1
|
Kazmierska-Grebowska P, Jankowski MM, Obrador E, Kolodziejczyk-Czepas J, Litwinienko G, Grebowski J. Nanotechnology meets radiobiology: Fullerenols and Metallofullerenols as nano-shields in radiotherapy. Biomed Pharmacother 2025; 184:117915. [PMID: 39983431 DOI: 10.1016/j.biopha.2025.117915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025] Open
Abstract
Despite significant advances in the development of radioprotective measures, the clinical application of radioprotectors and radiomitigators remains limited due to insufficient efficacy and high toxicity of most agents. Additionally, in oncological radiotherapy, these compounds may interfere with the therapeutic effectiveness. Recent progress in nanotechnology highlights fullerenols (FulOHs) and metallofullerenols (Me@FulOHs) as promising candidates for next-generation radioprotectors. These nanostructures possess unique antioxidant properties, demonstrating greater efficacy in rediucing oxidative stress compared to conventional agents. Moreover, their potential to minimize pro-oxidative risks depends on the precise identification of cellular environments and irradiation conditions that optimize their radioprotective effects. In parallel, Me@FulOHs serve as powerful theranostic tools in oncology. Their strong imaging signals enable high-resolution PET and MRI, facilitating early detection and accurate localization of pathogenic alterations. This dual functionality positions Me@FulOHs as key components in advanced radiotherapy. By integrating these nanomaterials with modern theranostic approaches, it is possible to enhance the precision of treatment while minimizing side effects, addressing a critical need in contemporary oncology. This review emphasizes the importance of systematic evaluation of context-dependent effects of Me@FulOHs, particularly in pre- and post-irradiation scenarios, to optimize their clinical relevance. The dual role of Me@FulOHs as both radioprotectors and diagnostic agents distinguishes them from traditional compounds, paving the way for innovative practical applications. Their use in radiotherapy represents a significant step toward the development of safer and more effective strategies in radiation protection and cancer treatment. We also review ionizing radiation effects, classifications, cancer radiotherapy applications, and countermeasures.
Collapse
Affiliation(s)
- Paulina Kazmierska-Grebowska
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, Lodz 90-236, Poland
| | - Maciej M Jankowski
- BioTechMed Center, Multimedia Systems Department, Faculty of Electronics, Telecommunications and Informatics, Gdansk University of Technology, Gabriela Narutowicza 11/12, Gdansk 80-233, Poland
| | - Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, Valencia 46010, Spain
| | - Joanna Kolodziejczyk-Czepas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, Lodz 90-236, Poland
| | | | - Jacek Grebowski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, Lodz 90-236, Poland; Military Institute of Medicine - National Research Institute, Szaserow 128, Warsaw 04-141, Poland.
| |
Collapse
|
2
|
Pan G, Xia Y, Hao M, Guan J, Zhu Q, Zha T, Sheng L, Zhao Z, Pan H, Fang W, Xu X, Chen X, Zhou S, Tong Z. EZH2 suppresses IR-induced ferroptosis by forming a co-repressor complex with HIF-1α to inhibit ACSL4: Targeting EZH2 enhances radiosensitivity in KDM6A-deficient esophageal squamous carcinoma. Cell Death Differ 2025:10.1038/s41418-025-01451-5. [PMID: 39920286 DOI: 10.1038/s41418-025-01451-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 12/29/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
The mutation status of the lysine demethylase 6 A (KDM6A), a gene antagonist to Enhancer of zeste homolog 2 (EZH2), is closely related to the therapeutic efficacy of EZH2 inhibitors in several malignancies. However, the mutational landscape of KDM6A and the therapeutic targetability of EZH2 inhibitors in esophageal squamous carcinoma (ESCC) remain unreported. Here, we found that approximately 9.18% (9/98) of our study ESCC tissues had KDM6A mutations of which 7 cases resulted in a complete loss of expression and consequent loss of demethylase function. We found that KDM6A-deficient ESCC cells exhibited increased sensitivity to EZH2 inhibitor, and the radiosensitizing activity of EZH2 inhibitor was evident in KDM6A-dficient ESCC cells. Further transcriptome analysis revealed that ferroptosis is implicated in the radiosensitizing effect exerted by EZH2 inhibition on KDM6A-deficient ESCC cells. The following Chromatin Immunoprecipitation (ChIP), co-immunoprecipitation, and luciferase reporter assays demonstrated that in KDM6A-deficient ESCC cells, (1) Acyl-CoA Synthetase Long Chain Family Member 4 (ACSL4) is the target gene for EZH2 to regulate ferroptosis; (2) The IR-induced hypoxia inducible factor 1 subunit alpha (HIF-1α) is a predominant mediator of EZH2 to repress ACSL4; (3) the HRE7-8 regions of the ACSL4 promoter are required for the repressive function of EZH2 on ACSL4; (4) EZH2 regulates ACSL4 by forming a co-repressive complex with HIF-1α. Our study provides preclinical evidence supporting that EZH2 inhibitors may confer therapeutic benefit in KDM6A-deficient ESCC patients.
Collapse
Affiliation(s)
- Guizhen Pan
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yeye Xia
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Oncology, Chengdu Fifth People's Hospital, Chengdu, China
| | - Mengyu Hao
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jiahao Guan
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qianqian Zhu
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Radiation Oncology, Fuyang Tumour Hospital, Fuyang, China
| | - Tianqi Zha
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lei Sheng
- Department of Radiation Oncology, the Chest Hospital of Anhui Province, Hefei, Anhui, China
| | - Zhenfeng Zhao
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Huaguang Pan
- Department of Thoracic Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Weiyang Fang
- Department of Electrocardiography, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiaoyong Xu
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiangcun Chen
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shuguang Zhou
- The Fifth Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Zhuting Tong
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
3
|
Sun LWH, Asana Marican HT, Shen H. In Vivo Imaging of Radiation-Induced Apoptosis at Single-Cell Resolution in Transgenic Zebrafish Embryos. Radiat Res 2023; 199:229-239. [PMID: 36745564 DOI: 10.1667/rade-22-00174.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/17/2023] [Indexed: 02/07/2023]
Abstract
Among the various types of cell death induced by ionizing radiation, apoptosis is a highly regulated and well-characterized form. Investigating radiation-induced apoptosis in an intact organism offers advantages in capturing the dynamics of apoptosis under preserved physiology, although high resolution imaging remains challenging. Owing to their optical transparency and genetic amenability, zebrafish is an ideal animal model for research into this aspect. In this study, we present a secA5 transgenic zebrafish expressing genetically encoded secreted ANNEXIN V fused with mVenus, a yellow fluorescent protein that enables reporting of radiation-induced apoptosis. Using in vivo imaging approach, we show that after 2 Gy total-body irradiation, apoptosis could be visualized at single-cell resolution in different cell types throughout the embryo. Elevated apoptosis could be imaged and quantified in the neuroepithelium of the embryonic brain, as well as the proliferative zone and parenchyma of the larval brain. In addition, clearance of apoptotic cells by microglia, the professional phagocytes residing in the brain, could be imaged at single-cell resolution in irradiated larvae. These results establish transgenic secA5 zebrafish as a useful and versatile in vivo system for investigating the dynamic process of radiation-induced apoptosis.
Collapse
Affiliation(s)
| | | | - Hongyuan Shen
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| |
Collapse
|
4
|
Tong Z, Fang W, Xu M, Xia Y, Wang R, Li Y, Zha T, Xiao L, Pan S, Chai H, Zhao L, Wang H, Pan H, Chen X. DAB2IP predicts treatment response and prognosis of ESCC patients and modulates its radiosensitivity through enhancing IR-induced activation of the ASK1-JNK pathway. Cancer Cell Int 2022; 22:106. [PMID: 35248066 PMCID: PMC8897861 DOI: 10.1186/s12935-022-02535-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/24/2022] [Indexed: 12/24/2022] Open
Abstract
Background Disabled homolog 2 interacting protein (DAB2IP) plays a tumor-suppressive role in several types of human cancers. However, the molecular status and function of the DAB2IP gene in esophageal squamous cell carcinoma (ESCC) patients who received definitive chemoradiotherapy is rarely reported. Methods We examined the expression dynamics of DAB2IP by immunohistochemistry (IHC) in 140 ESCC patients treated with definitive chemoradiotherapy. A series of in vivo and in vitro experiments were performed to elucidate the effect of DAB2IP on the chemoradiotherapy (CRT) response and its underlying mechanisms in ESCC. Results Decreased expression of DAB2IP in ESCCs correlated positively with ESCC resistance to CRT and was a strong and independent predictor for short disease-specific survival (DSS) of ESCC patients. Furthermore, the therapeutic sensitivity of CRT was substantially increased by ectopic overexpression of DAB2IP in ESCC cells. In addition, knockdown of DAB2IP dramatically enhanced resistance to CRT in ESCC. Finally, we demonstrated that DAB2IP regulates ESCC cell radiosensitivity through enhancing ionizing radiation (IR)-induced activation of the ASK1-JNK signaling pathway. Conclusions Our data highlight the molecular etiology and clinical significance of DAB2IP in ESCC, which may represent a new therapeutic strategy to improve therapy and survival for ESCC patients.
Collapse
|
5
|
Szejk-Arendt M, Czubak-Prowizor K, Macieja A, Poplawski T, Olejnik AK, Pawlaczyk-Graja I, Gancarz R, Zbikowska HM. Polyphenolic-polysaccharide conjugates from medicinal plants of Rosaceae/Asteraceae family protect human lymphocytes but not myeloid leukemia K562 cells against radiation-induced death. Int J Biol Macromol 2020; 156:1445-1454. [DOI: 10.1016/j.ijbiomac.2019.11.186] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/14/2019] [Accepted: 11/21/2019] [Indexed: 01/21/2023]
|
6
|
Murray D, Mirzayans R. Nonlinearities in the cellular response to ionizing radiation and the role of p53 therein. Int J Radiat Biol 2020; 97:1088-1098. [PMID: 31986075 DOI: 10.1080/09553002.2020.1721602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/06/2019] [Accepted: 12/26/2019] [Indexed: 10/25/2022]
Abstract
Many aspects of the cellular response to agents such as ionizing radiation that cause genotoxic and/or oxidative stress exhibit a nonlinear relationship to the applied stress level. These include elements of the antioxidant response and of the damage-signaling pathways that determine cell fate decisions. The wild-type p53 protein, which is mutated in many cancers, coordinates these responses and is a key determinant of this nonlinearity. Indeed, p53 has been referred to as a 'cellular rheostat' that favors antioxidant/cytoprotective functions at low stress levels while switching to a pro-oxidant/cytotoxic role under high-stress conditions. For solid tumor-derived cell lines, moderate doses of radiation, typical of those used to generate clonogenic survival curves (i.e. ≤10 Gy), predominantly invoke a dose-dependent cytostatic response. For cancer cell lines with wild-type p53, cytostasis is primarily associated with features of senescence, whereas cancer cells with aberrant p53 primarily undergo endopolyploidization and enlargement. In line with a commentary by Meyn et al. [Int J Radiat Biol. 2009, 85:107-115] concluding that apoptosis is not the primary cause of radiation-induced loss of clonogenicity in solid tumor-derived cell lines, significant levels of apoptosis are typically seen only after higher doses (≥5 Gy) and this is almost all of the delayed (rather than primary) type. Nonlinearity of the oxidative/genotoxic stress response is already apparent in the early antioxidant events activated by transcription factors such as p53 and Nrf2 and the Ref1 transcription coactivator. These cytoprotective pathways serve to minimize damage to important cellular targets caused by reactive oxygen species (ROS) and other electrophiles. After high/supra-lethal levels of stress these inducible antioxidant pathways can be deactivated in a manner that would reinforce the establishment of the pro-oxidant state, resulting in elevated ROS levels and to cytostasis or apoptosis. Understanding the complex regulation of these damage-signaling pathways in relation to the stress levels is important for the optimal utilization of radiation therapy for cancer.
