1
|
Jin M, Liu H, Zheng Z, Fang S, Xi Y, Liu K. CHI3L1 mediates radiation resistance in colorectal cancer by inhibiting ferroptosis via the p53/SLC7A11 pathway. J Transl Med 2025; 23:357. [PMID: 40119400 PMCID: PMC11929242 DOI: 10.1186/s12967-025-06378-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/12/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Radiotherapy is a key treatment for colorectal cancer (CRC), particularly rectal cancer; however, many patients are resistant to radiation. While it has been shown that CHI3L1 is associated with CRC progression, its specific function and regulatory mechanisms in radiation resistance remain unclear. METHODS The levels of CHI3L1 in CRC and normal tissue samples were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. To assess the effects of CHI3L1 on CRC cell proliferative, migratory, and invasive capacities, Cell Counting Kit-8 (CCK-8) and Transwell assays were performed. Radiation resistance in CRC cells with varying CHI3L1 expression levels was evaluated through colony formation assay. Western blot and immunofluorescence analyses were conducted to explore the correlation between CHI3L1 and p53 expression levels. Ferroptosis was assessed by determining reactive oxygen species (ROS), malondialdehyde (MDA), and glutathione (GSH) concentrations in cells with different CHI3L1 expression levels, and a xenograft mouse model was used to identify the molecular mechanisms of ferroptosis in vivo. RESULTS Significant CHI3L1 upregulated was observed in CRC tissues and was associated with promotion of malignant cell behaviors. The number of colonies in CHI3L1-overexpressing groups was significantly greater than that in the control groups following radiation, indicating increased radiation resistance in the former group. Furthermore, CHI3L1 overexpression was associated with p53 downregulation and elevated p53 ubiquitination. Notably, CHI3L1 inhibited the ferroptosis of CRC cells by suppressing p53 expression through the p53/SLC7A11 signaling pathway. CONCLUSIONS CHI3L1 overexpression promotes the proliferation, migration, invasion, and radiation resistance of CRC cells. Elevated CHI3L1 expression is associated with increased p53 ubiquitination and SLC7A11 upregulation. CHI3L1 promotes radiation resistance by suppressing ferroptosis in CRC cells through the p53/SLC7A11 axis.
Collapse
Affiliation(s)
- Ming Jin
- Department of Radiation Oncology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Hui Liu
- Department of Radiation Oncology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Zhen Zheng
- Department of Radiation Oncology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Shuai Fang
- The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Yang Xi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Kaitai Liu
- Department of Radiation Oncology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
2
|
Michaeli O, Luz I, Vatarescu M, Manko T, Weizman N, Korotinsky Y, Tsitrina A, Braiman A, Arazi L, Cooks T. APR-246 as a radiosensitization strategy for mutant p53 cancers treated with alpha-particles-based radiotherapy. Cell Death Dis 2024; 15:426. [PMID: 38890278 PMCID: PMC11189442 DOI: 10.1038/s41419-024-06830-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
Radiation therapy (RT) remains a common treatment for cancer patients worldwide, despite the development of targeted biological compounds and immunotherapeutic drugs. The challenge in RT lies in delivering a lethal dose to the cancerous site while sparing the surrounding healthy tissues. Low linear energy transfer (low-LET) and high linear energy transfer (high-LET) radiations have distinct effects on cells. High-LET radiation, such as alpha particles, induces clustered DNA double-strand breaks (DSBs), potentially inducing cell death more effectively. However, due to limited range, alpha-particle therapies have been restricted. In human cancer, mutations in TP53 (encoding for the p53 tumor suppressor) are the most common genetic alteration. It was previously reported that cells carrying wild-type (WT) p53 exhibit accelerated senescence and significant rates of apoptosis in response to RT, whereas cells harboring mutant p53 (mutp53) do not. This study investigated the combination of the alpha-emitting atoms RT based on internal Radium-224 (224Ra) sources and systemic APR-246 (a p53 reactivating compound) to treat tumors with mutant p53. Cellular models of colorectal cancer (CRC) or pancreatic ductal adenocarcinoma (PDAC) harboring mutant p53, were exposed to alpha particles, and tumor xenografts with mutant p53 were treated using 224Ra source and APR-246. Effects on cell survival and tumor growth, were assessed. The spread of alpha emitters in tumors was also evaluated as well as the spatial distribution of apoptosis within the treated tumors. We show that mutant p53 cancer cells exhibit radio-sensitivity to alpha particles in vitro and to alpha-particles-based RT in vivo. APR-246 treatment enhanced sensitivity to alpha radiation, leading to reduced tumor growth and increased rates of tumor eradication. Combining alpha-particles-based RT with p53 restoration via APR-246 triggered cell death, resulting in improved therapeutic outcomes. Further preclinical and clinical studies are needed to provide a promising approach for improving treatment outcomes in patients with mutant p53 tumors.
Collapse
Affiliation(s)
- Or Michaeli
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Ishai Luz
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Maayan Vatarescu
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
- Translational Research Laboratory, Alpha Tau Medical, Jerusalem, Israel
| | - Tal Manko
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Noam Weizman
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yevgeniya Korotinsky
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alexandra Tsitrina
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | - Lior Arazi
- Unit of Nuclear Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tomer Cooks
- The Shraga Segal Department of Microbiology, Immunology & Genetics, Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel.
| |
Collapse
|
3
|
Lieu DJ, Crowder MK, Kryza JR, Tamilselvam B, Kaminski PJ, Kim IJ, Li Y, Jeong E, Enkhbaatar M, Chen H, Son SB, Mok H, Bradley KA, Phillips H, Blanke SR. Autophagy suppression in DNA damaged cells occurs through a newly identified p53-proteasome-LC3 axis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595139. [PMID: 38826216 PMCID: PMC11142043 DOI: 10.1101/2024.05.21.595139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Macroautophagy is thought to have a critical role in shaping and refining cellular proteostasis in eukaryotic cells recovering from DNA damage. Here, we report a mechanism by which autophagy is suppressed in cells exposed to bacterial toxin-, chemical-, or radiation-mediated sources of genotoxicity. Autophagy suppression is directly linked to cellular responses to DNA damage, and specifically the stabilization of the tumor suppressor p53, which is both required and sufficient for regulating the ubiquitination and proteasome-dependent reduction in cellular pools of microtubule-associated protein 1 light chain 3 (LC3A/B), a key precursor of autophagosome biogenesis and maturation, in both epithelial cells and an ex vivo organoid model. Our data indicate that suppression of autophagy, through a newly identified p53-proteasome-LC3 axis, is a conserved cellular response to multiple sources of genotoxicity. Such a mechanism could potentially be important for realigning proteostasis in cells undergoing DNA damage repair.
Collapse
|
4
|
Leng JX, Su C, Carpenter DJ, Floyd W, Vaios E, Shenker R, Hendrickson PG, Giles W, Mullikin T, Floyd SR, Kirkpatrick JP, Green M, Reitman ZJ. Impact of TP53 mutations on brain metastasis control in non-small cell lung cancer patients undergoing stereotactic radiosurgery. JOURNAL OF RADIOSURGERY AND SBRT 2024; 9:91-99. [PMID: 39087065 PMCID: PMC11288656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/18/2023] [Indexed: 08/02/2024]
Abstract
Purpose To investigate whether TP53 variants may be correlated with overall survival and local control following stereotactic radiosurgery (SRS) for brain metastases (BMs) from non-small cell lung cancer (NSCLC). Methods Patients undergoing an initial course of SRS for NSCLC brain metastases between 1/2015 and 12/2020 were retrospectively identified. Overall survival and freedom from local intracranial progression (FFLIP) were estimated via Kaplan-Meier method. Cox models assessed TP53 variant status (pathogenic variant, PV; variant not detected, ND). Results 255 patients underwent molecular profiling for TP53, among whom 144 (56%) had a TP53 PV. Median follow-up was 11.6 months. OS was not significantly different across TP53 status. A trend toward superior FFLIP was observed for PV (95% CI 62.9 months-NR) versus ND patients (95% CI 29.4 months-NR; p=0.06). Superior FFLIP was observed for patients with one TP53 variant versus those with TP53 ND. Conclusion Among NSCLC patients with BMs, the potential association between TP53 status and post-SRS FFLIP warrants further investigation in a larger prospective cohort.
