximately 248 keV/μm down to 9.3 keV/μm. (2) Methods: Monte Carlo track chemistry calculations were used to simulate the radiation-induced chemistry of aerated Fricke-cystamine solutions across a broad spectrum of cystamine concentrations, ranging from 10-6 to 1 M. (3) Results: In irradiated Fricke solutions containing cystamine, cystamine is observed to hinder the oxidation of Fe2+ ions, an effect triggered by oxidizing agents from the radiolysis of acidic water, resulting in reduced Fe3+ ion production. Our simulations, conducted both with and without accounting for the multiple ionization of water, confirm cystamine's ability to capture free radicals, highlighting its strong antioxidant properties. Aligning with prior research, our simulations also indicate that the protective and antioxidant efficiency of cystamine diminishes with increasing LET of the radiation. This result can be attributed to the changes in the geometry of the track structures when transitioning from lower to higher LETs. (4) Conclusions: If we can apply these fundamental research findings to biological systems at a physiological pH, the use of cystamine alongside carbon-ion hadrontherapy could present a promising approach to further improve the therapeutic ratio in cancer treatments.
Collapse
Affiliation(s)
| | | | | | | | - Jean-Paul Jay-Gerin
- Département de Médecine Nucléaire et de Radiobiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, 3001, 12ème Avenue Nord, Sherbrooke, QC J1H 5N4, Canada; (S.P.); (E.S.); (A.M.Z.); (J.M.)
| |
Collapse
|
2
|
Mermer A, Tüzün B, Daştan SD, Koçyiğit ÜM, Çetin FN, Çevik Ö. Piperazin incorporated Schiff Base derivatives: Assessment of in vitro biological activities, metabolic enzyme inhibition properties, and molecular docking calculations. J Biochem Mol Toxicol 2023; 37:e23465. [PMID: 37462216 DOI: 10.1002/jbt.23465] [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: 06/13/2022] [Revised: 06/21/2023] [Accepted: 07/06/2023] [Indexed: 11/10/2023]
Abstract
The cytotoxic activities of the compounds were determined by the 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT) method in human breast cancer (MCF-7), human cervical cancer (HeLa), and mouse fibroblast (L929) cell lines. The compounds MAAS-5 and four modified the supercoiled tertiary structure of pBR322 plasmid DNA. MAAS-5 showed the highest cytotoxic activity in HeLa, MCF-7, and L929 cells with IC50 values of 16.76 ± 3.22, 28.83 ± 5.61, and 2.18 ± 1.22 µM, respectively. MAAS-3 was found to have almost the lowest cytotoxic activities with the IC50 values of 93.17 ± 9.28, 181.07 ± 11.54, and 16.86 ± 6.42 µM in HeLa, MCF-7, and L929 cells respectively at 24 h. Moreover, the antiepileptic potentials of these compounds were investigated in this study. To this end, the effect of newly synthesized Schiff base derivatives on the enzyme activities of carbonic anhydrase I and II isozymes (human carbonic anhydrase [hCA] I and hCA II) was evaluated spectrophotometrically. The target compounds demonstrated high inhibitory activities compared with standard inhibitors with Ki values in the range of 4.54 ± 0.86-15.46 ± 8.65 nM for hCA I (Ki value for standard inhibitor = 12.08 ± 2.00 nM), 1.09 ± 0.32-29.94 ± 0.82 nM for hCA II (Ki value for standard inhibitor = 18.22 ± 4.90 nM). Finally, the activities of the compounds were compared with the Gaussian programme in the B3lyp, HF, M062X base sets with 6-31++G (d,p) levels. In addition, the activities of five compounds against various breast cancer proteins and hCA I and II were compared with molecular docking calculations. Also, absorption, distribution, metabolism, excretion, and toxicity analysis was performed to investigate the possibility of using five compounds as drug candidates.
Collapse
Affiliation(s)
- Arif Mermer
- Experimental Medicine Application & Research Center, Validebağ Research Park, University of Health Sciences, Istanbul, Türkiye
- Department of Biotechnology, University of Health Sciences, Istanbul, Türkiye
| | - Burak Tüzün
- Plant and Animal Production Department, Technical Sciences Vocational School of Sivas, Sivas Cumhuriyet University, Sivas, Turkey
| | - Sevgi Durna Daştan
- Department of Biology, Faculty of Science, Sivas Cumhuriyet University, Sivas, Turkey
| | - Ümit M Koçyiğit
- Department of Basic Pharmaceutical Sciences, Sivas Cumhuriyet University, Sivas, Turkey
| | - Feyza Nur Çetin
- Department of Basic Pharmaceutical Sciences, Sivas Cumhuriyet University, Sivas, Turkey
| | - Özge Çevik
- Department of Biochemistry, Adnan Menderes University, Aydın, Turkey
| |
Collapse
|
3
|
Deng T, Zhu Q, Xie L, Liu Y, Peng Y, Yin L, Gao Y, Cao T, Fu Y, Qi X, Zhang S, Peng Y, Hou Y, Li X. Norcantharidin promotes cancer radiosensitization through Cullin1 neddylation-mediated CDC6 protein degradation. Mol Carcinog 2022; 61:812-824. [PMID: 35652616 DOI: 10.1002/mc.23435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/02/2022] [Accepted: 05/23/2022] [Indexed: 11/06/2022]
Abstract
Radiotherapy (RT) is a conventional cancer therapeutic modality. However, cancer cells tend to develop radioresistance after a period of treatment. Diagnostic markers and therapeutic targets for radiosensitivity are severely lacking. Our recently published studies demonstrated that the cell division cycle (CDC6) is a critical molecule contributing to radioresistance, and maybe a potential therapeutic target to overcome radioresistance. In the present study, we for the first time reported that Norcantharidin (NCTD), a demethylated form of cantharidin, re-sensitized radioresistant cancer cells to overcome radioresistance, and synergistically promoted irradiation (IR)-induced cell killing and apoptosis by inducing CDC6 protein degradation. Mechanistically, NCTD induced CDC6 protein degradation through the ubiquitin-proteasome pathways. By using small interfering RNA (siRNA) interference or small compound inhibitors, we further determined that NCTD induced CDC6 protein degradation through a neddylation-dependent pathway, but not through Huwe1, Cyclin F, and APC/C-mediated ubiquitin-proteasome pathways. We screened the six most relevant Cullin subunits (CUL1, 2, 3, 4A, 4B, and 5) using siRNAs. The knockdown of Cullin1 but not the other five cullins remarkably elevated CDC6 protein levels. NCTD promoted the binding of Cullin1 to CDC6, thereby promoting CDC6 protein degradation through a Cullin1 neddylation-mediated ubiquitin-proteasome pathway. NCTD can be used in combination with radiotherapy to achieve better anticancer efficacy, or work as a radiosensitizer to overcome cancer radioresistance.