Collapse
Affiliation(s)
- David Murray
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| | - Razmik Mirzayans
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
7
|
Mortensen ACL, Spiegelberg D, Brown CJ, Lane DP, Nestor M. The Stapled Peptide PM2 Stabilizes p53 Levels and Radiosensitizes Wild-Type p53 Cancer Cells. Front Oncol 2019; 9:923. [PMID: 31616635 PMCID: PMC6764291 DOI: 10.3389/fonc.2019.00923] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/03/2019] [Indexed: 12/30/2022] Open
Abstract
The tumor suppressor p53 is a key mediator of cellular stress and DNA damage response cascades and is activated after exposure to ionizing radiation. Amplifying wild-type p53 expression by targeting negative regulators such as MDM2 in combination with external beam radiotherapy (EBRT) may result in increased therapeutic effects. The novel stapled peptide PM2 prevents MDM2 from suppressing wild-type p53, and is thus a promising agent for therapeutic combination with EBRT. Effects of PM2 and potential PM2-induced radiosensitivity were assessed in a panel of cancer cell lines using 2D cell viability assays. Western Blot and flow cytometric analyses were used to investigate the mechanisms behind the observed effects in samples treated with PM2 and EBRT. Finally, PM2-treatment combined with EBRT was evaluated in an in vitro 3D spheroid model. PM2-therapy decreased cell viability in wild-type p53, HPV-negative cell lines. Western Blotting and flow cytometry confirmed upregulation of p53, as well as initiation of p53-mediated apoptosis measured by increased cleaved caspase-3 and Noxa activity. Furthermore, 3D in vitro tumor spheroid experiments confirmed the superior effects of the combination, as the only treatment regime resulting in growth inhibition and complete spheroid disintegration. We conclude that PM2 induces antitumorigenic effects in wt p53 HPV-negative cancer cells and potentiates the effects of EBRT, ultimately resulting in tumor eradication in a 3D spheroid model. This strategy shows great potential as a new wt p53 specific tumor-targeting compound, and the combination of PM2 and EBRT could be a promising strategy to increase therapeutic effects and decrease adverse effects from radiotherapy.
Collapse
Affiliation(s)
| | - Diana Spiegelberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | | | - David Philip Lane
- Agency for Science, Technology and Research (ASTAR), Singapore, Singapore.,Science for Life Laboratory, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Marika Nestor
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
8
|
Zhao Z, Zhou S, Li W, Zhong F, Zhang H, Sheng L, Li Y, Xu M, Xu J, Zhan L, Li B, Wang F, Xie D, Tong Z. AIB1 predicts tumor response to definitive chemoradiotherapy and prognosis in cervical squamous cell carcinoma. J Cancer 2019; 10:5212-5222. [PMID: 31602272 PMCID: PMC6775615 DOI: 10.7150/jca.31697] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 07/27/2019] [Indexed: 12/16/2022] Open
Abstract
Amplified in breast cancer 1 (AIB1) gene, has been reported to be associated with biological malignancy in several cancers. However, the molecular status of the AIB1 gene in cervical cancer and the clinicopathological/prognostic significance of AIB1 expression in chemoradiotherapy (CRT) sensitivity have not been determined. In our present study, we found that the high expression of AIB1 was frequent detected in specimens of cervical cancer patients, and this was significantly correlated with CRT response (P = 0.014), clinical stage (P = 0.003), T status (P = 0.027), N status (P = 0.021), M status (P = 0.015) and progression-free survival (P < 0.001). Moreover, the clonogenic survival fraction and cell apoptosis experiments showed that knockdown of AIB1 substantially increased cervical cancer cells sensitivity to ionizing radiation (IR) or cisplatin/5-fluorouracil. Collectively, our results demonstrated that the high expression of AIB1 in cervical cancer cells contributes to the resistance to CRT, which provides the evidence that AIB1 may be a promising predictor of aggressive cervical cancer patients with poor response to CRT.
Collapse
Affiliation(s)
- Zhenfeng Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Radiation Oncology, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shuguang Zhou
- Department of Gynecology, Maternity and Child Healthcare Hospital of Anhui Medical University, Hefei, China
| | - Wenyu Li
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fei Zhong
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, Anhui, China
| | - Heping Zhang
- Department of Pathology, Maternity and Child Healthcare Hospital of Anhui Medical University, Hefei, China
| | - Lei Sheng
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yue Li
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Meng Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jifei Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Zhan
- Pathology Department of Anhui Medical University, Hefei, China
| | - Bao Li
- The Comprehensive Lab, College of Basic medicine, Anhui Medical University, Hefei, China
| | - Fan Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dan Xie
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhuting Tong
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
9
|
Apoptosis and cell proliferation in short-term and long-term effects of radioiodine-131-induced kidney damage: an experimental and immunohistochemical study. Nucl Med Commun 2018; 39:131-139. [PMID: 29257007 DOI: 10.1097/mnm.0000000000000788] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Radioiodine-131 is a radionuclide that is used for therapeutic purposes in hyperthyroidism and thyroid cancer. The aim of this study was to evaluate apoptotosis and proliferative changes in radioiodine-related kidney damage. MATERIALS AND METHODS Three groups (n=10/group) of rats were used as follows: the rats were in group 1 untreated, and the rats in groups 2 and 3 were treated once with oral radioiodine (111 MBq). The animals in group 2 were killed at the end of the seventh day and the rats in group 3 were killed at the end of the 10th week. The kidneys were removed and evaluated immunohistochemically. The presence of radioiodine in the kidneys was shown by the Na+/I-symporter antibody and proliferating cell nuclear antigen, Ki-67, caspase-3, caspase-8, caspase-9, and terminal deoxynucleotidyl transferase mediated deoxyuridine triphosphate nick end labeling assay were used to detect cell proliferation and apoptosis. RESULTS Na+/I-symporter protein accumulation in the kidneys was observed to be significantly greater in group 2 than in group 3 (P<0.05). All the immunohistochemical analyses showed that cell proliferation and apoptosis began on the seventh day and peaked in the 10th week. The proliferating cell nuclear antigen, Ki-67, and caspase expressions and terminal deoxynucleotidyl transferase mediated deoxyuridine triphosphate nick end labeling values were all found to be statistically significantly increased in group 3 compared with the other groups (P<0.05). CONCLUSION Radioiodine caused cell proliferation and apoptosis as shown by immunohistochemistry.
Collapse
|
10
|
Zhou T, Cai W, Yang H, Zhang H, Hao M, Yuan L, Liu J, Zhang L, Yang Y, Liu X, Deng J, Zhao P, Yang G, Duan Y. Annexin V conjugated nanobubbles: A novel ultrasound contrast agent for in vivo assessment of the apoptotic response in cancer therapy. J Control Release 2018. [PMID: 29522835 DOI: 10.1016/j.jconrel.2018.03.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In vivo assessment of apoptotic response to cancer therapy is believed to be very important for optimizing management of treatment. However, few noninvasive strategies are currently available to monitor the therapeutic response in vivo. Ultrasonography has been used to detect apoptotic cell death in vivo, but a high-frequency transducer is needed. Fortunately, the capability of ultrasound contrast agents (UCAs) to exit the leaky vasculature of tumors enables ultrasound-targeted imaging of molecular events in response to cancer therapy. In this study, we prepared a novel nano-sized UCA, namely, Annexin V-conjugated nanobubbles (AV-NBs, 635.5 ± 25.4 nm). In vitro studies revealed that AV-NBs were relatively stable and highly echogenic. Moreover, these AV-NBs could easily extravasate into the tumor vasculature and recognize the apoptotic cells with high specificity and affinity in tumors sensitive to chemotherapy. Ultrasound imaging results demonstrated that AV-NBs had higher echogenicity and significantly greater enhancement compared with the untargeted control NBs (P < 0.01) inside the tumors after chemotherapy. Taken together, this study provides a promising method to accurately evaluate therapeutic effects at the molecular level to support cancer management.