Collapse
Affiliation(s)
- Jim X Leng
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Chang Su
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - David J Carpenter
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Warren Floyd
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene Vaios
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Rachel Shenker
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Peter G Hendrickson
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Will Giles
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Trey Mullikin
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Scott R Floyd
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - John P Kirkpatrick
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Michelle Green
- Department of Pathology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Zachary J Reitman
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
- Department of Pathology, Duke University Medical Center, Durham, NC, 27710, USA
| |
Collapse
|
5
|
Fu Z, Zhao PY, Yang XP, Li H, Hu SD, Xu YX, Du XH. Cannabidiol regulates apoptosis and autophagy in inflammation and cancer: A review. Front Pharmacol 2023; 14:1094020. [PMID: 36755953 PMCID: PMC9899821 DOI: 10.3389/fphar.2023.1094020] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023] Open
Abstract
Cannabidiol (CBD) is a terpenoid naturally found in plants. The purified compound is used in the treatment of mental disorders because of its antidepressive, anxiolytic, and antiepileptic effects. CBD can affect the regulation of several pathophysiologic processes, including autophagy, cytokine secretion, apoptosis, and innate and adaptive immune responses. However, several authors have reported contradictory findings concerning the magnitude and direction of CBD-mediated effects. For example, CBD treatment can increase, decrease, or have no significant effect on autophagy and apoptosis. These variable results can be attributed to the differences in the biological models, cell types, and CBD concentration used in these studies. This review focuses on the mechanism of regulation of autophagy and apoptosis in inflammatory response and cancer by CBD. Further, we broadly elaborated on the prospects of using CBD as an anti-inflammatory agent and in cancer therapy in the future.
Collapse
Affiliation(s)
- Ze Fu
- Medical School of Chinese PLA, Beijing, China
| | | | | | - Hao Li
- Medical School of Chinese PLA, Beijing, China
| | - Shi-Dong Hu
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ying-Xin Xu
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao-Hui Du
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing, China,*Correspondence: Xiao-Hui Du,
| |
Collapse
|
6
|
Mireștean CC, Iancu RI, Iancu DPT. p53 Modulates Radiosensitivity in Head and Neck Cancers-From Classic to Future Horizons. Diagnostics (Basel) 2022; 12:3052. [PMID: 36553058 PMCID: PMC9777383 DOI: 10.3390/diagnostics12123052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/08/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
p53, initially considered a tumor suppressor, has been the subject of research related to cancer treatment resistance in the last 30 years. The unfavorable response to multimodal therapy and the higher recurrence rate, despite an aggressive approach, make HNSCC a research topic of interest for improving therapeutic outcomes, even if it is only the sixth most common malignancy worldwide. New advances in molecular biology and genetics include the involvement of miRNA in the control of the p53 pathway, the understanding of mechanisms such as gain/loss of function, and the development of different methods to restore p53 function, especially for HPV-negative cases. The different ratio between mutant p53 status in the primary tumor and distant metastasis originating HNSCC may serve to select the best therapeutic target for activating an abscopal effect by radiotherapy as a "booster" of the immune system. P53 may also be a key player in choosing radiotherapy fractionation regimens. Targeting any pathway involving p53, including tumor metabolism, in particular the Warburg effect, could modulate the radiosensitivity and chemo-sensitivity of head and neck cancers.
Collapse
Affiliation(s)
- Camil Ciprian Mireștean
- Department of Oncology and Radiotherapy, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
- Department of Surgery, Railways Clinical Hospital Iasi, 700506 Iași, Romania
| | - Roxana Irina Iancu
- Oral Pathology Department, Faculty of Dental Medicine, “Gr. T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Department of Clinical Laboratory, “St. Spiridon” Emergency Universitary Hospital, 700111 Iași, Romania
| | - Dragoș Petru Teodor Iancu
- Oncology and Radiotherapy Department, Faculty of Medicine, “Gr. T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Department of Radiation Oncology, Regional Institute of Oncology, 700483 Iași, Romania
| |
Collapse
|
7
|
BPR0C261, An Analogous of Microtubule Disrupting Agent D-24851 Enhances the Radiosensitivity of Human Non-Small Cell Lung Cancer Cells via p53-Dependent and p53-Independent Pathways. Int J Mol Sci 2022; 23:ijms232214083. [PMID: 36430560 PMCID: PMC9692308 DOI: 10.3390/ijms232214083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/23/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
(1) Destabilization of microtubule dynamics is a primary strategy to inhibit fast growing tumor cells. The low cytotoxic derivative of microtubule inhibitor D-24851, named BPR0C261 exhibits antitumor activity via oral administration. In this study, we investigated if BPR0C261 could modulate the radiation response of human non-small cell lung cancer (NSCLC) cells with or without p53 expression. (2) Different doses of BPR0C261 was used to treat human NSCLC A549 (p53+/+) cells and H1299 (p53-/-) cells. The cytotoxicity, radiosensitivity, cell cycle distribution, DNA damage, and protein expression were evaluated using an MTT assay, a colony formation assay, flow cytometry, a comet assay, and an immunoblotting analysis, respectively. (3) BPR0C261 showed a dose-dependent cytotoxicity on A549 cells and H1299 cells with IC50 at 0.38 μM and 0.86 μM, respectively. BPR0C261 also induced maximum G2/M phase arrest and apoptosis in both cell lines after 24 h of treatment with a dose-dependent manner. The colony formation analysis demonstrated that a combination of low concentration of BPR0C261 and X-rays caused a synergistic radiosensitizing effect on NSCLC cells. Additionally, we found that a low concentration of BPR0C261 was sufficient to induce DNA damage in these cells, and it increased the level of DNA damage induced by a fractionation radiation dose (2 Gy) of conventional radiotherapy. Furthermore, the p53 protein level of A549 cell line was upregulated by BPR0C261. On the other hand, the expression of PTEN tumor suppressor was found to be upregulated in H1299 cells but not in A549 cells under the same treatment. Although radiation could not induce PTEN in H1299 cells, a combination of low concentration of BPR0C261 and radiation could reverse this situation. (4) BPR0C261 exhibits specific anticancer effects on NSCLC cells by the enhancement of DNA damage and radiosensitivity with p53-dependent and p53-independent/PTEN-dependent manners. The combination of radiation and BPR0C261 may provide an important strategy for the improvement of radiotherapeutic treatment.
Collapse
|
8
|
Niu H, Zhu Y, Wang J, Wang T, Wang X, Yan L. Effects of USP7 on radiation sensitivity through p53 pathway in laryngeal squamous cell carcinoma. Transl Oncol 2022; 22:101466. [PMID: 35696794 PMCID: PMC9194850 DOI: 10.1016/j.tranon.2022.101466] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/29/2022] [Accepted: 06/05/2022] [Indexed: 11/29/2022] Open
Abstract
The relationship between ubiquitin specific protease 7 (USP7) and radio-sensitivity in laryngeal squamous cell carcinoma (LSCC) has not been reported yet. Using gene chip and Label-Free mass spectrometry, we found that USP7 was significantly increased both in radioresistant LSCC patients and LSCC cells receiving irradiation. Since p53 is the most important downstream gene of USP7 and is frequently mutated in LSCC, we investigated the effects of USP7 on radioresistance of LSCC cells with or without p53 mutation. We found that knockdown of USP7 increased the radio-sensitivity in p53-mutated LSCC cells, while inhibiting the radio-sensitivity in p53-wild type cells. Knockdown of USP7 significantly inhibited the expression of the p53 and p53 pathway. Overexpressing endogenous p53 by CRISPR/dCas9 could reverse the effects of USP7 on radiosensitivity both in vitro and in vivo. Our results demonstrated the irradiation-induced USP7 led to radioresistance in p53-mutated LSCC cells but radio-sensitivity in p53-wild type cells. Therefore, the clinical application of USP7 inhibitors may improve the effects of radiotherapy in LSCC with p53 mutations and reduce the side effects on surrounding normal tissues without p53 mutation.