Collapse
Affiliation(s)
- Tanggang Deng
- Department of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China.,Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Qianling Zhu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Xie
- Department of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China.,Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Youhong Liu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuchong Peng
- Department of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China.,Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Linglong Yin
- Department of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China.,Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China.,School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yingxue Gao
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tuoyu Cao
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuxin Fu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuli Qi
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Songwei Zhang
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongbo Peng
- School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Youxiang Hou
- Department of Radiation Oncology, Tumor Hospital, Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Xiong Li
- Department of Clinical Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China.,Center for Clinical Precision Pharmacy, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China.,NMPA Key Laboratory for Technology Research and Evaluation of Pharmacovigilance, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Yamashita T, Kato T, Isogai T, Gu Y, Ito T, Ma N. Taurine Deficiency in Tissues Aggravates Radiation-Induced Gastrointestinal Syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1370:113-120. [DOI: 10.1007/978-3-030-93337-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
5
|
Saqib M, Arthur-Baidoo E, Ončák M, Denifl S. Electron Attachment Studies with the Potential Radiosensitizer 2-Nitrofuran. Int J Mol Sci 2020; 21:ijms21238906. [PMID: 33255344 PMCID: PMC7727711 DOI: 10.3390/ijms21238906] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
Nitrofurans belong to the class of drugs typically used as antibiotics or antimicrobials. The defining structural component is a furan ring with a nitro group attached. In the present investigation, electron attachment to 2-nitrofuran (C4H3NO3), which is considered as a potential radiosensitizer candidate for application in radiotherapy, has been studied in a crossed electron-molecular beams experiment. The present results indicate that low-energy electrons with kinetic energies of about 0-12 eV effectively decompose the molecule. In total, twelve fragment anions were detected within the detection limit of the apparatus, as well as the parent anion of 2-nitrofuran. One major resonance region of ≈0-5 eV is observed in which the most abundant anions NO2-, C4H3O-, and C4H3NO3- are detected. The experimental results are supported by ab initio calculations of electronic states in the resulting anion, thermochemical thresholds, connectivity between electronic states of the anion, and reactivity analysis in the hot ground state.
Collapse
Affiliation(s)
- Muhammad Saqib
- Institute for Ion Physics and Applied Physics, University of Innsbruck, Technikerstrasse 25, 6020 Innsbruck, Austria; (M.S.); (E.A.-B.)
- Center for Biomolecular Sciences Innsbruck, University of Innsbruck, Technikerstrasse 25, A-6020 Innsbruck, Austria
| | - Eugene Arthur-Baidoo
- Institute for Ion Physics and Applied Physics, University of Innsbruck, Technikerstrasse 25, 6020 Innsbruck, Austria; (M.S.); (E.A.-B.)
- Center for Biomolecular Sciences Innsbruck, University of Innsbruck, Technikerstrasse 25, A-6020 Innsbruck, Austria
| | - Milan Ončák
- Institute for Ion Physics and Applied Physics, University of Innsbruck, Technikerstrasse 25, 6020 Innsbruck, Austria; (M.S.); (E.A.-B.)
- Correspondence: (M.O.); (S.D.)
| | - Stephan Denifl
- Institute for Ion Physics and Applied Physics, University of Innsbruck, Technikerstrasse 25, 6020 Innsbruck, Austria; (M.S.); (E.A.-B.)
- Center for Biomolecular Sciences Innsbruck, University of Innsbruck, Technikerstrasse 25, A-6020 Innsbruck, Austria
- Correspondence: (M.O.); (S.D.)
| |
Collapse
|
6
|
Meißner R, Feketeová L, Ribar A, Fink K, Limão-Vieira P, Denifl S. Electron Ionization of Imidazole and Its Derivative 2-Nitroimidazole. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:2678-2691. [PMID: 31667709 PMCID: PMC6914720 DOI: 10.1007/s13361-019-02337-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/02/2019] [Accepted: 09/08/2019] [Indexed: 05/09/2023]
Abstract
Imidazole (IMI) is a basic building block of many biologically important compounds. Thus, its electron ionization properties are of major interest and essential for the comparison with other molecular targets containing its elemental structure. 2-Nitroimidazole (2NI) contains the imidazole ring together with nitrogen dioxide bound to the C2 position, making it a radiosensitizing compound in hypoxic tumors. In the present study, we investigated electron ionization of IMI and 2NI and determined the mass spectra, the ionization energies, and appearance energies of the most abundant fragment cations. The experiments were complemented by quantum chemical calculations on the thermodynamic thresholds and potential energy surfaces, with particular attention to the calculated transition states for the most important dissociation reactions. In the case of IMI, substantially lower threshold values (up to ~ 1.5 eV) were obtained in the present work compared to the only available previous electron ionization study. Closer agreement was found with recent photon ionization values, albeit the general trend of slightly higher values for the case of electron ionization. The only exception for imidazole was found in the molecular cation at m/z 40 which is tentatively assigned to the quasi-linear HCCNH+/ HCNCH+. Electron ionization of 2NI leads to analogous fragment cations as in imidazole, yet different dissociation pathways must be operative due to the presence of the NO2 group. Regarding the potential radiosensitization properties of 2NI, electron ionization is characterized by dominant parent cation formation and release of the neutral NO radical.
Collapse
Affiliation(s)
- Rebecca Meißner
- Institut für Ionenphysik und Angewandte Physik and Center for Molecular Biosciences Innsbruck (CMBI), Universität Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria.
- Atomic and Molecular Collisions Laboratory, CEFITEC, Department of Physics, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
| | - Linda Feketeová
- Institut für Ionenphysik und Angewandte Physik and Center for Molecular Biosciences Innsbruck (CMBI), Universität Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria.