Collapse
Affiliation(s)
- Tian Zhou
- Department of Ultrasound Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China; Department of Ultrasound Diagnosis, General Hospital of the PLA Rocket Force, Beijing 100088, China
| | - Wenbin Cai
- Department of Ultrasound Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Hengli Yang
- Department of Ultrasound Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Huizhong Zhang
- Department of Medical Laboratory and Research Center, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Minghua Hao
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Lijun Yuan
- Department of Ultrasound Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Jie Liu
- Department of Ultrasound Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Li Zhang
- Department of Ultrasound Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Yilin Yang
- Department of Ultrasound Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Xi Liu
- Department of Ultrasound Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | - Jianling Deng
- Department of Ultrasound Diagnosis, General Hospital of the PLA Rocket Force, Beijing 100088, China
| | - Ping Zhao
- Department of Ultrasound Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China.
| | - Guodong Yang
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China.
| | - Yunyou Duan
- Department of Ultrasound Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China.
| |
Collapse
|
11
|
Alongi F, Di Muzio N. Radiobiology and Molecular Oncology: How are they Changing Radiotherapy in Clinical Practice? TUMORI JOURNAL 2018; 96:175-7. [DOI: 10.1177/030089161009600131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Filippo Alongi
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Italy
- Radiotherapy, Scientific institute San Raffaele, Milan, Italy
- Intraoperative Radiotherapy, Breast Unit, San Raffaele-G. Giglio Foundation, Cefalù, Italy
| | - Nadia Di Muzio
- Radiotherapy, Scientific institute San Raffaele, Milan, Italy
| |
Collapse
|
12
|
Purgason A, Zhang Y, Hamilton SR, Gridley DS, Sodipe A, Jejelowo O, Ramesh GT, Moreno-Villanueva M, Wu H. Apoptosis and expression of apoptosis-related genes in mouse intestinal tissue after whole-body proton exposure. Mol Cell Biochem 2017; 442:155-168. [DOI: 10.1007/s11010-017-3200-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/06/2017] [Indexed: 12/11/2022]
|
13
|
Paths from DNA damage and signaling to genome rearrangements via homologous recombination. Mutat Res 2017; 806:64-74. [PMID: 28779875 DOI: 10.1016/j.mrfmmm.2017.07.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 07/21/2017] [Indexed: 12/31/2022]
Abstract
DNA damage is a constant threat to genome integrity. DNA repair and damage signaling networks play a central role maintaining genome stability, suppressing tumorigenesis, and determining tumor response to common cancer chemotherapeutic agents and radiotherapy. DNA double-strand breaks (DSBs) are critical lesions induced by ionizing radiation and when replication forks encounter damage. DSBs can result in mutations and large-scale genome rearrangements reflecting mis-repair by non-homologous end joining or homologous recombination. Ionizing radiation induces genetic change immediately, and it also triggers delayed events weeks or even years after exposure, long after the initial damage has been repaired or diluted through cell division. This review covers DNA damage signaling and repair pathways and cell fate following genotoxic insult, including immediate and delayed genome instability and cell survival/cell death pathways.
Collapse
|
14
|
de Bruin DM, Broekgaarden M, van Gemert MJC, Heger M, de la Rosette JJ, Van Leeuwen TG, Faber DJ. Assesment of apoptosis induced changes in scattering using optical coherence tomography. JOURNAL OF BIOPHOTONICS 2016; 9:913-923. [PMID: 26564260 DOI: 10.1002/jbio.201500198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/28/2015] [Accepted: 10/18/2015] [Indexed: 06/05/2023]
Abstract
The aim of this study is to identify changes in scattering with optical coherence tomography (OCT) and relate these measurements with mitochondrial changes during the initiation of apoptosis. Human retinal pigment epithelial cells were cultured and apoptosis was induced using 10% alcohol. Using the attenuation coefficient and backscattering, changes were measured during cell death in a cell-pellet and monolayer respectively. To confirm apoptosis, fluorescent activated cell sorting was used. Mitochondrial activity during apoptosis was assessed using an oxidative stress assay and fluorescent confocal microscopy. Pelleted apoptotic cells measured with OCT showed a clear rise while untreated cells showed a very small increase in attenuation coefficient. Monolayered apoptotic cells displayed a distinct increase, while untreated cells showed a small increase in the backscattering. Apoptosis was confirmed by FACS experiments. Mitochondrial changes during the onset of apoptosis were also measured. The results demonstrate that apoptotic cell death could be monitored in real-time by OCT. Changes in the scattering after induction of apoptosis are likely to be related to changes in the intracellular morphology. Oxidative stress-induced mitochondrial swelling could be responsible for the initial increase, while cell blebbing and secondary necrosis subsequently for the observed decrease in scattering.
Collapse
Affiliation(s)
- Daniel M de Bruin
- Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, Netherlands.
- Urology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, Netherlands.
| | - Mans Broekgaarden
- Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, Netherlands
| | - Martin J C van Gemert
- Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, Netherlands
| | - Michal Heger
- Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, Netherlands
| | - Jean J de la Rosette
- Urology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, Netherlands
| | - Ton G Van Leeuwen
- Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, Netherlands
| | - Dirk J Faber
- Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, Netherlands
| |
Collapse
|
15
|
Brooks AL, Hoel DG, Preston RJ. The role of dose rate in radiation cancer risk: evaluating the effect of dose rate at the molecular, cellular and tissue levels using key events in critical pathways following exposure to low LET radiation. Int J Radiat Biol 2016; 92:405-26. [PMID: 27266588 PMCID: PMC4975094 DOI: 10.1080/09553002.2016.1186301] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/14/2016] [Accepted: 05/02/2016] [Indexed: 12/19/2022]
Abstract
PURPOSE This review evaluates the role of dose rate on cell and molecular responses. It focuses on the influence of dose rate on key events in critical pathways in the development of cancer. This approach is similar to that used by the U.S. EPA and others to evaluate risk from chemicals. It provides a mechanistic method to account for the influence of the dose rate from low-LET radiation, especially in the low-dose region on cancer risk assessment. Molecular, cellular, and tissues changes are observed in many key events and change as a function of dose rate. The magnitude and direction of change can be used to help establish an appropriate dose rate effectiveness factor (DREF). CONCLUSIONS Extensive data on key events suggest that exposure to low dose-rates are less effective in producing changes than high dose rates. Most of these data at the molecular and cellular level support a large (2-30) DREF. In addition, some evidence suggests that doses delivered at a low dose rate decrease damage to levels below that observed in the controls. However, there are some data human and mechanistic data that support a dose-rate effectiveness factor of 1. In summary, a review of the available molecular, cellular and tissue data indicates that not only is dose rate an important variable in understanding radiation risk but it also supports the selection of a DREF greater than one as currently recommended by ICRP ( 2007 ) and BEIR VII (NRC/NAS 2006 ).
Collapse
Affiliation(s)
- Antone L. Brooks
- Retired Professor, Environmental Science, Washington State University,
Richland,
Washington,
USA
| | - David G. Hoel
- Medical University of South Carolina, Epidemiology,
Charleston South Carolina,
USA
| | - R. Julian Preston
- US Environmental Protection Agency, National Health and Environmental Effects Research Laboratory (NHEERL) (MD B105-01), RTP,
USA
| |
Collapse
|
16
|
Elvas F, Vangestel C, Pak K, Vermeulen P, Gray B, Stroobants S, Staelens S, Wyffels L. Early Prediction of Tumor Response to Treatment: Preclinical Validation of 99mTc-Duramycin. J Nucl Med 2016; 57:805-11. [PMID: 26837335 DOI: 10.2967/jnumed.115.168344] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 11/29/2015] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Noninvasive imaging of cell death can provide an early indication of the efficacy of tumor treatment, aiding clinicians in distinguishing responding patients from nonresponding patients early on. (99m)Tc-duramycin is a SPECT tracer for cell death imaging. In this study, our aim was to validate the use of (99m)Tc-duramycin for imaging the early response of tumors to treatment. METHODS An in vitro binding assay was performed on COLO205 cells treated with 5-fluorouracil (3.1, 31, or 310 μM) and oxaliplatin (0.7 or 7 μM) or radiation (2 or 4.5 Gy). (99m)Tc-duramycin cell binding and the levels of cell death were evaluated after treatment. In vivo imaging was performed on 4 groups of CD1-deficient mice bearing COLO205 human colorectal cancer tumors. Each group included 6 tumors. The first group was given irinotecan (100 mg/kg), the second oxaliplatin (5 mg/kg), the third irinotecan (80 mg/kg) plus oxaliplatin (5 mg/kg), and the fourth vehicle (0.9% NaCl and 5% glucose). For radiotherapy studies, COLO205 tumors received 4.5 Gy, 2 fractions of 4.5 Gy in a 24-h interval, pretreatment with an 80 mg/kg dose of irinotecan combined with 2 fractions of 4.5 Gy in a 24-h interval, or no treatment (n = 5-6/group). Therapy response was evaluated by (99m)Tc-duramycin SPECT 24 h after the last dose of therapy. Blocking was used to confirm tracer specificity. Radiotracer uptake in the tumors was validated ex vivo using γ-counting, cleaved caspase-3, and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) histology. RESULTS Chemotherapy and radiotherapy increased (99m)Tc-duramycin binding to COLO205 cells in a concentration/dose- and time-dependent manner, which correlated well with cell death levels (P < 0.05) as analyzed by annexin V and caspase 3/7 activity. In vivo, (99m)Tc-duramycin uptake in COLO205 xenografts was increased 2.3- and 2.8-fold (P < 0.001) in mice treated with irinotecan and combination therapy, respectively. Blocking with unlabeled duramycin demonstrated specific binding of the radiotracer. After tumor irradiation with 4.5 Gy, (99m)Tc-duramycin uptake in tumors increased significantly (1.24 ± 0.07 vs. 0.57 ± 0.08 percentage injected dose per gram in the unirradiated tumors; P < 0.001). γ-counting of radioactivity in the tumors positively correlated with cleaved caspase-3 (r = 0.85, P < 0.001) and TUNEL (r = 0.81, P < 0.001) staining. CONCLUSION We demonstrated that (99m)Tc-duramycin can be used to image induction of cell death early after chemotherapy and radiotherapy. It holds potential to be translated into clinical use for early assessment of treatment response.
Collapse
Affiliation(s)
- Filipe Elvas
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium Department of Nuclear Medicine, University Hospital Antwerp, Edegem, Belgium
| | - Christel Vangestel
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium Department of Nuclear Medicine, University Hospital Antwerp, Edegem, Belgium
| | - Koon Pak
- Molecular Targeting Technologies, Inc., West Chester, Pennsylvania; and
| | - Peter Vermeulen
- Laboratory of Pathology, General Hospital Sint-Augustinus, Antwerp, Belgium
| | - Brian Gray
- Molecular Targeting Technologies, Inc., West Chester, Pennsylvania; and
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium Department of Nuclear Medicine, University Hospital Antwerp, Edegem, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium
| | - Leonie Wyffels
- Molecular Imaging Center Antwerp, University of Antwerp, Wilrijk, Belgium Department of Nuclear Medicine, University Hospital Antwerp, Edegem, Belgium
| |
Collapse
|
17
|
Sun Y, Liu JH, Jin L, Sui YX, Han LL, Huang Y. Effect of autophagy-related beclin1 on sensitivity of cisplatin-resistant ovarian cancer cells to chemotherapeutic agents. Asian Pac J Cancer Prev 2016; 16:2785-91. [PMID: 25854363 DOI: 10.7314/apjcp.2015.16.7.2785] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The purpose of the study was to determine the effects of autophagy related gene Beclin1 at different levels of expression on the sensitivity of cisplatin-resistant ovarian cancer cells (SKOV3/DDP) to different chemotherapeutics. In pSUPER-Beclin1 transfected cells, real-time fluorescence quantitative RT-PCR and Western blot analysis showed that expression was significantly inhibited. Flow cytometry revealed that the mean fluorescence intensity (MDC), reflecting autophagy, and cells in the G0/G1 phase were markedly reduced. When compared with the blank control group, inhibition of Beclin1 expression in SKOV3/DDP cells not only increased the rate of apoptosis following treatment with chemotherapeutics, but also increased the sensitivity. These findings suggest that Beclin1 expression plays an important role in chemotherapeutic agent-induced death of SKOV3/DDP cells. Inhibition of autophagy related gene Beclin1 expression in SKOV3/DDP cells may increase the rate of apoptosis and elevate the sensitivity to chemotherapeutics.