Collapse
Affiliation(s)
- Hao Niu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China; Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, China
| | - Yi Zhu
- Department of Radiation Oncology, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Jie Wang
- Department of Radiation Oncology, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Tian Wang
- Department of Radiation Oncology, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Xiaosheng Wang
- Department of Radiation Oncology, Eye and ENT Hospital, Fudan University, Shanghai, China.
| | - Li Yan
- Department of Radiation Oncology, Eye and ENT Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Carlos-Reyes A, Muñiz-Lino MA, Romero-Garcia S, López-Camarillo C, Hernández-de la Cruz ON. Biological Adaptations of Tumor Cells to Radiation Therapy. Front Oncol 2021; 11:718636. [PMID: 34900673 PMCID: PMC8652287 DOI: 10.3389/fonc.2021.718636] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
Radiation therapy has been used worldwide for many decades as a therapeutic regimen for the treatment of different types of cancer. Just over 50% of cancer patients are treated with radiotherapy alone or with other types of antitumor therapy. Radiation can induce different types of cell damage: directly, it can induce DNA single- and double-strand breaks; indirectly, it can induce the formation of free radicals, which can interact with different components of cells, including the genome, promoting structural alterations. During treatment, radiosensitive tumor cells decrease their rate of cell proliferation through cell cycle arrest stimulated by DNA damage. Then, DNA repair mechanisms are turned on to alleviate the damage, but cell death mechanisms are activated if damage persists and cannot be repaired. Interestingly, some cells can evade apoptosis because genome damage triggers the cellular overactivation of some DNA repair pathways. Additionally, some surviving cells exposed to radiation may have alterations in the expression of tumor suppressor genes and oncogenes, enhancing different hallmarks of cancer, such as migration, invasion, and metastasis. The activation of these genetic pathways and other epigenetic and structural cellular changes in the irradiated cells and extracellular factors, such as the tumor microenvironment, is crucial in developing tumor radioresistance. The tumor microenvironment is largely responsible for the poor efficacy of antitumor therapy, tumor relapse, and poor prognosis observed in some patients. In this review, we describe strategies that tumor cells use to respond to radiation stress, adapt, and proliferate after radiotherapy, promoting the appearance of tumor radioresistance. Also, we discuss the clinical impact of radioresistance in patient outcomes. Knowledge of such cellular strategies could help the development of new clinical interventions, increasing the radiosensitization of tumor cells, improving the effectiveness of these therapies, and increasing the survival of patients.
Collapse
Affiliation(s)
- Angeles Carlos-Reyes
- Department of Chronic-Degenerative Diseases, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Marcos A. Muñiz-Lino
- Laboratorio de Patología y Medicina Bucal, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City, Mexico
| | - Susana Romero-Garcia
- Department of Chronic-Degenerative Diseases, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico, Mexico City
| | | |
Collapse
|
10
|
Kong X, Yu D, Wang Z, Li S. Relationship between p53 status and the bioeffect of ionizing radiation. Oncol Lett 2021; 22:661. [PMID: 34386083 PMCID: PMC8299044 DOI: 10.3892/ol.2021.12922] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
Radiotherapy is widely used in the clinical treatment of cancer patients and it may be used alone or in combination with surgery or chemotherapy to inhibit tumor development. However, radiotherapy may at times not kill all cancer cells completely, as certain cells may develop radioresistance that counteracts the effects of radiation. The emergence of radioresistance is associated with the genetic background and epigenetic regulation of cells. p53 is an important tumor suppressor gene that is expressed at low levels in cells. However, when cells are subjected to stress-induced stimulation, the expression level of p53 increases, thereby preventing genomic disruption. This mechanism has important roles in maintaining cell stability and inhibiting carcinogenesis. However, mutation and deletion destroy the anticancer function of p53 and may induce carcinogenesis. In tumor radiotherapy, the status of p53 expression in cancer cells has a close relationship with radiotherapeutic efficacy. Therefore, understanding how p53 expression affects the cellular response to radiation is of great significance for solving the problem of radioresistance and improving radiotherapeutic outcomes. For the present review, the literature was searched for studies published between 1979 and 2021 using the PubMed database (https://pubmed.ncbi.nlm.nih.gov/) with the following key words: Wild-type p53, mutant-type p53, long non-coding RNA, microRNA, gene mutation, radioresistance and radiosensitivity. From the relevant studies retrieved, the association between different p53 mutants and cellular radiosensitivity, as well as the molecular mechanisms of p53 affecting the radiosensitivity of cells, were summarized. The aim of the present study was to provide useful information for understanding and resolving radioresistance, to help clinical researchers develop more accurate treatment strategies and to improve radiotherapeutic outcomes for cancer patients with p53 mutations.
Collapse
Affiliation(s)
- Xiaohan Kong
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dehai Yu
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, Jilin 130061, P.R. China
| | - Zhaoyi Wang
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Sijie Li
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
11
|
Li L, Liu WL, Su L, Lu ZC, He XS. The Role of Autophagy in Cancer Radiotherapy. Curr Mol Pharmacol 2021; 13:31-40. [PMID: 31400274 DOI: 10.2174/1874467212666190809154518] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Autophagy, a pathway for lysosomal-mediated cellular degradation, is a catabolic process that recycles intracellular components to maintain metabolism and survival. It is classified into three major types: macroautophagy, microautophagy, and the chaperone-mediated autophagy (CMA). Autophagy is a dynamic and multistep process that includes four stages: nucleation, elongation, autophagosome formation, and fusion. Interestingly, the influence of autophagy in cancer development is complex and paradoxical, suppressive, or promotive in different contexts. Autophagy in cancer has been demonstrated to serve as both a tumour suppressor and promoter. Radiotherapy is a powerful and common strategy for many different types of cancer and can induce autophagy, which has been shown to modulate sensitivity of cancer to radiotherapy. However, the role of autophagy in radiation treatment is controversial. Some reports showed that the upregulation of autophagy was cytoprotective for cancer cells. Others, in contrast, showed that the induction of autophagy was advantageous. Here, we reviewed recent studies and attempted to discuss the various aspects of autophagy in response to radiotherapy of cancer. Thus, we could decrease the viability of cancer cell and increase the sensibility of cancer cells to radiation, providing a new basis for the application of autophagy in clinical tumor radiotherapy.
Collapse
Affiliation(s)
- Lei Li
- Cancer Research Institute, Hengyang Medical College of University of South China, No. 28, West Changsheng Road, Hengyang City, Hunan Province, China
| | - Wen-Ling Liu
- Cancer Research Institute, Hengyang Medical College of University of South China, No. 28, West Changsheng Road, Hengyang City, Hunan Province, China
| | - Lei Su
- Cancer Research Institute, Hengyang Medical College of University of South China, No. 28, West Changsheng Road, Hengyang City, Hunan Province, China
| | - Zhou-Cheng Lu
- Second Affiliated Hospital of South China University, Hengyang City, Hunan Province, China
| | - Xiu-Sheng He
- Cancer Research Institute, Hengyang Medical College of University of South China, No. 28, West Changsheng Road, Hengyang City, Hunan Province, China
| |
Collapse
|
12
|
Eichelmann AK, Mayne GC, Chiam K, Due SL, Bastian I, Butz F, Wang T, Sykes PJ, Clemons NJ, Liu DS, Michael MZ, Karapetis CS, Hummel R, Watson DI, Hussey DJ. Mutant p53 Mediates Sensitivity to Cancer Treatment Agents in Oesophageal Adenocarcinoma Associated with MicroRNA and SLC7A11 Expression. Int J Mol Sci 2021; 22:ijms22115547. [PMID: 34074015 PMCID: PMC8197322 DOI: 10.3390/ijms22115547] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/12/2021] [Accepted: 05/19/2021] [Indexed: 12/18/2022] Open
Abstract
TP53 gene mutations occur in 70% of oesophageal adenocarcinomas (OACs). Given the central role of p53 in controlling cellular response to therapy we investigated the role of mutant (mut-) p53 and SLC7A11 in a CRISPR-mediated JH-EsoAd1 TP53 knockout model. Response to 2 Gy irradiation, cisplatin, 5-FU, 4-hydroxytamoxifen, and endoxifen was assessed, followed by a TaqMan OpenArray qPCR screening for differences in miRNA expression. Knockout of mut-p53 resulted in increased chemo- and radioresistance (2 Gy survival fraction: 38% vs. 56%, p < 0.0001) and in altered miRNA expression levels. Target mRNA pathways analyses indicated several potential mechanisms of treatment resistance. SLC7A11 knockdown restored radiosensitivity (2 Gy SF: 46% vs. 73%; p = 0.0239), possibly via enhanced sensitivity to oxidative stress. Pathway analysis of the mRNA targets of differentially expressed miRNAs indicated potential involvement in several pathways associated with apoptosis, ribosomes, and p53 signaling pathways. The data suggest that mut-p53 in JH-EsoAd1, despite being classified as non-functional, has some function related to radio- and chemoresistance. The results also highlight the important role of SLC7A11 in cancer metabolism and redox balance and the influence of p53 on these processes. Inhibition of the SLC7A11-glutathione axis may represent a promising approach to overcome resistance associated with mut-p53.