- Institut de Physique des 2 Infinis de Lyon; CNRS/IN2P3, UMR5822, Université de Lyon, Université Claude Bernard Lyon 1, 43 Bd du 11 novembre 1918, 69622, Villeurbanne, France.
| | - Anita Ribar
- Institut für Ionenphysik und Angewandte Physik and Center for Molecular Biosciences Innsbruck (CMBI), Universität Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria
| | - Katharina Fink
- Institut für Ionenphysik und Angewandte Physik and Center for Molecular Biosciences Innsbruck (CMBI), Universität Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria
| | - Paulo Limão-Vieira
- Atomic and Molecular Collisions Laboratory, CEFITEC, Department of Physics, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
| | - Stephan Denifl
- Institut für Ionenphysik und Angewandte Physik and Center for Molecular Biosciences Innsbruck (CMBI), Universität Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria.
| |
Collapse
|
7
|
Zhang L, Wu C, Mu S, Xue W, Ma D. A chemotherapeutic self-sensibilized drug carrier delivering paclitaxel for the enhanced chemotherapy to human breast MDA-MB-231 cells. Colloids Surf B Biointerfaces 2019; 181:902-909. [DOI: 10.1016/j.colsurfb.2019.06.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/18/2019] [Accepted: 06/22/2019] [Indexed: 01/31/2023]
|
8
|
Sepulveda E, Sanguanmith S, Meesungnoen J, Jay-Gerin JP. Evaluation of the radioprotective ability of cystamine for 150 keV – 500 MeV proton irradiation: a Monte Carlo track chemistry simulation study. CAN J CHEM 2019. [DOI: 10.1139/cjc-2018-0382] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cystamine, an organic diamino-disulfide, is among the best of the known radiation-protective compounds, although the underlying molecular mechanisms by which it operates remain poorly understood. This study aims to use the aqueous ferrous sulfate (Fricke) dosimeter to evaluate the protective properties of this compound when present during irradiation by fast incident protons in the energy range of 150 keV – 500 MeV, that is, for “linear energy transfer” (LET) values ranging from ∼72.3 to 0.23 keV/μm. The presence of cystamine in irradiated Fricke solutions prevents the oxidation of Fe2+ ions by the oxidizing species produced in the radiolysis of acidic water, resulting in reduced Fe3+ ion yields. A Monte Carlo computer code is used to simulate the radiation-induced chemistry of the studied Fricke–cystamine solutions under aerated conditions while covering a wide range of cystamine concentrations from 5 × 10−7 to 1 mol/L. Results indicate that the protective activity of cystamine is due to its radical-capturing ability, a clear signature of the strong antioxidant profile of this compound. In addition, our simulations show that at low and intermediate concentrations of cystamine, its protective efficiency decreases with increasing LET, which is consistent with previous work. This finding stems from differences in the geometry of the track structures that change from low-LET isolated spherical “spurs” to high-LET dense continuous cylindrical tracks as LET increases. This study concludes that Monte Carlo simulations represent a powerful method for understanding, at the molecular level, indirect radiation damage to complex molecules such as cystamine.
Collapse
Affiliation(s)
- Esteban Sepulveda
- Département de médecine nucléaire et de radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12ème Avenue Nord, Sherbrooke QC J1H 5N4, Canada
- Département de médecine nucléaire et de radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12ème Avenue Nord, Sherbrooke QC J1H 5N4, Canada
| | - Sunuchakan Sanguanmith
- Département de médecine nucléaire et de radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12ème Avenue Nord, Sherbrooke QC J1H 5N4, Canada
- Département de médecine nucléaire et de radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12ème Avenue Nord, Sherbrooke QC J1H 5N4, Canada
| | - Jintana Meesungnoen
- Département de médecine nucléaire et de radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12ème Avenue Nord, Sherbrooke QC J1H 5N4, Canada
- Département de médecine nucléaire et de radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12ème Avenue Nord, Sherbrooke QC J1H 5N4, Canada
| | - Jean-Paul Jay-Gerin
- Département de médecine nucléaire et de radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12ème Avenue Nord, Sherbrooke QC J1H 5N4, Canada
- Département de médecine nucléaire et de radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12ème Avenue Nord, Sherbrooke QC J1H 5N4, Canada
| |
Collapse
|
9
|
Yamashita T, Kato T, Tunekawa M, Gu Y, Wang S, Ma N. Effect of Radiation on the Expression of Taurine Transporter in the Intestine of Mouse. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 2:729-740. [PMID: 28849495 DOI: 10.1007/978-94-024-1079-2_57] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
There has been a growing interest on the effects of radiation since the Fukushima nuclear power plant accident of 2011. Taurine has been reported to have a radioprotective effect in irradiated mice. However, the detailed mechanism of this radioprotective effect is still awaiting clarification. The aim of this study was to investigation how radiation affects the expression of taurine and to shed light on the mechanism accounting for radioprotective and radiation mitigating effect. Six-week-old male mice were randomly divided into two groups: IR group (7 Gy irradiation) and IR + Tau group (7 Gy irradiation + taurine 3000 mg/kg/day). We examined the survival rate, the expression of taurine and taurine transporter in the small intestine and the urinary taurine concentration. In this study, no statistically significant difference was found in the survival rate between IR Group and IR + Tau Group. Three days and 7 days after irradiation, the urinary taurine concentration of IR + Tau group increased more than that of IR group. Three days and 10 days after irradiation, the expression of taurine and taurine transporter in the small intestine of IR group and IR + Tau group decreased more than that of normal small intestine. It is reported that radiation exposure increases the urinary taurine concentration. We found that the radiation exposure decreases the expression of the taurine transporter in the small intestine of mouse. This finding suggests that a decrease in the expression of the taurine transporter promotes the release of taurine from the tissue into the urine.
Collapse
Affiliation(s)
- Takenori Yamashita
- Faculty of Health Science, Suzuka University of Medical Science, Suzuka, 513-8670, Mie, Japan
| | - Toshihiro Kato
- Sport Medical Center, Suzukakaisei Hospital, Suzuka, 513-0836, Mie, Japan
| | - Masahiro Tunekawa
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, 513-8670, Mie, Japan
| | - Yeunhwa Gu
- Faculty of Health Science, Junshin Gakuen University, Fukuoka, Japan
| | - Shumin Wang
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, 513-8670, Mie, Japan
| | - Ning Ma
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, 513-8670, Mie, Japan.
| |
Collapse
|
10
|
Gold nanoparticles as a potent radiosensitizer in neutron therapy. Oncotarget 2017; 8:112390-112400. [PMID: 29348833 PMCID: PMC5762518 DOI: 10.18632/oncotarget.19837] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/25/2017] [Indexed: 01/03/2023] Open
Abstract
The purpose of this study was to investigate the potential of gold nanoparticles as radiosensitizer for use in neutron therapy against hepatocellular carcinoma. The hepatocellular carcinoma cells lines Huh7 and HepG2 were irradiated with γ and neutron radiation in the presence or absence of gold nanoparticles. Effects were evaluated by transmission electron microscopy, cell survival, cell cycle, DNA damage, migration, and invasiveness. Gold nanoparticles significantly enhanced the radiosensitivity of Huh7 and HepG2 cells to γ-rays by 1.41- and 1.16-fold, respectively, and by 1.80- and 1.35-fold to neutron radiation, which has high linear energy transfer. Accordingly, exposure to neutron radiation in the presence of gold nanoparticles induced cell cycle arrest, DNA damage, and cell death to a significantly higher extent, and suppressed cell migration and invasiveness more robustly. These effects are presumably due to the ability of gold nanoparticles to amplify the effective dose from neutron radiation more efficiently. The data suggest that gold nanoparticles may be clinically useful in combination therapy against hepatocellular carcinoma by enhancing the toxicity of radiation with high linear energy transfer.