Collapse
Affiliation(s)
- Yang Sun
- Department of Gynecology, Fujian Provincial Hospital, Fujian Provincial Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China E-mail :
| | | | | | | | | | | |
Collapse
|
18
|
Belhocine TZ, Blankenberg FG, Kartachova MS, Stitt LW, Vanderheyden JL, Hoebers FJP, Van de Wiele C. (99m)Tc-Annexin A5 quantification of apoptotic tumor response: a systematic review and meta-analysis of clinical imaging trials. Eur J Nucl Med Mol Imaging 2015; 42:2083-97. [PMID: 26275392 DOI: 10.1007/s00259-015-3152-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 07/20/2015] [Indexed: 12/31/2022]
Abstract
PURPOSE (99m)Tc-Annexin A5 has been used as a molecular imaging probe for the visualization, characterization and measurement of apoptosis. In an effort to define the quantitative (99m)Tc-annexin A5 uptake criteria that best predict tumor response to treatment, we performed a systematic review and meta-analysis of the results of all clinical imaging trials found in the literature or publicly available databases. METHODS Included in this review were 17 clinical trials investigating quantitative (99m)Tc-annexin A5 (qAnx5) imaging using different parameters in cancer patients before and after the first course of chemotherapy and/or radiation therapy. Qualitative assessment of the clinical studies for diagnostic accuracy was performed using the QUADAS-2 criteria. Of these studies, five prospective single-center clinical trials (92 patients in total) were included in the meta-analysis after exclusion of one multicenter clinical trial due to heterogeneity. Pooled positive predictive values (PPV) and pooled negative predictive values (NPV) (with 95% CI) were calculated using Meta-Disc software version 1.4. RESULTS Absolute quantification and/or relative quantification of (99m)Tc-annexin A5 uptake were performed at baseline and after the start of treatment. Various quantitative parameters have been used for the calculation of (99m)Tc-annexin A5 tumor uptake and delta (Δ) tumor changes post-treatment compared to baseline including: tumor-to-background ratio (TBR), ΔTBR, tumor-to-noise ratio, relative tumor ratio (TR), ΔTR, standardized tumor uptake ratio (STU), ΔSTU, maximum count per pixel within the tumor volume (Cmax), Cmax%, absolute ΔU and percentage (ΔU%), maximum ΔU counts, semiquantitative visual scoring, percent injected dose (%ID) and %ID/cm(3). Clinical trials investigating qAnx5 imaging have included patients with lung cancer, lymphoma, breast cancer, head and neck cancer and other less common tumor types. In two phase I/II single-center clinical trials, an increase of ≥25% in uptake following treatment was considered a significant threshold for an apoptotic tumor response (partial response, complete response). In three other phase I/II clinical trials, increases of ≥28%, ≥42% and ≥47% in uptake following treatment were found to be the mean cut-off levels in responders. In a phase II/III multicenter clinical trial, an increase of ≥23% in uptake following treatment was found to be the minimum cut-off level for a tumor response. In one clinical trial, no significant difference in (99m)Tc-annexin A5 uptake in terms of %ID was found in healthy tissues after chemotherapy compared to baseline. In two other clinical trials, intraobserver and interobserver measurements of (99m)Tc-annexin A5 tumor uptake were found to be reproducible (mean difference <5%, kappa = 0.90 and 0.82, respectively) and to be highly correlated with treatment outcome (Spearman r = 0.99, p < 0.0001). The meta-analysis demonstrated a pooled positive PPV of 100% (95% CI 92 - 100%) and a pooled NPV of 70% (95% CI 55 - 82%) for prediction of a tumor response after the first course of chemotherapy and/or radiotherapy in terms of ΔU%. In a symmetric sROC analysis, the AUC was 0.919 and the Q* index was 85.21 %. CONCLUSION Quantitative (99m)Tc-annexin A5 imaging has been investigated in clinical trials for the assessment of apoptotic tumor responses. This meta-analysis showed a high pooled PPV and a moderate pooled NPV with ΔU cut-off values ranging between 20% and 30%. Standardization of quantification and harmonization of results are required for high-quality clinical research. A standardized uptake value score (SUV, ΔSUV) using quantitative SPECT/CT imaging may be a promising approach to the simple, reproducible and semiquantitative assessment of apoptotic tumor changes.
Collapse
Affiliation(s)
- Tarik Z Belhocine
- Biomedical Imaging Research Centre (BIRC), Western University, London, Ontario, Canada.
| | - Francis G Blankenberg
- Division of Pediatric Radiology, Department of Radiology, Lucile Salter Packard Children's Hospital, Stanford, Palo Alto, CA, USA
| | - Marina S Kartachova
- Department of Nuclear Medicine, Medical Center Alkmaar, Alkmaar, The Netherlands
| | - Larry W Stitt
- LW Stitt Statistical Services, London, Ontario, Canada
| | | | - Frank J P Hoebers
- Department of Radiation Oncology (MAASTRO Clinic), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | | |
Collapse
|
19
|
Rusinova GG, Vyazovskaya NS, Azizova TV, Revina VS, Glazkova IV, Generozov EV, Zakharzhevskaya NB, Guryanov MY, Belosokhov MV, Osovets SV. [Molecular mechanisms of lung cancer development at its different stages in nuclear industry workers]. Arkh Patol 2015; 77:10-15. [PMID: 26027393 DOI: 10.17116/patol201577210-15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE to assess mutational events in exons 5, 7, and 8 of the p53 gene and to reveal mutant p53 protein in verified cases of morphologically altered (proliferative and precancerous changes, lung cancer) and histologically unaltered, lung tissues in workers exposed to occupational radiation. MATERIAL AND METHODS The investigation used formalin-fixed paraffin-embedded unaltered and altered lung tissue blocks (FFPBs) obtained from the human radiobiological tissue repository. The shelf-life of FFPBs was 5-31 years. An immunohistochemical technique using mouse antibodies against p53 protein (<<DAKO>>, Denmark), stained with diaminobenzidine (DAB) chromogen, was employed to determine p53 protein. DNA was isolated from lung tissue FFPBs with QIAmp DNA FFPE Tissue Kit, (<<QIAGEN>>, USA). Polymerase chain reaction (PCR) was performed to amplify the p53 gene exons 5, 7, and 8 selected for examination, by applying the sequences of genes and primers, the specificity of which was checked using the online resource (http://www.ncbi.nlm.nih.gov/blast). PCR products were detected by temporal temperature gradient gel-electrophoresis and the Sanger sequencing method. The obtained DNA fragments were analyzed on a sequencer ABI Prism 3100 Genetic Analizer (<<Applied Biosystems>>, USA). Computer-aided DNA analysis was made using the BLAST program. A package of applied Statistica 6.0 programs was employed for statistical data processing. Results. Immunohistochemical analysis showed that mutant p53 protein was absent in the cells of unaltered lung tissue and the number of cells with mutant p53 protein increased in all the patients with proliferative and precancerous changes and lung cancer, suggesting p53 protein dysfunction. The total number of p53 gene mutations in exons 5, 7, and 8, if there were proliferative and precancerous lung tissue changes and lung cancer, were 25, 20, and 40%, respectively. All the found mutations were transversions (the substitution of purine for pyrimidine or, conversely), indicating the action of exogenous mutagens. CONCLUSION The results of this investigation have confirmed other investigators' data showing that p53 gene mutations in lung cancer are observed in 40-70% of cases. The differences in the number of cases of altered lung tissue with mutations in the p53 gene (not more than 40%) and in those of p53 protein expression were found in 100%, suggesting the regulation of p53 gene function in the cell at multiple levels.
Collapse
Affiliation(s)
- G G Rusinova
- South Ural Institute of Biophysics, Ozersk, Сhelyabinsk Region
| | - N S Vyazovskaya
- South Ural Institute of Biophysics, Ozersk, Сhelyabinsk Region
| | - T V Azizova
- South Ural Institute of Biophysics, Ozersk, Сhelyabinsk Region
| | - V S Revina
- South Ural Institute of Biophysics, Ozersk, Сhelyabinsk Region
| | - I V Glazkova
- South Ural Institute of Biophysics, Ozersk, Сhelyabinsk Region
| | - E V Generozov
- Laboratory of Human Molecular Genetics, Research Institute of Physicochemical Medicine, Ministry of Health of Russia, Moscow, Russian Federation
| | - N B Zakharzhevskaya
- Laboratory of Human Molecular Genetics, Research Institute of Physicochemical Medicine, Ministry of Health of Russia, Moscow, Russian Federation
| | - M Yu Guryanov
- South Ural Institute of Biophysics, Ozersk, Сhelyabinsk Region
| | - M V Belosokhov
- South Ural Institute of Biophysics, Ozersk, Сhelyabinsk Region
| | - S V Osovets
- South Ural Institute of Biophysics, Ozersk, Сhelyabinsk Region
| |
Collapse
|
20
|
Bloy N, Pol J, Manic G, Vitale I, Eggermont A, Galon J, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Radioimmunotherapy for oncological indications. Oncoimmunology 2014; 3:e954929. [PMID: 25941606 DOI: 10.4161/21624011.2014.954929] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 07/18/2014] [Indexed: 02/06/2023] Open
Abstract
During the past two decades, it has become increasingly clear that the antineoplastic effects of radiation therapy do not simply reflect the ability of X-, β- and γ-rays to damage transformed cells and directly cause their permanent proliferative arrest or demise, but also involve cancer cell-extrinsic mechanisms. Indeed, among other activities, radiotherapy has been shown to favor the establishment of tumor-specific immune responses that operate systemically, underpinning the so-called 'out-of-field' or 'abscopal' effect. Thus, ionizing rays appear to elicit immunogenic cell death, a functionally peculiar variant of apoptosis associated with the emission of a particularly immunostimulatory combination of damage-associated molecular patterns. In line with this notion, radiation therapy fosters, and thus exacerbates, the antineoplastic effects of various treatment modalities, including surgery, chemotherapy and various immunotherapeutic agents. Here, we summarize recent advances in the use of ionizing rays as a means to induce or potentiate therapeutically relevant anticancer immune responses. In addition, we present clinical trials initiated during the past 12 months to test the actual benefit of radioimmunotherapy in cancer patients.