Collapse
Affiliation(s)
- Ann-Kathrin Eichelmann
- Flinders Health and Medical Research Institute—Cancer Program, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (G.C.M.); (K.C.); (S.L.D.); (I.B.); (F.B.); (T.W.); (P.J.S.); (M.Z.M.); (C.S.K.); (D.I.W.)
- Department of General, Visceral and Transplant Surgery, University Hospital of Münster, Waldeyerstrasse 1, 48149 Münster, Germany
- Correspondence: (A.-K.E.); (D.J.H.)
| | - George C. Mayne
- Flinders Health and Medical Research Institute—Cancer Program, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (G.C.M.); (K.C.); (S.L.D.); (I.B.); (F.B.); (T.W.); (P.J.S.); (M.Z.M.); (C.S.K.); (D.I.W.)
- Department of Surgery, Flinders Medical Centre, Bedford Park, Adelaide, SA 5042, Australia
| | - Karen Chiam
- Flinders Health and Medical Research Institute—Cancer Program, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (G.C.M.); (K.C.); (S.L.D.); (I.B.); (F.B.); (T.W.); (P.J.S.); (M.Z.M.); (C.S.K.); (D.I.W.)
| | - Steven L. Due
- Flinders Health and Medical Research Institute—Cancer Program, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (G.C.M.); (K.C.); (S.L.D.); (I.B.); (F.B.); (T.W.); (P.J.S.); (M.Z.M.); (C.S.K.); (D.I.W.)
| | - Isabell Bastian
- Flinders Health and Medical Research Institute—Cancer Program, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (G.C.M.); (K.C.); (S.L.D.); (I.B.); (F.B.); (T.W.); (P.J.S.); (M.Z.M.); (C.S.K.); (D.I.W.)
| | - Frederike Butz
- Flinders Health and Medical Research Institute—Cancer Program, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (G.C.M.); (K.C.); (S.L.D.); (I.B.); (F.B.); (T.W.); (P.J.S.); (M.Z.M.); (C.S.K.); (D.I.W.)
| | - Tingting Wang
- Flinders Health and Medical Research Institute—Cancer Program, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (G.C.M.); (K.C.); (S.L.D.); (I.B.); (F.B.); (T.W.); (P.J.S.); (M.Z.M.); (C.S.K.); (D.I.W.)
| | - Pamela J. Sykes
- Flinders Health and Medical Research Institute—Cancer Program, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (G.C.M.); (K.C.); (S.L.D.); (I.B.); (F.B.); (T.W.); (P.J.S.); (M.Z.M.); (C.S.K.); (D.I.W.)
| | - Nicholas J. Clemons
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; (N.J.C.); (D.S.L.)
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - David S. Liu
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; (N.J.C.); (D.S.L.)
- Department of Surgery, Austin Health, Heidelberg, VIC 3084, Australia
| | - Michael Z. Michael
- Flinders Health and Medical Research Institute—Cancer Program, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (G.C.M.); (K.C.); (S.L.D.); (I.B.); (F.B.); (T.W.); (P.J.S.); (M.Z.M.); (C.S.K.); (D.I.W.)
- Department of Gastroenterology, Flinders Medical Centre, Bedford Park, Adelaide, SA 5042, Australia
| | - Christos S. Karapetis
- Flinders Health and Medical Research Institute—Cancer Program, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (G.C.M.); (K.C.); (S.L.D.); (I.B.); (F.B.); (T.W.); (P.J.S.); (M.Z.M.); (C.S.K.); (D.I.W.)
- Department of Medical Oncology, Flinders Medical Centre, Bedford Park, Adelaide, SA 5042, Australia
| | - Richard Hummel
- Department of Surgery, University Hospital of Schleswig-Holstein, Ratzeburger Allee 160, 23538 Lübeck, Germany;
| | - David I. Watson
- Flinders Health and Medical Research Institute—Cancer Program, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (G.C.M.); (K.C.); (S.L.D.); (I.B.); (F.B.); (T.W.); (P.J.S.); (M.Z.M.); (C.S.K.); (D.I.W.)
- Department of Surgery, Flinders Medical Centre, Bedford Park, Adelaide, SA 5042, Australia
| | - Damian J. Hussey
- Flinders Health and Medical Research Institute—Cancer Program, Flinders University, Bedford Park, Adelaide, SA 5042, Australia; (G.C.M.); (K.C.); (S.L.D.); (I.B.); (F.B.); (T.W.); (P.J.S.); (M.Z.M.); (C.S.K.); (D.I.W.)
- Department of Surgery, Flinders Medical Centre, Bedford Park, Adelaide, SA 5042, Australia
- Correspondence: (A.-K.E.); (D.J.H.)
| |
Collapse
|
13
|
Wu K, Chen X, Chen X, Zhang S, Xu Y, Xia B, Ma S. Suberoylanilide hydroxamic acid enhances the radiosensitivity of lung cancer cells through acetylated wild-type and mutant p53-dependent modulation of mitochondrial apoptosis. J Int Med Res 2021; 49:300060520981545. [PMID: 33557658 PMCID: PMC7876760 DOI: 10.1177/0300060520981545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Objective Suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor, has
shown potential as a candidate radiosensitizer for many types of cancers.
This study aimed to explore the radiosensitization mechanism of SAHA in lung
cancer cells. Methods Mutations in p53 were generated by site-directed mutagenesis using polymerase
chain reaction. Transfection was performed to generate H1299 cells carrying
wild-type or mutant p53. The radiosensitizing enhancement ratio was
determined by clonogenic assays. Mitochondrial apoptosis was detected using
JC-1 staining and flow cytometry analysis. Results Our results showed that SAHA induced radiosensitization in H1299 cells
expressing wild-type p53, p53R175H or p53P223L, but
this enhanced clonogenic cell death was not observed in parental H1299
(p53-null) cells or H1299 cells expressing p53 with K120R, A161T and V274R
mutations. In SAHA-sensitized cells, mitochondrial apoptosis was induced
following exposure to irradiation. Additionally, we observed that a
secondary mutation at K120 (K120R) could eliminate p53-mediated
radiosensitization and mitochondrial apoptosis. Conclusions The results of this study suggest that wild-type and specific mutant forms of
p53 mediate SAHA-induced radiosensitization by regulating mitochondrial
apoptosis, and the stabilization of K120 acetylation by SAHA is the
molecular basis contributing to radiosensitization in lung cancer cells.
Collapse
Affiliation(s)
- Kan Wu
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Xueqin Chen
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Xufeng Chen
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, USA
| | - Shirong Zhang
- Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Yasi Xu
- Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Bing Xia
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Department of Oncology, Jiande Second People's Hospital, Jiande, P.R. China
| | - Shenglin Ma
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| |
Collapse
|
14
|
Lu Y, Liu B, Liu Y, Yu X, Cheng G. Dual effects of active ERK in cancer: A potential target for enhancing radiosensitivity. Oncol Lett 2020; 20:993-1000. [PMID: 32724338 PMCID: PMC7377092 DOI: 10.3892/ol.2020.11684] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/20/2020] [Indexed: 12/20/2022] Open
Abstract
Ionizing radiation (IR) is an important cancer treatment approach. However, radioresistance eventually occurs, resulting in poor outcomes in patients with cancer. Radioresistance is associated with multiple signaling pathways, particularly pro-survival signaling pathways. The extracellular signal-regulated kinase 1/2 (ERK1/2) cascade is an important signaling pathway that initiates several cellular processes and is regulated by various stimuli, including IR. Although numerous studies have demonstrated the pro-survival effects of active ERK, activation of ERK has also been associated with cell death, indicating that radiosensitization may occur by ERK stimulation. In this context, the present review describes the associations between ERK signaling, cancer and IR, and discusses the association between ERK and its pro-survival function in cancer cells, including stimuli, molecular mechanisms, clinical use of inhibitors and underlying limitations. Additionally, the present review introduces the view that active ERK may induce cell death, and describes the potential factors associated with this process. This review describes the various outcomes induced by active ERK to prompt future studies to aim to enhance radiosensitivity in the treatment of cancer.