Collapse
|
11
|
Khoei S, Poorabdollahi R, Mostaar A, Faeghi F. Methoxyamine Enhances 5-Fluorouracil-Induced Radiosensitization in Colon Cancer Cell Line HT29. CELL JOURNAL 2017; 19:283-291. [PMID: 28670521 PMCID: PMC5412787 DOI: 10.22074/cellj.2016.4295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/03/2016] [Indexed: 11/24/2022]
Abstract
Objective This study intended to observe the effects of methoxyamine (Mx) on cytotoxic
effects and DNA damage caused by 5-Fluorouracil (5-FU) in combination with gamma
radiation in a human colon cancer cell line, HT29.
Materials and Methods In this experimental study, HT29 cells were cultured as a monolayer and treated with different concentrations of 5-FU along with 1 mM Mx for 24 hours.
Next, the cells were irradiated with 2 Gy gamma radiation. After the treatments, we assessed for DNA damage, cytotoxicity, and viability by alkaline comet, clonogenic survival,
and trypan blue dye exclusion assays.
Results Cytotoxicity and DNA damage increased with increasing 5-FU concentration.
The 1 mM Mx concentration had no significant effect on cytotoxicity and DNA damage
from 5-FU; however, it increased the cytotoxic and genotoxic effects of different concentrations of 5-FU when used in combination with 2 Gy gamma radiation.
Conclusion Mx combined with 5-FU enhanced the radiosensitivity of colon cancer cells.
Collapse
Affiliation(s)
- Samideh Khoei
- Razi Drug Research Centre, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Roghayeh Poorabdollahi
- Department of Radiology Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Mostaar
- Department of Medical Physics and Medical Engineering, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariborz Faeghi
- Department of Radiology Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
GuhaSarkar S, Pathak K, Sudhalkar N, More P, Goda JS, Gota V, Banerjee R. Synergistic locoregional chemoradiotherapy using a composite liposome-in-gel system as an injectable drug depot. Int J Nanomedicine 2016; 11:6435-6448. [PMID: 27942215 PMCID: PMC5138055 DOI: 10.2147/ijn.s110525] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The use of radiosensitizers in clinical radiotherapy is limited by systemic toxicity. The biopolymeric, biodegradable, injectable liposome-in-gel-paclitaxel (LG-PTX) system was developed for regional delivery of the radiosensitizer paclitaxel (PTX), and its efficacy was evaluated with concurrent fractionated radiation. LG-PTX is composed of nano-sized drug-loaded fluidizing liposomes, which are incorporated into a porous biodegradable gellan hydrogel. This allows enhanced drug permeation while maintaining a localization of the drug depot. LG-PTX had an IC50 of 325±117 nM in B16F10 melanoma cells, and cytotoxicity with concurrent doses of fractionated radiation showed significant increase in apoptotic cells (75%) compared to radiation (39%) or LG-PTX (43%) alone. Peri-tumoral injection in tumor-bearing mice showed PTX localization in the tumor 2 hours after administration, with no drug detected in plasma or other organs. LG-PTX administration with doses of focal radiation (5×3 Gy) significantly reduced tumor volumes compared to control (6.4 times) and radiation alone (1.6 times) and improved animal survival. LG-PTX thus efficiently localizes the drug at the tumor site and synergistically enhances the effect of concurrent radiotherapy. This novel liposome-in-gel system can potentially be used as a platform technology for the delivery of radiosensitizing drugs to enhance the efficacy of chemoradiotherapy.
Collapse
Affiliation(s)
- Shruti GuhaSarkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai
| | | | - Niyati Sudhalkar
- Department of Radiation Oncology, Advanced Centre for Treatment Research and Education in Cancer, Navi Mumbai, Maharashtra, India
| | - Prachi More
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai
| | - Jayant Sastri Goda
- Department of Radiation Oncology, Advanced Centre for Treatment Research and Education in Cancer, Navi Mumbai, Maharashtra, India
| | | | - Rinti Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai
| |
Collapse
|
13
|
Bolognesi P, Casavola AR, Cartoni A, Richter R, Markus P, Borocci S, Chiarinelli J, Tošić S, Sa’adeh H, Masič M, Marinković B, Prince K, Avaldi L. Communication: “Position” does matter: The photofragmentation of the nitroimidazole isomers. J Chem Phys 2016; 145:191102. [DOI: 10.1063/1.4967770] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- P. Bolognesi
- CNR-Istituto di Struttura della Materia, Area della Ricerca di Roma1, Monterotondo, Italy
| | - A. R. Casavola
- CNR-Istituto di Struttura della Materia, Area della Ricerca di Roma1, Monterotondo, Italy
| | - A. Cartoni
- CNR-Istituto di Struttura della Materia, Area della Ricerca di Roma1, Monterotondo, Italy
- Dipartimento di Chimica, Sapienza Università di Roma, Roma, Italy
| | - R. Richter
- Elettra-Sincrotrone Trieste, Basovizza, Italy
| | - P. Markus
- CNR-Istituto di Struttura della Materia, Area della Ricerca di Roma1, Monterotondo, Italy
| | - S. Borocci
- Dipartimento per l’Innovazione nei Sistemi Biologici, Agroalimentari e Forestali (DIBAF), Università della Tuscia, Viterbo, Italy
| | - J. Chiarinelli
- CNR-Istituto di Struttura della Materia, Area della Ricerca di Roma1, Monterotondo, Italy
| | - S. Tošić
- Institute of Physics Belgrade, University of Belgrade, Belgrade, Serbia
| | - H. Sa’adeh
- Department of Physics, The University of Jordan, Amman, Jordan
| | - M. Masič
- School of Chemistry, Cardiff University, Cardiff, United Kingdom
| | - B.P. Marinković
- Institute of Physics Belgrade, University of Belgrade, Belgrade, Serbia
| | - K.C. Prince
- Elettra-Sincrotrone Trieste, Basovizza, Italy
| | - L. Avaldi
- CNR-Istituto di Struttura della Materia, Area della Ricerca di Roma1, Monterotondo, Italy
| |
Collapse
|
14
|
Bolognesi P, Kettunen JA, Cartoni A, Richter R, Tosic S, Maclot S, Rousseau P, Delaunay R, Avaldi L. Site- and state-selected photofragmentation of 2Br-pyrimidine. Phys Chem Chem Phys 2015; 17:24063-9. [PMID: 26314495 DOI: 10.1039/c5cp02601f] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The fragmentation of the 2Br-pyrimidine molecule following direct valence photoionization or inner shell excitation has been studied by electron-ion coincidence experiments. 2Br-pyrimidine has been chosen as a model for the class of pyrimidinic building blocks of three nucleic acids and several radiosensitizers. It is known that the site- and state-localization of energy deposition, typical of inner shell excitation, results in the enhancement of the total ion yield as well as in changes in the relative intensity of the different fragmentation channels. Here we address the question of the origin of this selective fragmentation by using electron-ion coincidence techniques. The results show that the fragmentation is strongly selective in the final singly charged ion state, independently of the process that leads to the population of that state, and the dominant fragmentation patterns correlate with the nearest appearance potential.