Collapse
Affiliation(s)
- Norma Bloy
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris-Sud/Paris XI ; Paris, France
| | - Jonathan Pol
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France
| | - Gwenola Manic
- Regina Elena National Cancer Institute ; Rome, Italy
| | - Ilio Vitale
- Regina Elena National Cancer Institute ; Rome, Italy
| | | | - Jérôme Galon
- INSERM, U1138 ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers ; Paris, France
| | - Eric Tartour
- Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; INSERM, U970 ; Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP ; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1015; CICBT507 ; Villejuif, France
| | - Guido Kroemer
- INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP ; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus ; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus ; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France
| |
Collapse
|
21
|
Wang XY, Ma ZC, Wang YG, Tan HL, Xiao CR, Liang QD, Tang XL, Cheng Y, Gao Y. Tetramethylpyrazine protects lymphocytes from radiation-induced apoptosis through nuclear factor-κB. Chin J Nat Med 2014; 12:730-7. [PMID: 25443365 DOI: 10.1016/s1875-5364(14)60112-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Indexed: 11/18/2022]
Abstract
AIM Radiation induces an important apoptosis response in irradiated organs. The objective of this study was to investigate the radioprotective effect of tetramethylpyrazine (TMP) on irradiated lymphocytes and discover the possible mechanism of protection. METHOD Lymphocytes were pretreated for 12 h with TMP (25-200 μmol·L(-1)) and then exposed to 4 Gy radiation. Cell apoptosis and the signaling pathway were analyzed. RESULTS Irradiation increased cell death, DNA fragmentation, activated caspase activation and cytochrome c translocation, downregulated B-cell lymphoma 2 (Bcl-2) and up-regulated Bcl-2-associated X protein (Bax). Pretreated with TMP significantly reversed this tendency. Several anti-apoptotic characteristics of TMP, including the ability to increase cell viability, inhibit caspase-9 activation, and upregulate Bcl-2 and down-regulate Bax in 4Gy-irradiated lymphocytes were determined. Signal pathway analysis showed TMP could translate nuclear factor-κB (NF-κB) from cytosol into the nucleus. CONCLUSION The results suggest that TMP had a radioprotective effect through the NF-κB pathway to inhibit apoptosis, and it may be an effective candidate for treating radiation diseases associated with cell apoptosis.
Collapse
Affiliation(s)
- Xiao-Yan Wang
- Institute of Radiation Medicine, Academy of Military Medical Science, Beijing 100850, China; Chongqing Maternal and Child Health-Care Hospital, Chongqing 400016, China
| | - Zeng-Chun Ma
- Institute of Radiation Medicine, Academy of Military Medical Science, Beijing 100850, China.
| | - Yu-Guang Wang
- Institute of Radiation Medicine, Academy of Military Medical Science, Beijing 100850, China
| | - Hong-Ling Tan
- Institute of Radiation Medicine, Academy of Military Medical Science, Beijing 100850, China
| | - Cheng-Rong Xiao
- Institute of Radiation Medicine, Academy of Military Medical Science, Beijing 100850, China
| | - Qian-De Liang
- Institute of Radiation Medicine, Academy of Military Medical Science, Beijing 100850, China
| | - Xiang-Lin Tang
- Institute of Radiation Medicine, Academy of Military Medical Science, Beijing 100850, China
| | - Yu Cheng
- Chongqing Maternal and Child Health-Care Hospital, Chongqing 400016, China
| | - Yue Gao
- Institute of Radiation Medicine, Academy of Military Medical Science, Beijing 100850, China.
| |
Collapse
|
22
|
Human phosphatidylethanolamine-binding protein 4 promoted the radioresistance of human rectal cancer by activating Akt in an ROS-dependent way. PLoS One 2014; 9:e90062. [PMID: 24594691 PMCID: PMC3940727 DOI: 10.1371/journal.pone.0090062] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 01/28/2014] [Indexed: 12/28/2022] Open
Abstract
Human phosphatidylethanolamine-binding protein 4(hPEBP4) is a novel anti-apoptosis molecule associated with the resistance of tumors to apoptotic agents. Here we sought to investigate the role of hPEBP4 in the radioresistance of rectal cancer. Immunohistochemistry analysis showed hPEBP4 was expressed in 27/33 of rectal cancer specimens, but only in 2/33 of neighboring normal mucosa. Silencing the expression of hPEBP4 with siRNA significantly reduced the clonogenic survival and enhanced the apoptosis of rectal cancer cells on irradiation. Instead, forced overexpression of hPEBP4 promoted its survival and decreased the apoptosis. Western blot showed hPEBP4 could increase the radiation-induced Akt activation, for which reactive oxygen specimen(ROS) was required. The radioresistance effect of hPEBP4 was reversed after given LY-294002 to inhibit Akt activation or antioxidant to abolish the ROS production. We also confirmed that effect of hPEBP4 in vivo with nude mice. Thus we concluded that hPEBP4, specifically expressed in rectal cancer cells, is associated with radioresistance of rectal cancer, implying that modulation of hPEBP4 may have important therapeutic implications in radiotherapy of rectal cancer.
Collapse
|
23
|
Stegeman H, Span PN, Cockx SC, Peters JPW, Rijken PFJW, van der Kogel AJ, Kaanders JHAM, Bussink J. EGFR-Inhibition Enhances Apoptosis in Irradiated Human Head and Neck Xenograft Tumors Independent of Effects on DNA Repair. Radiat Res 2013; 180:414-21. [DOI: 10.1667/rr3349.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- H. Stegeman
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - P. N. Span
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - S. C. Cockx
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - J. P. W. Peters
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - P. F. J. W. Rijken
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - A. J. van der Kogel
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - J. H. A. M. Kaanders
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - J. Bussink
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Vacchelli E, Vitale I, Tartour E, Eggermont A, Sautès-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Anticancer radioimmunotherapy. Oncoimmunology 2013; 2:e25595. [PMID: 24319634 PMCID: PMC3850274 DOI: 10.4161/onci.25595] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 06/28/2013] [Indexed: 12/18/2022] Open
Abstract
Radiotherapy has extensively been employed as a curative or palliative intervention against cancer throughout the last century, with a varying degree of success. For a long time, the antineoplastic activity of X- and γ-rays was entirely ascribed to their capacity of damaging macromolecules, in particular DNA, and hence triggering the (apoptotic) demise of malignant cells. However, accumulating evidence indicates that (at least part of) the clinical potential of radiotherapy stems from cancer cell-extrinsic mechanisms, including the normalization of tumor vasculature as well as short- and long-range bystander effects. Local bystander effects involve either the direct transmission of lethal signals between cells connected by gap junctions or the production of diffusible cytotoxic mediators, including reactive oxygen species, nitric oxide and cytokines. Conversely, long-range bystander effects, also known as out-of-field or abscopal effects, presumably reflect the elicitation of tumor-specific adaptive immune responses. Ionizing rays have indeed been shown to promote the immunogenic demise of malignant cells, a process that relies on the spatiotemporally defined emanation of specific damage-associated molecular patterns (DAMPs). Thus, irradiation reportedly improves the clinical efficacy of other treatment modalities such as surgery (both in neo-adjuvant and adjuvant settings) or chemotherapy. Moreover, at least under some circumstances, radiotherapy may potentiate anticancer immune responses as elicited by various immunotherapeutic agents, including (but presumably not limited to) immunomodulatory monoclonal antibodies, cancer-specific vaccines, dendritic cell-based interventions and Toll-like receptor agonists. Here, we review the rationale of using radiotherapy, alone or combined with immunomodulatory agents, as a means to elicit or boost anticancer immune responses, and present recent clinical trials investigating the therapeutic potential of this approach in cancer patients.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, U848; Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
- National Institute of Health; Rome, Italy
| | - Eric Tartour
- INSERM, U970; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; Assistance Publique-Hôpitaux de Paris; Paris, France
| | | | - Catherine Sautès-Fridman
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; Assistance Publique-Hôpitaux de Paris; Paris, France
- Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | - Jérôme Galon
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Equipe 15, Centre de Recherche des Cordeliers; Paris, France
- INSERM, U872; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
| | - Laurence Zitvogel
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, U1015; Villejuif, France
| | - Guido Kroemer
- INSERM, U848; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; Assistance Publique-Hôpitaux de Paris; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
- Metabolomics and Cell Biology Platforms; Institut Gustave Roussy; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| |
Collapse
|
25
|
Qiu J, Yang G, Shen Z, Xie Y, Wang L. hPEBP4 as a predictive marker for the pathological response of rectal cancer to preoperative radiotherapy. Int J Colorectal Dis 2013; 28:241-6. [PMID: 22801881 DOI: 10.1007/s00384-012-1534-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/01/2012] [Indexed: 02/04/2023]
Abstract
PURPOSE The ability to predict tumor sensitivity toward radiotherapy is important in the personalized application of preoperative radiotherapy for patients with rectal cancer. The aim of the present study was to test the human phosphatidylethanolamine-binding protein 4 (hPEBP4) as a predictive marker of tumor response to preoperative radiotherapy in patients with rectal cancer. METHOD A retrospective analysis was conducted, consisting of 86 patients with locally advanced rectal cancer who underwent short-course preoperative radiotherapy (20 Gy in five fractions for 1 week) followed by a radical resection. Both pretreatment biopsy specimens and resected primary tumor tissue were collected. Immunohistochemistry and tumor regression grading system were used to evaluate the expression of hPEBP4 in the pretreatment biopsy specimens and the response of rectal cancer to radiotherapy, respectively. Expression of hPEBP4 was correlated with tumor regression in the resected specimen and the clinical outcome of the patients as well. RESULTS We found that high expression of hPEBP4 was associated with radioresistance in both univariate and multivariate analyses, and patients with a high hPEBP4 expression had a poorer progression-free survival than those with low hPEBP4 expression. CONCLUSION Our study revealed the independent predictive values of hPEBP4 in response of rectal cancer to preoperative radiotherapy, which suggests that upregulating hPEBP4 might be a potential mechanism by which rectal cancer cells avoid the destructive effects of radiotherapy.