Collapse
Affiliation(s)
- Yinliang Lu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Baocai Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Ying Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xinyue Yu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Guanghui Cheng
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
15
|
A Potent Autophagy Inhibitor (Lys05) Enhances the Impact of Ionizing Radiation on Human Lung Cancer Cells H1299. Int J Mol Sci 2019; 20:ijms20235881. [PMID: 31771188 PMCID: PMC6928878 DOI: 10.3390/ijms20235881] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/15/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy inhibition through small-molecule inhibitors is one of the approaches to increase the efficiency of radiotherapy in oncological patients. A new inhibitor-Lys05-with the potential to accumulate within lysosomes and to block autophagy was discovered a few years ago. Several studies have addressed its chemosensitizing effects but nothing is known about its impact in the context of ionizing radiation (IR). To describe its role in radiosensitization, we employed radioresistant human non-small cell lung carcinoma cells (H1299, p53-negative). Combined treatment of H1299 cells by Lys05 together with IR decreased cell survival in the clonogenic assay and real-time monitoring of cell growth more than either Lys05 or IR alone. Immunodetection of LC3 and p62/SQSTM1 indicated that autophagy was inhibited, which correlated with increased SQSTM1 and decreased BNIP3 gene expression determined by qRT-PCR. Fluorescence microscopy and flow cytometry uncovered an accumulation of lysosomes. Similarly, transmission electron microscopy demonstrated the accumulation of autophagosomes confirming the ability of Lys05 to potentiate autophagy inhibition in H1299 cells. We report here for the first time that Lys05 could be utilized in combination with IR as a promising future strategy in the eradication of lung cancer cells.
Collapse
|
16
|
Zhang WW, Li L, Li D, Liu J, Li X, Li W, Xu X, Zhang MJ, Chandler LA, Lin H, Hu A, Xu W, Lam DMK. The First Approved Gene Therapy Product for Cancer Ad-p53 (Gendicine): 12 Years in the Clinic. Hum Gene Ther 2019; 29:160-179. [PMID: 29338444 DOI: 10.1089/hum.2017.218] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Gendicine (recombinant human p53 adenovirus), developed by Shenzhen SiBiono GeneTech Co. Ltd., was approved in 2003 by the China Food and Drug Administration (CFDA) as a first-in-class gene therapy product to treat head and neck cancer, and entered the commercial market in 2004. Gendicine is a biological therapy that is delivered via minimally invasive intratumoral injection, as well as by intracavity or intravascular infusion. The wild-type (wt) p53 protein expressed by Gendicine-transduced cells is a tumor suppressor that is activated by cellular stress, and mediates cell-cycle arrest and DNA repair, or induces apoptosis, senescence, and/or autophagy, depending upon cellular stress conditions. Based on 12 years of commercial use in >30,000 patients, and >30 published clinical studies, Gendicine has exhibited an exemplary safety record, and when combined with chemotherapy and radiotherapy has demonstrated significantly higher response rates than for standard therapies alone. In addition to head and neck cancer, Gendicine has been successfully applied to treat various other cancer types and different stages of disease. Thirteen published studies that include long-term survival data showed that Gendicine combination regimens yield progression-free survival times that are significantly longer than standard therapies alone. Although the p53 gene is mutated in >50% of all human cancers, p53 mutation status did not significantly influence efficacy outcomes and long-term survival rate for Ad-p53-treated patients. To date, Shenzhen SiBiono GeneTech has manufactured 41 batches of Gendicine in compliance with CFDA QC/QA requirements, and 169,571 vials (1.0 × 1012 vector particles per vial) have been used to treat patients. No serious adverse events have been reported, except for vector-associated transient fever, which occurred in 50-60% of patients and persisted for only a few hours. The manufacturing accomplishments and clinical experience with Gendicine, as well as the understanding of its cellular mechanisms of action and implications, could provide valuable insights for the international gene therapy community and add valuable data to promote further developments and advancements in the gene therapy field.
Collapse
Affiliation(s)
- Wei-Wei Zhang
- 1 LifeTech Biosciences Group, Hong Kong .,2 Angionetics, Inc., San Diego, California
| | - Longjiang Li
- 3 State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dinggang Li
- 4 Beijing Haidian Hospital Center for Cancer Gene Therapy, Beijing, China
| | - Jiliang Liu
- 5 Shenzhen Hengsheng Hospital Cancer Center, Shenzhen, China
| | - Xiuqin Li
- 6 China Medical University Shengjing Hospital Department of Obstetrics and Gynecology, Shenyang, China
| | - Wei Li
- 7 Shenzhen SiBiono GeneTech Co. Ltd., Shenzhen, China
| | - Xiaolong Xu
- 7 Shenzhen SiBiono GeneTech Co. Ltd., Shenzhen, China
| | - Michael J Zhang
- 8 Department of Medicine University of Minnesota Medical School, Minneapolis, Minnesota
| | | | - Hong Lin
- 7 Shenzhen SiBiono GeneTech Co. Ltd., Shenzhen, China
| | - Aiguo Hu
- 7 Shenzhen SiBiono GeneTech Co. Ltd., Shenzhen, China
| | - Wei Xu
- 7 Shenzhen SiBiono GeneTech Co. Ltd., Shenzhen, China
| | | |
Collapse
|
17
|
Wu X, Zhang J, Ma C, Li W, Zeng J, Wang Y, Deng G. A role for Wnt/β-catenin signalling in suppressing Bacillus Calmette-Guerin-induced macrophage autophagy. Microb Pathog 2018; 127:277-287. [PMID: 30550847 DOI: 10.1016/j.micpath.2018.12.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/08/2018] [Accepted: 12/10/2018] [Indexed: 12/11/2022]
Abstract
Mycobacterium tuberculosis (Mtb)-induced autophagy of alveolar macrophages has been confirmed to play a central role in the pathogenesis of tuberculosis. Growing evidence indicates that excessive or uncontrolled autophagic activity, which results in type II programmed cell death, can be regulated by many factors, including Wnt/β-catenin signalling. Wnt/β-catenin signalling has been demonstrated to be involved in multiple diseases through the regulation of autophagy; however, its exact role in regulating autophagy induced by Mtb remains unclear. Accordingly, this study examined the function of the Wnt/β-catenin signalling pathway in regulating Mycobacterium bovis Bacillus Calmette-Guerin (BCG)-induced autophagy in RAW264.7 macrophage cell line. In the present study, we found that BCG induced the autophagy of RAW264.7 cells in a time- and dose-dependent manner along with an accumulation of LC3 (Microtubule-associated protein 1 light chain 3) protein. Intriguingly, Wnt3a, a Wnt/β-catenin signalling ligand, significantly inhibited autophagy, with decreased autophagy rates and autophagic flux. An immunoblot analysis further revealed that Wnt/β-catenin signalling was capable of inhibiting the expression of the LC3 and autophagy-associated gene (Atg) cascade proteins in BCG-infected cells. Mechanistically, Wnt/β-catenin signalling may inhibit autophagy in BCG-infected macrophages by activating mTOR-dependent pathways. Our findings reveal the mechanisms of Wnt/β-catenin signalling regulates cellular autophagy induced by Mtb and provide novel insights into physiological and immune control of tuberculosis by modulating autophagy processes.
Collapse
Affiliation(s)
- Xiaoling Wu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Jiamei Zhang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Chenjie Ma
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Wu Li
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Jin Zeng
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Yujiong Wang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China.
| | - Guangcun Deng
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, Yinchuan, 750021, Ningxia, China; College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China.
| |
Collapse
|
18
|
Udagawa H, Umemura S, Murakami I, Mimaki S, Makinoshima H, Ishii G, Miyoshi T, Kirita K, Matsumoto S, Yoh K, Niho S, Tsuchihara K, Goto K. Genetic profiling-based prognostic prediction of patients with advanced small-cell lung cancer in large scale analysis. Lung Cancer 2018; 126:182-188. [PMID: 30527185 DOI: 10.1016/j.lungcan.2018.11.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/17/2018] [Accepted: 11/10/2018] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Comprehensive genomic analysis of small-cell lung cancer (SCLC) revealed various genetic alterations. However, obtaining suitable samples for genetic analysis is difficult in advanced SCLC. Thus, the prognostic effect of genetic alterations on the outcome of SCLC patients has not been well investigated. Therefore, this study evaluated the effect of genetic alterations on the survival of SCLC patients. MATERIALS AND METHODS We collected samples obtained from 220 patients with advanced SCLC before cancer treatment. Genomic DNA extracted from the samples was subjected to a 1.499 Mb-sized custom panel that captured all exons of 244 cancer-related genes, and the captured DNA was analyzed through next-generation sequencing. The associations between genetic alterations and overall survival were evaluated. RESULTS Genetic analysis was successful in 204 samples (93%). Genetic alterations in the PI3K/AKT/mTOR pathway and inactivating mutations inTP53 and RB1 were detected in 14 (7%), 150 (74%), and 85 (42%) of the tumors. In extensive disease (ED, N = 126) patients, multivariate analysis revealed that the presence of genetic alterations in the PI3K/AKT/mTOR pathway was significantly associated with unfavorable survival [hazard ratio (HR), 2.14; 95% CI 1.02-4.06; P = 0.04]. In limited disease (LD, N = 78) patients, the presence of TP53 mutation and the absence of RB1 mutation were significantly associated with unfavorable survival (HR, 2.41; 95% CI 1.21-5.34; P = 0.01, and HR, 0.45; 95% CI 0.25-0.79; P < 0.01, respectively). CONCLUSIONS Sequencing-based genetic profiling is feasible and useful to predict the prognosis in advanced SCLC. Genetic alterations in the PI3K/AKT/mTOR pathway, TP53 mutations and RB1 mutations were associated with prognosis in SCLC patients. The genetic alterations associated with the prognosis were different between ED-SCLC and LD-SCLC.