Collapse
Affiliation(s)
- P Bolognesi
- CNR-Istituto di Struttura della Materia, Area della Ricerca di Roma1, Monterotondo Scalo, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Kim EH, Kim MS, Jeong YK, Cho I, You SH, Cho SH, Lee H, Jung WG, Kim HD, Kim J. Mechanisms for SU5416 as a radiosensitizer of endothelial cells. Int J Oncol 2015; 47:1440-50. [PMID: 26314590 DOI: 10.3892/ijo.2015.3127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/22/2015] [Indexed: 11/06/2022] Open
Abstract
Endothelial cells (ECs), that comprise the tumor vasculature, are critical targets for anticancer radiotherapy. The aim of this work was to study the mechanism by which SU5416, a known anti-angiogenesis inhibitor, modifies the radiation responses of human vascular ECs. Two human endothelial cell lines (HUVEC and 2H11) were treated with SU5416 alone, radiation alone, or a combination of both. In vitro tests were performed using colony forming assays, FACS analysis, western blotting, immunohistochemistry, migration assay, invasion assays and endothelial tube formation assays. The combination of radiation and SU5416 significantly inhibited cell survival, the repair of radiation-induced DNA damage, and induced apoptosis. It also caused cell cycle arrest, inhibited cell migration and invasion, and suppressed angiogenesis. In this study, our results first provide a scientific rationale to combine SU5416 with radiotherapy to target ECs and suggest its clinical application in combination cancer treatment with radiotherapy.
Collapse
Affiliation(s)
- Eun Ho Kim
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Mi-Sook Kim
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Youn Kyoung Jeong
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Ilsung Cho
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Seung Hoon You
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Sung Ho Cho
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Hanna Lee
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Won-Gyun Jung
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Republic of Korea
| | - Hag Dong Kim
- Laboratory of Biochemistry, Division of Life Sciences, Korea University, Seoul 136-701, Republic of Korea
| | - Joon Kim
- Laboratory of Biochemistry, Division of Life Sciences, Korea University, Seoul 136-701, Republic of Korea
| |
Collapse
|
16
|
Chen W, Hu GH. Biomarkers for enhancing the radiosensitivity of nasopharyngeal carcinoma. Cancer Biol Med 2015; 12:23-32. [PMID: 25859408 PMCID: PMC4383846 DOI: 10.7497/j.issn.2095-3941.2014.0015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 01/07/2015] [Indexed: 12/14/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a common head and neck malignancy. The incidence of NPC is higher in Southern China and Southeast Asia compared with Western countries. Given its high radiosensitivity, the standard treatment for NPC is radiotherapy. However, radioresistance remains a serious obstacle to successful treatment. Radioresistance can cause local recurrence and distant metastases in some patients after treatment by radiation. Thus, special emphasis has been given to the discovery of effective radiosensitizers. This review aims to discuss the biomarkers, classified according to the main mechanisms of radiosensitization, which can enhance the sensitivity of NPC cells to ionizing radiation.
Collapse
Affiliation(s)
- Wei Chen
- Department of Otorhinolaryngology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Guo-Hua Hu
- Department of Otorhinolaryngology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
17
|
Anzai K, Ueno M, Matsumoto KI, Ikota N, Takata J. Gamma-tocopherol-N,N-dimethylglycine ester as a potent post-irradiation mitigator against whole body X-irradiation-induced bone marrow death in mice. JOURNAL OF RADIATION RESEARCH 2014; 55:67-74. [PMID: 23908555 PMCID: PMC3885127 DOI: 10.1093/jrr/rrt094] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 06/05/2013] [Accepted: 06/24/2013] [Indexed: 06/02/2023]
Abstract
We examined the radioprotective and mitigative effects of gamma-tocopherol-N,N-dimethylglycine ester (GTDMG), a novel water-soluble gamma-tocopherol derivative, against X-irradiation-induced bone marrow death in mice. Mice (C3H, 10 weeks, male) were injected intraperitoneally with GTDMG suspended in a 0.5% methyl cellulose solution before or after receiving of 7.5-Gy whole body X-irradiation. GTDMG significantly enhanced the 30-day survival rate when given 30 min before or immediately after the irradiation. Its mitigative activity (administered after exposure) was examined further in detail. The optimal concentration of GTDMG given immediately after irradiation was around 100 mg/kg body weight (bw) and the 30-day survival rate was 97.6 ± 2.4%. When GTDMG was administered 1, 10 and 24 h post-irradiation, the survival rate was 85.7 ± 7.6, 75.0 ± 9.7 and 36.7 ± 8.8%, respectively, showing significant mitigation even at 24 h after irradiation (P < 0.05). The value of the dose reduction factor (100 mg/kg bw, given intraperitoneally (i.p.) immediately after irradiation) was 1.25. GTDMG enhanced the recovery of red blood cell-, white blood cell-, and platelet-counts after irradiation and significantly increased the number of endogenous spleen colonies (P < 0.05). Subcutaneous (s.c.) administration also had mitigative effects. In conclusion, GTDMG is a potent radiation mitigator.