Collapse
Affiliation(s)
- Jianming Qiu
- Department of Colorectal Surgery, The Third People's Hospital of Hangzhou, Hangzhou, China.
| | | | | | | | | |
Collapse
|
26
|
Abstract
There are several well-established mechanisms involved in radiation-induced cell death in mammalian cell systems. The p53-mediated apoptotic pathway is the most widely recognized mechanism (Lowe et al. Nature 362:847-849, 1993), although apoptosis has long been considered a less relevant mechanism of radiation-induced cell death (Steel, Acta Oncol 40:968-975, 2001; Brown and Wouters, Cancer Res 59:1391-1399, 1999; Olive and Durand, Int J Radiat Biol 71:695-707, 1997). We and others have recently focused instead on the emerging links between radiation, apoptosis, and ceramide and showed that ceramide is a sphingolipid-derived second messenger capable of initiating apoptotic cascades in response to various stress stimuli, including radiation.Ceramide, the backbone of all sphingolipids, is synthesized by a family of ceramide synthases (CerS), each using acyl-CoAs of defined chain length for N-acylation of the sphingoid long-chain base. Six mammalian CerS homologs have been cloned that demonstrated high selectivity towards acyl-CoAs (Lahiri et al. FEBS Lett 581:5289-5294, 2007), and more recently, it was shown that their activity can be modulated by dimer formation (Mesicek et al. Cell Signal 22:1300-1307, 2010; Laviad et al. J Biol Chem 283:5677-5684, 2008).This de novo ceramide synthesis has been observed in irradiated cells through a pathway normally suppressed by ataxia telangiectasia-mutated (ATM) protein, a key component of the cellular response to DNA double-strand breaks (Liao et al. J Biol Chem 274:17908-17917, 1999). ATM is not the sole factor known to affect apoptotic potential by modulating CerS activity. Recent work has also implicated protein kinase Cα (PKCα) as a potential CerS activator (Truman et al. Cancer Biol Ther 8:54-63, 2009).In this review, we summarize involvement of CerS in sphingolipid-mediated apoptosis in irradiated human prostate cancer cells and discuss future directions in this field.
Collapse
Affiliation(s)
- Carla Hajj
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | | |
Collapse
|
27
|
Liang B, Kong D, Liu Y, Liang N, He M, Ma S, Liu X. Autophagy inhibition plays the synergetic killing roles with radiation in the multi-drug resistant SKVCR ovarian cancer cells. Radiat Oncol 2012; 7:213. [PMID: 23244773 PMCID: PMC3552756 DOI: 10.1186/1748-717x-7-213] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 12/11/2012] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Autophagy has attracted attentions as a novel mechanism for tumor development. In this study Human ovarian carcinoma cell line SKOV3 and multidrug-resistant phenotype SKVCR cells were used and the roles of autophagy in radiation-induced cell death were analyzed. METHODS AND MATERIALS Cell viability was examined by colony formation and cell counting kit-8 (CCK-8) assay, 3MA and ZVAD were used to block autophagy and apoptosis, respectively. Quantitative real-time PCR was used to detect mRNA level and Western blot was used to detect protein expression, monodansylcadaverine (MDC) staining and flow cytometery were used for autophagy, apoptosis and cell cycle dynamics, respectively. RESULTS (1) The radiosensitivity exhibited differently in SKOV3 and SKVCR cells (SKOV3: D0=3.37, SKVCR: D0= 4.18); compared with SKOV3 the constitutive expression of MAPLC3 in SKVCR was higher, but no change of Caspase-3 and cleaved Caspase-3. (2) The ionizing radiation (IR)- induced apoptosis and autophagy were significant in both cells (P<0.05); inhibition of apoptosis with ZVAD showed no impact on survival of SKOV3 and SKVCR cells after radiation, while inhibition of autophagy significantly decreased viability in SKVCR cells, for SKVO3 cells only low level of radiation (2 Gy and 4 Gy) could decrease the viability(P<0.05). (3) ZVAD inhibited apoptosis and autophagy in both cells, 3MA inhibit apoptosis in SKOV3, and promote apoptosis in SKVCR, together with inhibition of autophagy. (4) G2/M arrest was induced by radiation in both cells; the accumulation of G2/M was more significant in SKOV3, 3MA attenuated the radiation-induced S phase delay in SKVCR. CONCLUSION IR-induced autophagy provides a self-protective mechanism against radiotherapy in SKVCR cells, the use of autophagy inhibitor, 3MA, increases the killing effects of radiation by inhibiting autophagy and radiation- induced S phase delay, also by the increase of apoptosis, which suggests a better therapeutic strategy in drug- resistant SKVCR ovarian cancer cells.
Collapse
Affiliation(s)
- Bing Liang
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin 130021, China
| | | | | | | | | | | | | |
Collapse
|
28
|
Bonnema SJ, Hegedüs L. Radioiodine therapy in benign thyroid diseases: effects, side effects, and factors affecting therapeutic outcome. Endocr Rev 2012; 33:920-80. [PMID: 22961916 DOI: 10.1210/er.2012-1030] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Radioiodine ((131)I) therapy of benign thyroid diseases was introduced 70 yr ago, and the patients treated since then are probably numbered in the millions. Fifty to 90% of hyperthyroid patients are cured within 1 yr after (131)I therapy. With longer follow-up, permanent hypothyroidism seems inevitable in Graves' disease, whereas this risk is much lower when treating toxic nodular goiter. The side effect causing most concern is the potential induction of ophthalmopathy in predisposed individuals. The response to (131)I therapy is to some extent related to the radiation dose. However, calculation of an exact thyroid dose is error-prone due to imprecise measurement of the (131)I biokinetics, and the importance of internal dosimetric factors, such as the thyroid follicle size, is probably underestimated. Besides these obstacles, several potential confounders interfere with the efficacy of (131)I therapy, and they may even interact mutually and counteract each other. Numerous studies have evaluated the effect of (131)I therapy, but results have been conflicting due to differences in design, sample size, patient selection, and dose calculation. It seems clear that no single factor reliably predicts the outcome from (131)I therapy. The individual radiosensitivity, still poorly defined and impossible to quantify, may be a major determinant of the outcome from (131)I therapy. Above all, the impact of (131)I therapy relies on the iodine-concentrating ability of the thyroid gland. The thyroid (131)I uptake (or retention) can be stimulated in several ways, including dietary iodine restriction and use of lithium. In particular, recombinant human thyrotropin has gained interest because this compound significantly amplifies the effect of (131)I therapy in patients with nontoxic nodular goiter.
Collapse
Affiliation(s)
- Steen Joop Bonnema
- Department of Endocrinology, Odense University Hospital, DK-5000 Odense C, Denmark.
| | | |
Collapse
|
29
|
Jia LT, Chen SY, Yang AG. Cancer gene therapy targeting cellular apoptosis machinery. Cancer Treat Rev 2012; 38:868-876. [PMID: 22800735 DOI: 10.1016/j.ctrv.2012.06.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 06/21/2012] [Accepted: 06/22/2012] [Indexed: 01/14/2023]
Abstract
The unraveling of cellular apoptosis machinery provides novel targets for cancer treatment, and gene therapy targeting this suicidal system has been corroborated to cause inflammation-free autonomous elimination of neoplastic cells. The apoptotic machinery can be targeted by introduction of a gene encoding an inducer, mediator or executioner of apoptotic cell death or by inhibition of anti-apoptotic gene expression. Strategies targeting cancer cells, which are achieved by selective gene delivery, specific gene expression or secretion of target proteins via genetic modification of autologous cells, dictate the outcome of apoptosis-based cancer gene therapy. Despite so far limited clinical success, gene therapy targeting the apoptotic machinery has great potential to benefit patients with threatening malignancies provided the availability of efficient and specific gene delivery and administration systems.
Collapse
Affiliation(s)
- Lin-Tao Jia
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China.
| | | | | |
Collapse
|
30
|
Raju U, Riesterer O, Wang ZQ, Molkentine DP, Molkentine JM, Johnson FM, Glisson B, Milas L, Ang KK. Dasatinib, a multi-kinase inhibitor increased radiation sensitivity by interfering with nuclear localization of epidermal growth factor receptor and by blocking DNA repair pathways. Radiother Oncol 2012; 105:241-9. [DOI: 10.1016/j.radonc.2012.08.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 07/24/2012] [Accepted: 08/14/2012] [Indexed: 11/28/2022]
|
31
|
Yu H, Li X, Sun S, Gao X, Zhou D. c-Met inhibitor SU11274 enhances the response of the prostate cancer cell line DU145 to ionizing radiation. Biochem Biophys Res Commun 2012; 427:659-65. [PMID: 23026049 DOI: 10.1016/j.bbrc.2012.09.117] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 09/21/2012] [Indexed: 02/08/2023]
Abstract
Hormone-refractory prostate cancer shows substantial resistance to most conventional therapies including radiotherapy, constitutes a key impediment to curing patients with the disease. c-Met overexpression plays a key role in prostate cancer tumorigenesis and disease progression. Here, we demonstrate that c-Met inhibition by SU11274 could significantly suppress cell survival and proliferation as well as enhance the radiosensitivity of DU145 cells. The underlying mechanisms of the effects of SU11274 on DU145 cells may include the inhibition of c-Met signaling, depolarization of the mitochondrial membrane potential, impairment of DNA repair function, abrogation of cell cycle arrest, and enhancement of cell death. Our study is the first to show the effectiveness of combining c-Met inhibition with ionizing radiation to cure hormone-refractory prostate cancer.
Collapse
Affiliation(s)
- Hongliang Yu
- Department of Radiation Oncology, Peking University First Hospital, Peking University, Beijing, China
| | | | | | | | | |
Collapse
|
32
|
Havelund BM, Sørensen FB, Pløen J, Lindebjerg J, Spindler KLG, Jakobsen A. Immunohistological expression of HIF-1α, GLUT-1, Bcl-2 and Ki-67 in consecutive biopsies during chemoradiotherapy in patients with rectal cancer. APMIS 2012; 121:127-38. [PMID: 23030583 DOI: 10.1111/j.1600-0463.2012.02949.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 06/22/2012] [Indexed: 12/16/2022]
Abstract
The aim of this study was to describe the dynamics of HIF-1α, GLUT-1, Bcl-2 and Ki-67 during chemoradiotherapy (CRT) of rectal cancer, and to investigate the fluctuation of these biomarkers in relation to pathological response to CRT. The study included 86 patients with rectal adenocarcinoma receiving preoperative CRT (>50.4 Gy and Uracil/Tegafur). Immunohistological expressions of HIF-1α, GLUT-1, Bcl-2 and Ki-67 were investigated in biopsies taken before treatment, after 2, 4 and 6 weeks of CRT and in specimens from the operation. Decreasing expressions of HIF-1α, Bcl-2 and Ki-67 were observed during CRT, whereas GLUT-1 overall was unchanged. No significant changes of the markers were observed in the interval between CRT and surgery. A significant association was observed between the presence of residual carcinoma after 6 weeks of treatment and pathological response to CRT, but no association was seen between the fluctuations of any of the markers and response to CRT. This unique material containing specimens before, after and during CRT for rectal cancer demonstrated biological dynamics in HIF-1α, Bcl-2 and Ki-67, but not GLUT-1, expression during CRT, and a significant association was seen between the presence of residual carcinoma after 6 weeks of treatment and pathological response to CRT.