Collapse
Affiliation(s)
- Hibiki Udagawa
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan; Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.
| | - Shigeki Umemura
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan; Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Isao Murakami
- Department of Respiratory Medicine, Higashi-Hiroshima Medical Center, Higashi-Hiroshima, Japan
| | - Sachiyo Mimaki
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Hideki Makinoshima
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Genichiro Ishii
- Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Tomohiro Miyoshi
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Keisuke Kirita
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shingo Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan; Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Kiyotaka Yoh
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Seiji Niho
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Katsuya Tsuchihara
- Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
19
|
Xu J, Patel NH, Saleh T, Cudjoe EK, Alotaibi M, Wu Y, Lima S, Hawkridge AM, Gewirtz DA. Differential Radiation Sensitivity in p53 Wild-Type and p53-Deficient Tumor Cells Associated with Senescence but not Apoptosis or (Nonprotective) Autophagy. Radiat Res 2018; 190:538-557. [PMID: 30132722 DOI: 10.1667/rr15099.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Studies of radiation interaction with tumor cells often focus on apoptosis as an end point; however, clinically relevant doses of radiation also promote autophagy and senescence. Moreover, functional p53 has frequently been implicated in contributing to radiation sensitivity through the facilitation of apoptosis. To address the involvement of apoptosis, autophagy, senescence and p53 status in the response to radiation, the current studies utilized isogenic H460 non-small cell lung cancer cells that were either p53-wild type (H460wt) or null (H460crp53). As anticipated, radiosensitivity was higher in the H460wt cells than in the H460crp53 cell line; however, this differential radiation sensitivity did not appear to be a consequence of apoptosis. Furthermore, radiosensitivity did not appear to be reduced in association with the promotion of autophagy, as autophagy was markedly higher in the H460wt cells. Despite radiosensitization by chloroquine in the H460wt cells, the radiation-induced autophagy proved to be essentially nonprotective, as inhibition of autophagy via 3-methyl adenine (3-MA), bafilomycin A1 or ATG5 silencing failed to alter radiation sensitivity or promote apoptosis in either the H460wt or H460crp53 cells. Radiosensitivity appeared to be most closely associated with senescence, which occurred earlier and to a greater extent in the H460wt cells. This finding is consistent with the in-depth proteomics analysis on the secretomes from the H460wt and H460crp53 cells (with or without radiation exposure) that showed no significant association with radioresistance-related proteins, whereas several senescence-associated secretory phenotype (SASP) factors were upregulated in H460wt cells relative to H460crp53 cells. Taken together, these findings indicate that senescence, rather than apoptosis, plays a central role in determination of radiosensitivity; furthermore, autophagy is likely to have minimal influence on radiosensitivity under conditions where autophagy takes the nonprotective form.
Collapse
Affiliation(s)
- Jingwen Xu
- a Department of Pharmacology and Toxicology, Shenyang Pharmaceutical University, Liaoning, China
| | - Nipa H Patel
- b Department of Pharmacology and Toxicology and Medicine, Virginia Commonwealth University, Richmond, Virginia.,e Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Tareq Saleh
- b Department of Pharmacology and Toxicology and Medicine, Virginia Commonwealth University, Richmond, Virginia.,e Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Emmanuel K Cudjoe
- c Department of Pharmacotherapy and Outcome Sciences and Pharmaceutics, Virginia Commonwealth University, Richmond, Virginia
| | - Moureq Alotaibi
- f Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Yingliang Wu
- a Department of Pharmacology and Toxicology, Shenyang Pharmaceutical University, Liaoning, China
| | - Santiago Lima
- d Department of Biology, Virginia Commonwealth University, Richmond, Virginia.,e Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Adam M Hawkridge
- c Department of Pharmacotherapy and Outcome Sciences and Pharmaceutics, Virginia Commonwealth University, Richmond, Virginia.,e Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - David A Gewirtz
- b Department of Pharmacology and Toxicology and Medicine, Virginia Commonwealth University, Richmond, Virginia.,e Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
20
|
Simabuco FM, Morale MG, Pavan IC, Morelli AP, Silva FR, Tamura RE. p53 and metabolism: from mechanism to therapeutics. Oncotarget 2018; 9:23780-23823. [PMID: 29805774 PMCID: PMC5955117 DOI: 10.18632/oncotarget.25267] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/06/2018] [Indexed: 11/25/2022] Open
Abstract
The tumor cell changes itself and its microenvironment to adapt to different situations, including action of drugs and other agents targeting tumor control. Therefore, metabolism plays an important role in the activation of survival mechanisms to keep the cell proliferative potential. The Warburg effect directs the cellular metabolism towards an aerobic glycolytic pathway, despite the fact that it generates less adenosine triphosphate than oxidative phosphorylation; because it creates the building blocks necessary for cell proliferation. The transcription factor p53 is the master tumor suppressor; it binds to more than 4,000 sites in the genome and regulates the expression of more than 500 genes. Among these genes are important regulators of metabolism, affecting glucose, lipids and amino acids metabolism, oxidative phosphorylation, reactive oxygen species (ROS) generation and growth factors signaling. Wild-type and mutant p53 may have opposing effects in the expression of these metabolic genes. Therefore, depending on the p53 status of the cell, drugs that target metabolism may have different outcomes and metabolism may modulate drug resistance. Conversely, induction of p53 expression may regulate differently the tumor cell metabolism, inducing senescence, autophagy and apoptosis, which are dependent on the regulation of the PI3K/AKT/mTOR pathway and/or ROS induction. The interplay between p53 and metabolism is essential in the decision of cell fate and for cancer therapeutics.
Collapse
Affiliation(s)
- Fernando M. Simabuco
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Mirian G. Morale
- Center for Translational Investigation in Oncology/LIM24, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Isadora C.B. Pavan
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Ana P. Morelli
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Fernando R. Silva
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rodrigo E. Tamura
- Center for Translational Investigation in Oncology/LIM24, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
21
|
Khaodee W, Inboot N, Udomsom S, Kumsaiyai W, Cressey R. Glucosidase II beta subunit (GluIIβ) plays a role in autophagy and apoptosis regulation in lung carcinoma cells in a p53-dependent manner. Cell Oncol (Dordr) 2017; 40:579-591. [PMID: 28929344 DOI: 10.1007/s13402-017-0349-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2017] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Glucosidase II plays a major role in regulating the post-translational modification of N-linked glycoproteins. Previously, we found that the beta subunit of glucosidase II (GluIIβ) levels are significantly increased in lung carcinoma tissues, indicating a potential role in lung tumorigenesis. Here, we investigated the role of GluIIβ in the regulation of autophagy and apoptosis in lung carcinoma- and immortalized human bronchial epithelial-derived cells. METHODS A selective glucosidase II inhibitor, bromoconduritol, was used to inhibit GluII enzyme activity and a siRNA-based technology was used to suppress the expression of the GluIIβ encoding gene PRKCSH in lung carcinoma cells differing in p53 status. Cell viability was assessed using a MTT assay, cell cycle progression was assessed using flow cytometry, autophagy was assessed using Western blotting and apoptosis was assessed using an annexin V-FITC/PI double labeling method. RESULTS We found that GluIIβ inhibition resulted in the induction of autophagy in all cell lines tested, but apoptosis in only wild-type p53 cells. We also found that GluIIβ inhibition dose-dependently decreased activation of the EGFR/RTK and PI3K/AKT signaling pathways. Although the apoptosis inducing effect of GluIIβ inhibition appeared to be p53-dependent, we found that a combined treatment with lysosomal inhibitors to block autophagy enhanced the apoptotic effect of GluIIβ inhibition in both wild-type p53 and p53-null cells. CONCLUSIONS Our data indicate that GluIIβ inhibition results in autophagy and apoptosis in lung carcinoma-derived cells, supporting the hypothesis that this enzyme may play a role in blocking these two tumor suppressive processes. Since blocking autophagy by lysosomal inhibitors enhanced the apoptosis-inducing effect of bromoconduritol, independent of p53 status, their combined use may hold promise for the treatment of cancer, particularly lung cancer.