Collapse
Affiliation(s)
- Kazunori Anzai
- Nihon Pharmaceutical University
- National Institute of Radiological Sciences
| | | | | | | | - Jiro Takata
- Faculty of Pharmaceutical Sciences, Fukuoka University
| |
Collapse
|
18
|
Wilde F, Chamseddin C, Lemmerhirt H, Bednarski PJ, Jira T, Link A. Evaluation of (S)- and (R)-misonidazole as GPX inhibitors: synthesis, characterization including circular dichroism and in vitro testing on bovine GPx-1. Arch Pharm (Weinheim) 2013; 347:153-60. [PMID: 24375829 DOI: 10.1002/ardp.201300285] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/24/2013] [Accepted: 09/24/2013] [Indexed: 11/06/2022]
Abstract
Racemic misonidazole, a radiosensitizer formally used in radiation therapy of cancer and to date still applied, was once reported to exhibit strong inhibitory effects on mouse glutathione peroxidases (GPX). This appeared to qualify misonidazole as a lead structure for the development of novel GPX inhibitors to cause oxidative stress in chemotherapy-resistant tumors. A unique feature of misonidazole as an inhibitor of GPX is the absence of a thiol functionality. Therefore, it was expected to selectively target inhibition devoid of promiscuous interactions with cations and sulfhydryl groups. We synthesized the isomers of misonidazole and analyzed the ability of chiroptical high-performance liquid chromatography (HPLC) to identify the particular enantiomers. Due to the chiral pool synthesis, the assignment of the correct configuration could be verified. Finally, we evaluated both isomers for their inhibitory activities on bovine erythrocyte GPx-1, which is 87% homologous to the human enzyme. Despite the previously reported inhibition of racemic misonidazole on the less homologous mouse GPx-1, we did not find any significant inhibitory activity on the bovine enzyme for either isomer. Though misonidazole appears unlikely to be an inhibitor of human GPx-1 activity, we still spotlight misonidazole as a promising fragment-like lead structure in general.
Collapse
Affiliation(s)
- Felix Wilde
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | | | | | | | | | | |
Collapse
|
19
|
Tchanque-Fossuo CN, Gong B, Poushanchi B, Donneys A, Sarhaddi D, Gallagher KK, Deshpande SS, Goldstein SA, Morris MD, Buchman SR. Raman spectroscopy demonstrates Amifostine induced preservation of bone mineralization patterns in the irradiated murine mandible. Bone 2013; 52:712-717. [PMID: 22885239 PMCID: PMC3789510 DOI: 10.1016/j.bone.2012.07.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 07/26/2012] [Accepted: 07/27/2012] [Indexed: 12/28/2022]
Abstract
PURPOSE Adjuvant radiotherapy in the management of head and neck cancer remains severely debilitating. Fortunately, newly developed agents aimed at decreasing radiation-induced damage have shown great promise. Amifostine (AMF) is a compound, which confers radio-protection to the exposed normal tissues, such as bone. Our intent is to utilize Raman spectroscopy to demonstrate how AMF preserves the mineral composition of the murine mandible following human equivalent radiation. METHODS Sprague Dawley rats were randomized into 3 experimental groups: control (n=5), XRT (n=5), and AMF-XRT (n=5). Both XRT and AMF groups underwent bioequivalent radiation of 70Gy in 5 fractions to the left hemimandible. AMF-XRT received Amifostine prior to radiation. Fifty-six days post-radiation, the hemimandibles were harvested, and Raman spectra were taken in the region of interest spanning 2mm behind the last molar. Bone mineral and matrix-specific Raman bands were analyzed using one-way ANOVA, with statistical significance at p<0.05. RESULTS The full-width at half-maximum of the primary phosphate band (FWHM) and the ratio of carbonate/phosphate intensities demonstrated significant differences between AMF-XRT versus XRT (p<0.01) and XRT versus control (p<0.01). There was no difference between AMF-XRT and control (p>0.05) in both Raman metrics. Computer-aided spectral subtraction further confirmed these results where AMF-XRT was spectrally similar to the control. Interestingly, the collagen cross-link ratio did not differ between XRT and AMF-XRT (p<0.01) but was significantly different from the control (p<0.01). CONCLUSION Our novel findings demonstrate that AMF prophylaxis maintains and protects bone mineral quality in the setting of radiation. Raman spectroscopy is an emerging and exceptionally attractive clinical translational technology to investigate and monitor both the destructive effects of radiation and the therapeutic remediation of AMF on the structural, physical and chemical qualities of bone.
Collapse
Affiliation(s)
- Catherine N. Tchanque-Fossuo
- University of Michigan Hospital and Health Systems, Pediatric Plastic Surgery Section, 1540 E. Hospital Drive, MI 48109, USA
| | - Bo Gong
- University of Michigan Hospital and Health Systems, Department of Chemistry, 930 N. University, Ann Arbor, MI 48109, USA
| | - Behdod Poushanchi
- University of Michigan Hospital and Health Systems, Pediatric Plastic Surgery Section, 1540 E. Hospital Drive, MI 48109, USA
| | - Alexis Donneys
- University of Michigan Hospital and Health Systems, Pediatric Plastic Surgery Section, 1540 E. Hospital Drive, MI 48109, USA
| | - Deniz Sarhaddi
- University of Michigan Hospital and Health Systems, Pediatric Plastic Surgery Section, 1540 E. Hospital Drive, MI 48109, USA
| | - K. Kelly Gallagher
- University of Michigan Hospital and Health Systems, Department of Otolaryngology–Head and Neck Surgery, University of Michigan Medical School, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Sagar S. Deshpande
- University of Michigan Hospital and Health Systems, Pediatric Plastic Surgery Section, 1540 E. Hospital Drive, MI 48109, USA
| | - Steven A. Goldstein
- University of Michigan, Department of Orthopaedic Surgery, A. Alfred Taubman Biomedical Science Research Building, 109 Zina Pitcher Pl, Ann Arbor, MI, 48109, USA
| | - Michael D. Morris
- University of Michigan Hospital and Health Systems, Department of Chemistry, 930 N. University, Ann Arbor, MI 48109, USA
| | - Steven R. Buchman
- University of Michigan Hospital and Health Systems, Pediatric Plastic Surgery Section, 1540 E. Hospital Drive, MI 48109, USA
| |
Collapse
|
20
|
Zhang L, Yang L, Li JJ, Sun L. Potential use of nucleic acid-based agents in the sensitization of nasopharyngeal carcinoma to radiotherapy. Cancer Lett 2012; 323:1-10. [DOI: 10.1016/j.canlet.2012.03.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 03/26/2012] [Accepted: 03/26/2012] [Indexed: 11/27/2022]
|
21
|
Ueno M, Inano H, Onoda M, Murase H, Ikota N, Kagiya TV, Anzai K. Modification of Mortality and Tumorigenesis by Tocopherol-mono-glucoside (TMG) Administered after X Irradiation in Mice and Rats. Radiat Res 2009; 172:519-24. [DOI: 10.1667/rr1695.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
22
|
Radiation Effects and Radioprotection in MC3T3-E1 Mouse Calvarial Osteoblastic Cells. Plast Reconstr Surg 2008; 122:1025-1035. [DOI: 10.1097/prs.0b013e3181845931] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
23
|