Collapse
|
33
|
Allen AM, Ben-Ami M, Reshef A, Steinmetz A, Kundel Y, Inbar E, Djaldetti R, Davidson T, Fenig E, Ziv I. Assessment of response of brain metastases to radiotherapy by PET imaging of apoptosis with ¹⁸F-ML-10. Eur J Nucl Med Mol Imaging 2012; 39:1400-8. [PMID: 22699524 DOI: 10.1007/s00259-012-2150-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 04/30/2012] [Indexed: 01/01/2023]
Abstract
PURPOSE Early assessment of tumor response to therapy is vital for treatment optimization for the individual cancer patient. Induction of apoptosis is an early and nearly universal effect of anticancer therapies. The purpose of this study was to assess the performance of (18)F-ML-10, a novel PET radiotracer for apoptosis, as a tool for the early detection of response of brain metastases to whole-brain radiation therapy (WBRT). MATERIALS AND METHODS Ten patients with brain metastases treated with WBRT at 30 Gy in ten daily fractions were enrolled in this trial. Each patient underwent two (18)F-ML-10 PET scans, one prior to the radiation therapy (baseline scan), and the second after nine or ten fractions of radiotherapy (follow-up scan). MRI was performed at 6-8 weeks following completion of the radiation therapy. Early treatment-induced changes in tumor (18)F-ML-10 uptake on the PET scan were measured by voxel-based analysis, and were then evaluated by correlation analysis as predictors of the extent of later changes in tumor anatomical dimensions as seen on MRI scans 6-8 weeks after completion of therapy. RESULTS In all ten patients, all brain lesions were detected by both MRI and the (18)F-ML-10 PET scan. A highly significant correlation was found between early changes on the (18)F-ML-10 scan and later changes in tumor anatomical dimensions (r = 0.9). CONCLUSION These results support the potential of (18)F-ML-10 PET as a novel tool for the early detection of response of brain metastases to WBRT.
Collapse
Affiliation(s)
- Aaron M Allen
- Department of Radiation Oncology, Nuclear Medicine, Radiology and Neurology, Rabin Medical Center, Petach-Tikvah, Israel.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Haimovitz-Friedman A, Yang TIJ, Thin TH, Verheij M. Imaging Radiotherapy-Induced Apoptosis. Radiat Res 2012; 177:467-82. [DOI: 10.1667/rr2576.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
35
|
Suzuki M, Yamauchi M, Oka Y, Suzuki K, Yamashita S. Live-cell imaging visualizes frequent mitotic skipping during senescence-like growth arrest in mammary carcinoma cells exposed to ionizing radiation. Int J Radiat Oncol Biol Phys 2012; 83:e241-50. [PMID: 22365624 DOI: 10.1016/j.ijrobp.2011.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 11/24/2011] [Accepted: 11/30/2011] [Indexed: 12/22/2022]
Abstract
PURPOSE Senescence-like growth arrest in human solid carcinomas is now recognized as the major outcome of radiotherapy. This study was designed to analyze cell cycle during the process of senescence-like growth arrest in mammary carcinoma cells exposed to X-rays. METHODS AND MATERIALS Fluorescent ubiquitination-based cell cycle indicators were introduced into the human mammary carcinoma cell line MCF-7. Cell cycle was sequentially monitored by live-cell imaging for up to 5 days after exposure to 10 Gy of X-rays. RESULTS Live-cell imaging revealed that cell cycle transition from G2 to G1 phase without mitosis, so-called mitotic skipping, was observed in 17.1% and 69.8% of G1- and G2-irradiated cells, respectively. Entry to G1 phase was confirmed by the nuclear accumulation of mKO(2)-hCdt1 as well as cyclin E, which was inversely correlated to the accumulation of G2-specific markers such as mAG-hGeminin and CENP-F. More than 90% of cells skipping mitosis were persistently arrested in G1 phase and showed positive staining for the senescent biochemical marker, which is senescence-associated ß-galactosidase, indicating induction of senescence-like growth arrest accompanied by mitotic skipping. While G2 irradiation with higher doses of X-rays induced mitotic skipping in approximately 80% of cells, transduction of short hairpin RNA (shRNA) for p53 significantly suppressed mitotic skipping, suggesting that ionizing radiation-induced mitotic skipping is associated with p53 function. CONCLUSIONS The present study found the pathway of senescence-like growth arrest in G1 phase without mitotic entry following G2-irradiation.
Collapse
Affiliation(s)
- Masatoshi Suzuki
- Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | | | | | | | | |
Collapse
|
36
|
Stewart FA, Akleyev AV, Hauer-Jensen M, Hendry JH, Kleiman NJ, Macvittie TJ, Aleman BM, Edgar AB, Mabuchi K, Muirhead CR, Shore RE, Wallace WH. ICRP publication 118: ICRP statement on tissue reactions and early and late effects of radiation in normal tissues and organs--threshold doses for tissue reactions in a radiation protection context. Ann ICRP 2012; 41:1-322. [PMID: 22925378 DOI: 10.1016/j.icrp.2012.02.001] [Citation(s) in RCA: 846] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
This report provides a review of early and late effects of radiation in normal tissues and organs with respect to radiation protection. It was instigated following a recommendation in Publication 103 (ICRP, 2007), and it provides updated estimates of 'practical' threshold doses for tissue injury defined at the level of 1% incidence. Estimates are given for morbidity and mortality endpoints in all organ systems following acute, fractionated, or chronic exposure. The organ systems comprise the haematopoietic, immune, reproductive, circulatory, respiratory, musculoskeletal, endocrine, and nervous systems; the digestive and urinary tracts; the skin; and the eye. Particular attention is paid to circulatory disease and cataracts because of recent evidence of higher incidences of injury than expected after lower doses; hence, threshold doses appear to be lower than previously considered. This is largely because of the increasing incidences with increasing times after exposure. In the context of protection, it is the threshold doses for very long follow-up times that are the most relevant for workers and the public; for example, the atomic bomb survivors with 40-50years of follow-up. Radiotherapy data generally apply for shorter follow-up times because of competing causes of death in cancer patients, and hence the risks of radiation-induced circulatory disease at those earlier times are lower. A variety of biological response modifiers have been used to help reduce late reactions in many tissues. These include antioxidants, radical scavengers, inhibitors of apoptosis, anti-inflammatory drugs, angiotensin-converting enzyme inhibitors, growth factors, and cytokines. In many cases, these give dose modification factors of 1.1-1.2, and in a few cases 1.5-2, indicating the potential for increasing threshold doses in known exposure cases. In contrast, there are agents that enhance radiation responses, notably other cytotoxic agents such as antimetabolites, alkylating agents, anti-angiogenic drugs, and antibiotics, as well as genetic and comorbidity factors. Most tissues show a sparing effect of dose fractionation, so that total doses for a given endpoint are higher if the dose is fractionated rather than when given as a single dose. However, for reactions manifesting very late after low total doses, particularly for cataracts and circulatory disease, it appears that the rate of dose delivery does not modify the low incidence. This implies that the injury in these cases and at these low dose levels is caused by single-hit irreparable-type events. For these two tissues, a threshold dose of 0.5Gy is proposed herein for practical purposes, irrespective of the rate of dose delivery, and future studies may elucidate this judgement further.
Collapse
|
37
|
Exposure to hypoxia following irradiation increases radioresistance in prostate cancer cells. Urol Oncol 2011; 31:1106-16. [PMID: 22130126 DOI: 10.1016/j.urolonc.2011.10.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 10/14/2011] [Accepted: 10/19/2011] [Indexed: 01/26/2023]
Abstract
BACKGROUND Tumor hypoxia is a common feature of prostate tumors associated with the stabilization of hypoxia-inducible-factor 1alpha (HIF-1α) and poor prognosis following radiation therapy. Lack of oxygen at the time of irradiation is associated with radioresistance, but recent reports suggest radioresponse is also modulated by the dynamic nature of tumor hypoxia. OBJECTIVE We proposed to evaluate the effect of post-irradiation hypoxic exposure on the radioresponse of 2 prostate cancer (CaP) cell lines (22Rv1, DU145) and to examine whether it correlates with modified cellular responses induced by hypoxia. METHODS AND RESULTS Aerobic and hypoxic CaP cells exposed to hypoxia (24 h) after irradiation (4 Gy) gained a survival advantage compared with cells fully oxygenated after irradiation. This survival advantage was associated with induction of a G2/M cell cycle arrest, reduced induction of apoptosis and decreased amount of senescent cells. These modified cellular responses appeared mediated by HIF-1α. CONCLUSION Our data suggest that targeting hypoxia after irradiation may benefit patients with aggressive hypoxic prostate tumors.
Collapse
|
38
|
Tsang TY, Tang WY, Chan JYW, Co NN, Au Yeung CL, Yau PL, Kong SK, Fung KP, Kwok TT. P-glycoprotein enhances radiation-induced apoptotic cell death through the regulation of miR-16 and Bcl-2 expressions in hepatocellular carcinoma cells. Apoptosis 2011; 16:524-35. [PMID: 21336967 DOI: 10.1007/s10495-011-0581-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
P-glycoprotein (Pgp), an efflux pump, was confirmed the first time to regulate the expressions of miR/gene in cells. Pgp is known to be associated with multidrug resistance. RHepG2 cells, the multidrug resistant subline of human hepatocellular carcinoma HepG2 cells, expressed higher levels of Pgp as well as miR-16, and lower level of Bcl-2 than the parental cells. In addition, RHepG2 cells were more radiation sensitive and showed more pronounced radiation-induced apoptotic cell death than the parental cells. Mechanistic analysis revealed that transfection with mdr1 specific antisense oligos suppressed radiation-induced apoptosis in HepG2 cells. On the other hand, ectopic mdr1 expression enhanced radiation-induced apoptosis in HepG2 cells, SK-HEP-1 cells, MiHa cells, and furthermore, induced miR-16 and suppressed its target gene Bcl-2 in HepG2 cells. Moreover, the enhancement effects of Pgp and miR-16 on radiation-induced apoptosis were counteracted by overexpression of Bcl-2. The Pgp effect on miR-16/Bcl-2 was suppressed by Pgp blocker verapamil indicating the importance of the efflux of Pgp substrates. The present study is the first to reveal the role of Pgp in regulation of miRNA/gene expressions. The findings may provide new perspective in understanding the biological function of Pgp.