Collapse
Affiliation(s)
- Worapong Khaodee
- Division of Clinical Chemistry, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Nichanan Inboot
- Division of Clinical Chemistry, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Suruk Udomsom
- Biomedical Engineering Program, Faculty of Engineering, Chiang Mai University, Chiang Mai, Thailand.,Biomedical Engineering Center, Chiang Mai University, Chiang Mai, Thailand
| | - Warunee Kumsaiyai
- Division of Clinical Chemistry, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Ratchada Cressey
- Division of Clinical Chemistry, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand. .,MT Cancer Research Unit, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
22
|
Abstract
Oncolytic virus (OV) therapy utilizes replication-competent viruses to kill cancer cells, leaving non-malignant cells unharmed. With the first U.S. Food and Drug Administration-approved OV, dozens of clinical trials ongoing, and an abundance of translational research in the field, OV therapy is poised to be one of the leading treatments for cancer. A number of recombinant OVs expressing a transgene for p53 (TP53) or another p53 family member (TP63 or TP73) were engineered with the goal of generating more potent OVs that function synergistically with host immunity and/or other therapies to reduce or eliminate tumor burden. Such transgenes have proven effective at improving OV therapies, and basic research has shown mechanisms of p53-mediated enhancement of OV therapy, provided optimized p53 transgenes, explored drug-OV combinational treatments, and challenged canonical roles for p53 in virus-host interactions and tumor suppression. This review summarizes studies combining p53 gene therapy with replication-competent OV therapy, reviews preclinical and clinical studies with replication-deficient gene therapy vectors expressing p53 transgene, examines how wild-type p53 and p53 modifications affect OV replication and anti-tumor effects of OV therapy, and explores future directions for rational design of OV therapy combined with p53 gene therapy.
Collapse
|
23
|
Yang YM, Fang F, Li X, Yu L, Wang ZC. TRAIL overexpression co-regulated by Egr1 and HRE enhances radiosensitivity of hypoxic A549 cells depending on its apoptosis inducing role. Oncol Rep 2016; 37:533-539. [PMID: 27878298 DOI: 10.3892/or.2016.5271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/17/2016] [Indexed: 11/05/2022] Open
Abstract
Ionizing radiation can upregulate the expression levels of TRAIL and enhance tumor cell apoptosis. While Early growth response 1 (Egr1) gene promoter has radiation inducible characteristics, the expression for exogenous gene controlled by Egr1 promoter could be enhanced by ionizing radiation, but its efficiency is limited by tissue hypoxia. Hypoxia response elements (HREs) are important hypoxic response regulatory sequences and sensitivity enhancers. Therefore, we chose TRAIL as the gene radiotherapy to observe whether it is regulated by Egr1 and HER and its effects on A549 cells and its mechanism. The pcDNA3.1-Egr1-TRAIL (pc-E-hsT) and pcDNA3.1-HRE/Egr1-TRAIL (pc-H/E-hsT) plasmids containing Egr1-hsTRAIL and HRE/Egr1-hsTRAIL were transfected into A549 cells, the cells were treated by hypoxia and radiation. The TRAIL mRNA in the cells and protein concentration in the culture supernatants were measured by RT-PCR and ELISA, respectively. Mean lethal dose D0 value was evaluated with colony forming assay. The cell apoptotic rates were analyzed by FCM and TUNEL assay. Expression of DR4, DR5 and cleaved caspase-3 proteins were analyzed by western blotting. It showed that TRAIL mRNA expression and TRAIL concentration all significantly increased under hypoxia and/or radiation. D0 value of pc-H/E‑hsT transfected cells under hypoxia was lowest, indicating more high radiosensitivity. Hypoxia could not cause the pc-E-hsT transfected cell apoptotic rate increase, but there were promoting effects in pc-H/E-hsT transfected cells. DR4 had not obvious change in pc-E-hsT and pc-H/E-hsT transfected cells under normoxic and hypoxic condition, otherwise, DR5 and cleaved caspase-3 increased mostly in pc-H/E-hsT transfected cells under hypoxic condition. TRAIL overexpression was co-regulated by Egr1 and HRE. TRAIL might promote hypoxic A549 cell radiosensitivity and induce apoptosis depending on DR5 to caspase-3 pathways.
Collapse
Affiliation(s)
- Yan-Ming Yang
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Fang Fang
- Key Laboratory of Radiobiology, Ministry of Health, School of Public Health, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xin Li
- Key Laboratory of Radiobiology, Ministry of Health, School of Public Health, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lei Yu
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Zhi-Cheng Wang
- Key Laboratory of Radiobiology, Ministry of Health, School of Public Health, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
24
|
Liu HC, Ma F, Shen Y, Hu YQ, Pan S. Overexpression of SMAR1 Enhances Radiosensitivity in Human Breast Cancer Cell Line MCF7 via Activation of p53 Signaling Pathway. Oncol Res 2016; 22:293-300. [PMID: 26629941 PMCID: PMC7842601 DOI: 10.3727/096504015x14424348426035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This study sought to investigate the effect of overexpression of SMAR1 (scaffold/matrix-associated region-binding protein 1) on cell radiosensitivity in breast cancer, as well as elucidate its regulatory mechanism. We constructed a lentiviral expression system to successfully overexpress SMAR1 in human breast cancer cell line MCF7. In addition, overexpression of SMAR1 in MCF7 cells enhanced the radiosensitivity to (89)SrCl2. Moreover, overexpression of SMAR1 significantly induced cell apoptosis rate and G2/M phase arrest under the irradiation of (89)SrCl2. In addition, Western blot analysis showed that overexpression of SMAR1 in MCF cells significantly increased the expression levels of pP53 (ser15), pP53 (ser20), acP53, and p21 and obviously decreased the expression of MDM2 under the irradiation of (89)SrCl2. Notably, these expression changes could be neutralized by PFTα, an inhibitor of p53 signaling pathway that could inhibit p53-dependent transactivation of p53-responsive genes. Therefore, overexpression of SMAR1 may increase radiosensitivity to (89)SrCl2 in breast cancer cell line MCF7 by p53-dependent G2/M checkpoint arrest and apoptosis. Enhanced expression of SMAR1 in tumors will help to improve the clinical efficiency of radiation therapy.
Collapse
Affiliation(s)
- Heng-chao Liu
- Department of Nuclear Medicine, First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui, China
| | | | | | | | | |
Collapse
|
25
|
Cui D, Xiong X, Zhao Y. Cullin-RING ligases in regulation of autophagy. Cell Div 2016; 11:8. [PMID: 27293474 PMCID: PMC4902950 DOI: 10.1186/s13008-016-0022-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/27/2016] [Indexed: 12/25/2022] Open
Abstract
Cullin-RING ligases (CRLs), the largest E3 ubiquitin ligase family, promote ubiquitination and degradation of various cellular key regulators involved in a broad array of physiological and pathological processes, including cell cycle progression, signal transduction, transcription, cardiomyopathy, and tumorigenesis. Autophagy, an intracellular catabolic reaction that delivers cytoplasmic components to lysosomes for degradation, is crucial for cellular metabolism and homeostasis. The dysfunction of autophagy has been proved to associate with a variety of human diseases. Recent evidences revealed the emerging roles of CRLs in the regulation of autophagy. In this review, we will focus mainly on recent advances in our understandings of the regulation of autophagy by CRLs and the cross-talk between CRLs and autophagy, two degradation systems. We will also discuss the pathogenesis of human diseases associated with the dysregulation of CRLs and autophagy. Finally, we will discuss current efforts and future perspectives on basic and translational research on CRLs and autophagy.