Isenberg JS, Maxhimer JB, Hyodo F, Pendrak ML, Ridnour LA, DeGraff WG, Tsokos M, Wink DA, Roberts DD. Thrombospondin-1 and CD47 limit cell and tissue survival of radiation injury. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:1100-12. [PMID: 18787106 DOI: 10.2353/ajpath.2008.080237] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Radiation, a primary mode of cancer therapy, acutely damages cellular macromolecules and DNA and elicits stress responses that lead to cell death. The known cytoprotective activity of nitric oxide (NO) is blocked by thrombospondin-1, a potent antagonist of NO/cGMP signaling in ischemic soft tissues, suggesting that thrombospondin-1 signaling via its receptor CD47 could correspondingly increase radiosensitivity. We show here that soft tissues in thrombospondin-1-null mice are remarkably resistant to radiation injury. Twelve hours after 25-Gy hindlimb irradiation, thrombospondin-1-null mice showed significantly less cell death in both muscle and bone marrow. Two months after irradiation, skin and muscle units in null mice showed minimal histological evidence of radiation injury and near full retention of mitochondrial function. Additionally, both tissue perfusion and acute vascular responses to NO were preserved in irradiated thrombospondin-1-null hindlimbs. The role of thrombospondin-1 in radiosensitization is specific because thrombospondin-2-null mice were not protected. However, mice lacking CD47 showed radioresistance similar to thrombospondin-1-null mice. Both thrombospondin-1- and CD47-dependent radiosensitization is cell autonomous because vascular cells isolated from the respective null mice showed dramatically increased survival and improved proliferative capacity after irradiation in vitro. Therefore, thrombospondin-1/CD47 antagonists may have selective radioprotective activity for normal tissues.
Collapse
Affiliation(s)
- Jeff S Isenberg
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1500, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Anzai K, Ikota N, Ueno M, Nyui M, Kagiya TV. Heat-treated mineral-yeast as a potent post-irradiation radioprotector. JOURNAL OF RADIATION RESEARCH 2008; 49:425-430. [PMID: 18480592 DOI: 10.1269/jrr.07127] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
In vivo radioprotection of C3H mice by i.p. administration of Zn-, Mn-, Cu-, or Se-containing heat-treated Saccharomyces serevisiae yeast sample was examined. The 30-day survival of the group treated 30 min before 7.5 Gy whole-body X-irradiation with mineral-containing yeast powders suspended in 0.5% methylcellulose was significantly higher than that of control group. When mineral-yeast was administered immediately after irradiation, the survival rate was even higher and Zn- or Cu-yeast showed the highest rate (more than 90%). Although treatment with simple yeast showed a high survival rate (73%), it was significantly lower than that obtained by the Zn-yeast treatment. The effects of Zn-yeast were studied further. When the interval between irradiation and administration was varied, the protective activity of Zn-yeast decreased gradually by increasing the interval but was still significantly high for the administration at 10 h post-irradiation. The dose reduction factor of Zn-yeast (100 mg/kg, i.p. administration immediately after irradiation) was about 1.2. When the suspension of Zn-yeast was fractionated by centrifugation, the insoluble fraction showed a potent effect, while the soluble fraction had only a moderate effect. In conclusion, mineral-yeast, especially Zn-yeast, provides remarkable post-irradiation protection against lethal whole body X-irradiation. The activity is mainly attributable to the insoluble fraction, whereas some soluble components might contribute to the additional protective activity.
Collapse
Affiliation(s)
- Kazunori Anzai
- Research Center for Charged Particle Therapy, National Institute of Radiological Sciences.
| | | | | | | | | |
Collapse
|
25
|
Gevorgyan A, La Scala GC, Neligan PC, Pang CY, Forrest CR. Radioprotection of craniofacial bone growth. J Craniofac Surg 2007; 18:995-1000. [PMID: 17912071 DOI: 10.1097/scs.0b013e31812f7596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
In this review, the potential of pharmacologic therapy for prevention of radiation-induced bone growth inhibition is discussed. Significant radioprotection using the radioprotector Amifostine has been achieved in animal models of radiation-induced retardation of long and craniofacial bone growth. Moreover, radioprotection in vitro has been achieved in a number of cell lines, including osteoblast-like, endothelial, and fibroblastic. This evidence may support future clinical investigations of radioprotector Amifostine or similar substances for radioprotection of the growing craniofacial skeleton.
Collapse
Affiliation(s)
- Artur Gevorgyan
- Division of Plastic Surgery, Hospital for Sick Children, Toronto, Canada
| | | | | | | | | |
Collapse
|
26
|
Wardman P. Chemical radiosensitizers for use in radiotherapy. Clin Oncol (R Coll Radiol) 2007; 19:397-417. [PMID: 17478086 DOI: 10.1016/j.clon.2007.03.010] [Citation(s) in RCA: 318] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Accepted: 03/13/2007] [Indexed: 12/21/2022]
Abstract
Radiosensitizers are intended to enhance tumour cell killing while having much less effect on normal tissues. Some drugs target different physiological characteristics of the tumour, particularly hypoxia associated with radioresistance. Oxygen is the definitive hypoxic cell radiosensitizer, the large differential radiosensitivity of oxic vs hypoxic cells being an attractive factor. The combination of nicotinamide to reduce acute hypoxia with normobaric carbogen breathing is showing clinical promise. 'Electron-affinic' chemicals that react with DNA free radicals have the potential for universal activity to combat hypoxia-associated radioresistance; a nitroimidazole, nimorazole, is clinically effective at tolerable doses. Hypoxia-specific cytotoxins, such as tirapazamine, are valuable adjuncts to radiotherapy. Nitric oxide is a potent hypoxic cell radiosensitizer; variations in endogenous levels might have prognostic significance, and routes to deliver nitric oxide specifically to tumours are being developed. In principle, many drugs can be delivered selectively to hypoxic tumours using either reductase enzymes or radiation-produced free radicals to activate drug release from electron-affinic prodrugs. A redox-active agent based on a gadolinium chelate is being evaluated clinically. Pyrimidines substituted with bromine or iodine are incorporated into DNA and enhance free radical damage; fluoropyrimidines act by different mechanisms. A wide variety of drugs that influence the nature or repair of DNA damage are being evaluated in conjunction with radiation; it is often difficult to define the mechanisms underlying chemoradiation regimens. Drugs being evaluated include topoisomerase inhibitors (e.g. camptothecin, topotecan), and the hypoxia-activated anthraquinone AQ4N; alkylating agents include temozolomide. Drugs involved in DNA repair pathways being investigated include the potent poly(ADP ribose)polymerase inhibitor, AG14,361. Proteins involved in cell signalling, such as the Ras family, are attractive targets linked to radioresistance, as are epidermal growth factor receptors and linked kinases (drugs including vandetanib [ZD6,474], cetuximab and gefitinib), and cyclooxygenase-2 (celecoxib). The suppression of radioprotective thiols seems to offer more potential with alkylating agents than with radiotherapy, although it remains a strategy worthy of exploration.