Collapse
Affiliation(s)
- Tsun Yee Tsang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chaudhary P, Shukla SK, Sharma RK. REC-2006-A Fractionated Extract of Podophyllum hexandrum Protects Cellular DNA from Radiation-Induced Damage by Reducing the Initial Damage and Enhancing Its Repair In Vivo. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2011; 2011:473953. [PMID: 20008078 PMCID: PMC3136531 DOI: 10.1093/ecam/nep212] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 11/09/2009] [Indexed: 11/13/2022]
Abstract
Podophyllum hexandrum, a perennial herb commonly known as the Himalayan May Apple, is well known in Indian and Chinese traditional systems of medicine. P. hexandrum has been widely used for the treatment of venereal warts, skin infections, bacterial and viral infections, and different cancers of the brain, lung and bladder. This study aimed at elucidating the effect of REC-2006, a bioactive fractionated extract from the rhizome of P. hexandrum, on the kinetics of induction and repair of radiation-induced DNA damage in murine thymocytes in vivo. We evaluated its effect on non-specific radiation-induced DNA damage by the alkaline halo assay in terms of relative nuclear spreading factor (RNSF) and gene-specific radiation-induced DNA damage via semi-quantitative polymerase chain reaction. Whole body exposure of animals with gamma rays (10 Gy) caused a significant amount of DNA damage in thymocytes (RNSF values 17.7 ± 0.47, 12.96 ± 1.64 and 3.3 ± 0.014) and a reduction in the amplification of β-globin gene to 0, 28 and 43% at 0, 15 and 60 min, respectively. Administrating REC-2006 at a radioprotective concentration (15 mg kg−1 body weight) 1 h before irradiation resulted in time-dependent reduction of DNA damage evident as a decrease in RNSF values 6.156 ± 0.576, 1.647 ± 0.534 and 0.496 ± 0.012, and an increase in β-globin gene amplification 36, 95 and 99%, at 0, 15 and 60 min, respectively. REC-2006 scavenged radiation-induced hydroxyl radicals in a dose-dependent manner stabilized DPPH free radicals and also inhibited superoxide anions. Various polyphenols and flavonoides present in REC-2006 might contribute to scavenging of radiation-induced free radicals, thereby preventing DNA damage and stimulating its repair.
Collapse
Affiliation(s)
- Pankaj Chaudhary
- Institute of Nuclear Medicine and Allied Sciences (INMAS), Delhi, India
| | | | | |
Collapse
|
40
|
Kapty J, Murray D, Mercer J. Radiotracers for noninvasive molecular imaging of tumor cell death. Cancer Biother Radiopharm 2011; 25:615-28. [PMID: 21204755 DOI: 10.1089/cbr.2010.0793] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The need to monitor cancer therapy-induced cellular and tissue changes using noninvasive imaging techniques continues to stimulate both basic and clinical research. Monitoring changes in cellular proliferative capacity that occur after treatment with radiation and/or chemotherapy has the potential to provide longitudinal information on the cellular dynamics of tumors before, during, and after therapeutic intervention. Cells can lose their reproductive potential through one of several mechanisms, including apoptosis and autophagy (which are forms of programmed cell death), premature senescence, or necrosis. When a tumor responds to therapy, current imaging methods do not provide information about the exact mechanism of cell death executed. We are now beginning to develop the molecular imaging tools that will enable us to noninvasively image cell death mechanisms both in experimental models and in the clinical cancer environment. Studies with these imaging tools will contribute to a better understanding of therapeutic responses and assist in the design and evaluation of more effective treatments. This review examines the state-of-the-art in the use of (radio)tracers for the purpose of imaging mechanisms of tumor cell inactivation (cell death) in animal models and in clinical trials.
Collapse
Affiliation(s)
- Janice Kapty
- Department of Oncology, University of Alberta, Edmonton, Canada
| | | | | |
Collapse
|
41
|
Park SR, Lee KD, Kim UK, Gil YG, Oh KS, Park BS, Kim GC. Pseudomonas aeruginosa exotoxin A reduces chemoresistance of oral squamous carcinoma cell via inhibition of heat shock proteins 70 (HSP70). Yonsei Med J 2010; 51:708-16. [PMID: 20635445 PMCID: PMC2908850 DOI: 10.3349/ymj.2010.51.5.708] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 12/24/2009] [Accepted: 12/24/2009] [Indexed: 12/04/2022] Open
Abstract
PURPOSE Oral squamous carcinoma (OSCC) cells exhibit resistance to chemotherapeutic agent-mediated apoptosis in the late stage of malignancy. Increased levels of heat shock proteins 70 (HSP70) in cancer cells are known to confer resistance to apoptosis. Since recent advances in the understanding of bacterial toxins have produced new strategies for the treatment of cancers, we investigated the effect of Pseudomonas aeruginosa exotoxin A (PEA) on HSP70 expression and induction of apoptosis in chemoresistant OSCC cell line (YD-9). MATERIALS AND METHODS The apoptotic effect of PEA on chemoresistant YD-9 cells was confirmed by MTT, Hoechst and TUNEL stains, DNA electrophoresis, and Western blot analysis. RESULTS While YD-9 cells showed high resistance to chemotherapeutic agents such as etoposide and 5-fluorouraci (5-FU), HSP70 antisense oligonucelotides sensitized chemoresistant YD-9 cells to etoposide and 5-FU. On the other hand, PEA significantly decreased the viability of YD-9 cells by deteriorating the HSP70-relating protecting system through inhibition of HSP70 expression and inducing apoptosis in YD-9 cells. Apoptotic manifestations were evidenced by changes in nuclear morphology, generation of DNA fragmentation, and activation of caspases. While p53, p21, and E2F-1 were upregulated, cdk2 and cyclin B were downregulated by PEA treatment, suggesting that PEA caused cell cycle arrest at the G2/M checkpoint. CONCLUSION Therefore, these results indicate that PEA reduced the chemoresistance through inhibition of HSP70 expression and also induced apoptosis in chemoresistant YD-9 cells.
Collapse
Affiliation(s)
- Sang Rye Park
- Department of Oral Anatomy, School of Dentistry, Research Institute for Oral Biotechnology, Pusan National University, Yangsan, Korea
| | - Kyoung Duk Lee
- Department of Oral Anatomy, School of Dentistry, Research Institute for Oral Biotechnology, Pusan National University, Yangsan, Korea
| | - Uk Kyu Kim
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Research Institute for Oral Biotechnology, Pusan National University, Yangsan, Korea
| | - Young Gi Gil
- Department of Anatomy, College of Medicine, Kosin University, Busan, Korea
| | - Kyu Seon Oh
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Bong Soo Park
- Department of Oral Anatomy, School of Dentistry, Research Institute for Oral Biotechnology, Pusan National University, Yangsan, Korea
| | - Gyoo Cheon Kim
- Department of Oral Anatomy, School of Dentistry, Research Institute for Oral Biotechnology, Pusan National University, Yangsan, Korea
| |
Collapse
|
42
|
Firat E, Heinemann F, Grosu AL, Hermann F, Niedermann G. Molecular radiobiology meets clinical radiation oncology. Int J Radiat Biol 2010; 86:252-9. [PMID: 20201653 DOI: 10.3109/09553000903419320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND The 2nd Langendorff Congress in Freiburg in Breisgau (Germany) gathered basic and translational scientists as well as clinicians interested in recent developments in molecular and clinical radiobiology. The topics ranged from the most recent insight into the organisation of the DNA damage response and radiotherapeutically relevant cell death mechanisms to biological imaging for treatment planning and advances in the understanding of the molecular biological effects of particle beams. Clinical aspects of stem cell and tumour stem cell biology as well as of angiogenesis and hypoxia, the search for novel molecular radiosensitisers and potential strategies for exploitation of the immune system to further improve tumour radiotherapy were also discussed. RESULTS AND CONCLUSION This report surveys the presentations at the meeting, considering their significance in light of the literature, and documents the increasing importance of molecular radiobiology for clinical radiooncology.
Collapse
Affiliation(s)
- Elke Firat
- Department of Radiation Oncology, University of Freiburg, Freiburg, Germany
| | | | | | | | | |
Collapse
|
43
|
Zhao H, Xu S, Wang Z, Li Y, Guo W, Lin C, Gong S, Li C, Wang G, Cai L. Repetitive exposures to low-dose X-rays attenuate testicular apoptotic cell death in streptozotocin-induced diabetes rats. Toxicol Lett 2010; 192:356-364. [PMID: 19931367 DOI: 10.1016/j.toxlet.2009.11.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Revised: 11/06/2009] [Accepted: 11/09/2009] [Indexed: 11/17/2022]
Abstract
To define whether repetitive exposures to low-dose radiation (LDR) can attenuate diabetes-induced testicular cell death, Type 1 diabetic rats were produced by single injection of streptozotocin (STZ). Once hyperglycemia was diagnosed, diabetic rats were treated with and without LDR (25 and 50 mGy X-rays) daily for 4 weeks. Eight and 12 weeks after diabetes onset, testicular apoptotic cell death was examined by flow cytometry with Annexin V/PI staining, Western blotting assay for caspase-3 cleavage, and TUNEL staining for localization of apoptotic cells. Diabetes induced a significant increase in testicular apoptotic cell death, which was able to be attenuated by repetitive exposures to LDR. Diabetes-induced testicular cell death was associated with increased mitochondrial dysfunction, shown by the decreased mitochondrial potential and increased expressions of Bax mRNA and protein. All these changes were significantly attenuated in certain extends by repetitive exposures to LDR. To investigate the mechanisms by which LDR attenuates diabetes-induced testicular apoptotic cell death, serum sex hormone (testosterone, luteinizing hormone and follicle stimulating hormone) levels, and both serum and testicular oxidative damage (lipid peroxides) and antioxidant contents (superoxide dismutase, catalase and glutathione) were measured. Serum sex hormones were significantly decreased in diabetic rats, but not significantly in diabetic rats with multiple exposures to LDR; serum and testicular oxidative damage was significantly increased along with significant decreases in serum and testicular antioxidants in diabetic rats; however, these changes were significantly prevented by repetitive exposures to LDR. Furthermore, diabetic effects on the testicular oxidative damage and cell death were all attenuated by antioxidant N-acetylcysteine. These results suggest that diabetes-induced testicular cell death is probably mediated by increased oxidative stress. LDR protection from diabetes-induced testicular cell death is most likely mediated by its preserving antioxidants.
Collapse
Affiliation(s)
- Hongguang Zhao
- Nuclear Medicine, The First Hospital of Jilin University, Changchun, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|