Collapse
Affiliation(s)
- P Wardman
- University of Oxford, Gray Cancer Institute, PO Box 100, Mount Vernon Hospital, Northwood HA6 2JR, UK.
| |
Collapse
|
27
|
Bai A, Meier GP, Wang Y, Luberto C, Hannun YA, Zhou D. Prodrug Modification Increases Potassium Tricyclo[5.2.1.02,6]-decan-8-yl Dithiocarbonate (D609) Chemical Stability and Cytotoxicity against U937 Leukemia Cells. J Pharmacol Exp Ther 2004; 309:1051-9. [PMID: 14960662 DOI: 10.1124/jpet.103.064600] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Potassium tricyclo[5.2.1.0(2,6)]-decan-8-yl dithiocarbonate (D609) is a selective antitumor agent, potent antioxidant, and cytoprotectant. It has the potential to be developed as a unique chemotherapeutic agent that may provide dual therapeutic benefits against cancer, e.g., enhancing tumor cell death while protecting normal tissues from damage. However, D609 contains a dithiocarbonate (xanthate) group [O-C(=S)S(-)/O-C(=S)SH], which is chemically unstable, being readily oxidized to form a disulfide bond with subsequent loss of all biological activities. Therefore, we developed the synthesis of a series of S-(alkoxyacyl) D609 prodrugs by connecting the xanthate group of D609 to an ester via a self-immolative methyleneoxyl group. These S-(alkoxylacyl)-D609 prodrugs are designed to release D609 in two steps: esterase-catalyzed hydrolysis of the acyl ester bond followed by conversion of the resulting hydroxymethyl D609 to formaldehyde and D609. Three S-(alkoxyacyl) D609 prodrugs were synthesized by varying the steric bulkiness of the acyl group. These prodrugs are stable to ambient conditions, but readily hydrolyzed by esterases to liberate D609 in a controlled manner. More importantly, the lead prodrug methyleneoxybutyryl D609 is biologically more effective than D609 in inhibiting sphingomyelin synthase, thereby increasing the level of ceramide and inducing apoptosis in U937 leukemia cells. The prodrug has a significantly lower LD(50) value than that of D609 (56.6 versus 117 microM) against U937 cells. These findings demonstrate that prodrug modification of the xanthate moiety with an alkoxyacyl group can improve D609 oxidative stability and enhance its antitumor activity.
Collapse
Affiliation(s)
- Aiping Bai
- Department of Pathology, Medical University of South Carolina, 165 Ashley Ave., Suite 309, P.O. Box 250908, Charleston, SC 29425, USA
| | | | | | | | | | | |
Collapse
|
28
|
Riesterer O, Tenzer A, Zingg D, Hofstetter B, Vuong V, Pruschy M, Bodis S. Novel radiosensitizers for locally advanced epithelial tumors: inhibition of the PI3K/Akt survival pathway in tumor cells and in tumor-associated endothelial cells as a novel treatment strategy? Int J Radiat Oncol Biol Phys 2004; 58:361-8. [PMID: 14751504 DOI: 10.1016/j.ijrobp.2003.09.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
In locally advanced epithelial malignancies, local control can be achieved with high doses of radiotherapy (RT). Concurrent chemoradiotherapy can improve tumor control in selected solid epithelial adult tumors; however, treatment-related toxicity is of major concern and the therapeutic window often small. Therefore, novel pharmacologic radiosensitizers with a tumor-specific molecular target and a broad therapeutic window are attractive. Because of clonal heterogeneity and the high mutation rate of these tumors, combined treatment with single molecular target radiosensitizers and RT are unlikely to improve sustained local tumor control substantially. Therefore, radiosensitizers modulating entire tumor cell survival pathways in epithelial tumors are of potential clinical use. We discuss the preclinical efficacy and the mechanism of three different, potential radiosensitizers targeting the PTEN/PI3K/Akt survival pathway. These compounds were initially thought to act as single-target agents against growth factor receptors (PKI 166 and PTK 787) or protein kinase C isoforms (PKC 412). We describe an additional target for these compounds. PKI 166 (an epidermal growth factor [EGF] receptor inhibitor) and PKC 412, target the PTEN/PI3K/Akt pathway mainly in tumor cells, and PTK 787 (a vascular endothelial growth factor [VEGF] receptor inhibitor) in endothelial cells. Even for these broader range molecular radiosensitizers, the benefit could be restricted to human epithelial tumor cell clones with a distinct molecular profile. Therefore, these potential radiosensitizers have to be carefully tested in specific model systems before introduction in early clinical trials.
Collapse
Affiliation(s)
- Oliver Riesterer
- Department of Radiation Oncology, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
The RAF-1 serine-threonine kinase plays a central role in signal transduction pathways involved in cell survival and proliferation. The concept of RAF-1-targeted disruption of cell signaling for therapeutic purposes was first advanced in 1989 with the demonstration of tumor growth inhibition in athymic mice and radiosensitization of human squamous carcinoma cells transfected with a vector expressing antisense cDNA. However, the clinical application of antisense strategies has awaited the development of improved antisense oligonucleotide technologies and drug delivery methods. Nuclease-resistant phosphorothioated antisense oligonucleotides have been the focus of pharmaceutical industry attention. In vivo delivery of nuclease-sensitive, natural backbone/phosphodiester oligonucleotides has remained a formidable challenge. Liposomal encapsulation of antisense oligonucleotides protects them from degradation and enhances drug delivery. Here, we review the importance of targeting RAF-1 signaling in cancer therapy and the preclinical and clinical experiences with a liposomal formulation of a nuclease-sensitive, ends-modified antisense RAF oligonucleotide.
Collapse
Affiliation(s)
- Usha Kasid
- Department of Radiation Medicine, Lombardi Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| | | |
Collapse
|