Collapse
Affiliation(s)
- Danrui Cui
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qing-Chun Road, Hangzhou, Zhejiang 310003 People's Republic of China ; Institute of Translational Medicine, Zhejiang University School of Medicine, 268 Kai-Xuan Road, Hangzhou, Zhejiang 310029 People's Republic of China
| | - Xiufang Xiong
- Institute of Translational Medicine, Zhejiang University School of Medicine, 268 Kai-Xuan Road, Hangzhou, Zhejiang 310029 People's Republic of China
| | - Yongchao Zhao
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qing-Chun Road, Hangzhou, Zhejiang 310003 People's Republic of China ; Institute of Translational Medicine, Zhejiang University School of Medicine, 268 Kai-Xuan Road, Hangzhou, Zhejiang 310029 People's Republic of China
| |
Collapse
|
26
|
Ondrej M, Cechakova L, Durisova K, Pejchal J, Tichy A. To live or let die: Unclear task of autophagy in the radiosensitization battle. Radiother Oncol 2016; 119:265-75. [PMID: 26993419 DOI: 10.1016/j.radonc.2016.02.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/26/2016] [Accepted: 02/18/2016] [Indexed: 02/06/2023]
Abstract
Radiation-induced autophagy is believed to represent a radioprotective mechanism of cancer cells. Thus, its inhibition should support radiation treatment and increase its efficacy. On the other hand, there is evidence that radiation alone or in combination with various chemical agents can induce autophagy that results into increased cell death, especially within transformed apoptosis-resistant cells. In this paper, besides description of autophagic process and its relation to cancer and radiotherapy, we compared two contradictory radiosensitization approaches that employ inhibition and induction of autophagy. In spite of the classical concept based on cytoprotective model, there is a plethora of recently developed inducers of autophagy, which indicates the future trend in radiosensitization via modulation of autophagy. Because contemporary literature is conflicting and inconsistent in this respect, we reviewed the recent studies focused on enhancement of sensitivity of cancer cells toward radiation in regard to autophagy, revealing some striking discrepancies. The deeper the knowledge, the more complex this situation is. To interpret results of various studies correctly one has to take into account the methodology of autophagy assessment and also the fact that radiosensitization might be mediated by other than intrinsic mechanisms related to autophagy. Notwithstanding, targeting autophagy remains an attractive anti-tumor strategy.
Collapse
Affiliation(s)
- Martin Ondrej
- Department of Radiobiology, Faculty of Military Health Sciences in Hradec Kralove, University of Defense in Brno, Czech Republic
| | - Lucie Cechakova
- Department of Radiobiology, Faculty of Military Health Sciences in Hradec Kralove, University of Defense in Brno, Czech Republic
| | - Kamila Durisova
- Department of Radiobiology, Faculty of Military Health Sciences in Hradec Kralove, University of Defense in Brno, Czech Republic
| | - Jaroslav Pejchal
- Department of Radiobiology, Faculty of Military Health Sciences in Hradec Kralove, University of Defense in Brno, Czech Republic
| | - Ales Tichy
- Department of Radiobiology, Faculty of Military Health Sciences in Hradec Kralove, University of Defense in Brno, Czech Republic; Centre of Biomedical Research, University Hospital, Hradec Kralove, Czech Republic.
| |
Collapse
|
27
|
Li Y, Liu F, Wang Y, Li D, Guo F, Xu L, Zeng Z, Zhong X, Qian K. Rapamycin-induced autophagy sensitizes A549 cells to radiation associated with DNA damage repair inhibition. Thorac Cancer 2016; 7:379-86. [PMID: 27385978 PMCID: PMC4930955 DOI: 10.1111/1759-7714.12332] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/14/2015] [Indexed: 01/09/2023] Open
Abstract
Background Autophagy has been reported to increase in cancer cells after radiation. However, it remains unknown whether increased autophagy as a result of radiation affects DNA damage repair and sensitizes cancer cells. In this study, the radiosensitization effect of rapamycin, a mammalian target of rapamycin inhibitor that induces autophagy, on human lung adenocarcinoma A549 cells was investigated. Methods A549 cells were treated with different concentrations of rapamycin. Cell viability was evaluated by methyl‐thiazolyl‐tetrazolium assay. Survival fraction values of A549 cells after radiotherapy were detected by colony formation assay. Autophagosome was observed by a transmission electron microscope. Furthermore, Western blot was employed to examine alterations in autophagy protein LC3 and p62, DNA damage protein γ–H2AX, and DNA damage repair proteins Rad51, Ku70, and Ku80. Rad51, Ku70, and Ku80 messenger ribonucleic acid (mRNA) expression levels were examined by real‐time polymerase chain reaction. Results Rapamycin suppressed A549 cell proliferation in dose and time‐dependent manners. An inhibitory concentration (IC)10 dose of rapamycin could induce autophagy in A549 cells. Rapamycin combined with radiation significantly decreased the colony forming ability of cells, compared with rapamycin or radiation alone. Rapamycin and radiation combined increased γ–H2AX expression levels and decreased Rad51 and Ku80 expression levels, compared with single regimens. However, rapamycin treatment did not induce any change in Rad51, Ku70, and Ku80 mRNA levels, regardless of radiation. Conclusions These findings indicate that increasing autophagy sensitizes lung cancer cells to radiation.
Collapse
Affiliation(s)
- Yong Li
- Department of Medical Oncology The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Fen Liu
- Critical Care Medicine The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Yong Wang
- Department of Medical Oncology The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Donghai Li
- Department of Neurosurgery The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Fei Guo
- The institute of Burn Research The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Liyao Xu
- Department of Medical Oncology The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Zhengguo Zeng
- Critical Care Medicine The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Xiaojun Zhong
- Department of Medical Oncology The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Kejian Qian
- Critical Care Medicine The First Affiliated Hospital of Nanchang University Nanchang Jiangxi China
| |
Collapse
|
28
|
Ye F, Zhao T, Liu X, Jin X, Liu X, Wang T, Li Q. Long-term Autophagy and Nrf2 Signaling in the Hippocampi of Developing Mice after Carbon Ion Exposure. Sci Rep 2015; 5:18636. [PMID: 26689155 PMCID: PMC4686898 DOI: 10.1038/srep18636] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/20/2015] [Indexed: 12/29/2022] Open
Abstract
To explore charged particle radiation-induced long-term hippocampus damage, we investigated the expression of autophagy and antioxidant Nrf2 signaling-related proteins in the mouse hippocampus after carbon ion radiation. Heads of immature female Balb/c mice were irradiated with carbon ions of different LETs at various doses. Behavioral tests were performed on the mice after maturation. Acute and chronic expression of LC3-II, p62/SQSTM1, nuclear Nrf2, activated caspase-3 and the Bax/Bcl-2 ratio were measured in the hippocampi. Secondary X-ray insult was adopted to amplify potential damages. Long-term behavioral changes were observed in high-LET carbon ion-irradiated mice. There were no differences in the rates of LC3-II induction and p62/SQSTM1 degradation compared to the control group regardless of whether the mice received the secondary X-ray insult. A high nuclear Nrf2 content and low apoptosis level in hippocampal cells subjected to secondary X-rays were observed for the mice exposed to relatively low-LET carbon ions. Therefore, carbon ion exposure in the immature mouse led to an LET-dependent behavioral change after maturation. Although autophagy was intact, the persistently high nuclear Nrf2 content in the hippocampus might account for the unchanged behavioral pattern in mice exposed to the relatively low-LET carbon ions and the subsequent increased radioresistance of the hippocampus.
Collapse
Affiliation(s)
- Fei Ye
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China.,Department of Modern Physics, Lanzhou University, Lanzhou 730000, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
| | - Ting Zhao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
| | - Xiongxiong Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
| | - Xinguo Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
| | - Tieshan Wang
- Department of Modern Physics, Lanzhou University, Lanzhou 730000, China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
| |
Collapse
|
29
|
Sharma K, Goehe RW, Di X, Hicks MA, Torti SV, Torti FM, Harada H, Gewirtz DA. A novel cytostatic form of autophagy in sensitization of non-small cell lung cancer cells to radiation by vitamin D and the vitamin D analog, EB 1089. Autophagy 2015; 10:2346-61. [PMID: 25629933 DOI: 10.4161/15548627.2014.993283] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The standard of care for unresectable lung cancer is chemoradiation. However, therapeutic options are limited and patients are rarely cured. We have previously shown that vitamin D and vitamin D analogs such as EB 1089 can enhance the response to radiation in breast cancer through the promotion of a cytotoxic form of autophagy. In A549 and H460 non-small cell lung cancer (NSCLC) cells, 1,25-D3 (the hormonally active form of vitamin D) and EB 1089 prolonged the growth arrest induced by radiation alone and suppressed proliferative recovery, which translated to a significant reduction in clonogenic survival. In H838 or H358 NSCLC cells, which lack VDR/vitamin D receptor or functional TP53, respectively, 1,25-D3 failed to modify the extent of radiation-induced growth arrest or suppress proliferative recovery post-irradiation. Sensitization to radiation in H1299 NSCLC cells was evident only when TP53 was induced in otherwise tp53-null H1299 NSCLC cells. Sensitization was not associated with increased DNA damage, decreased DNA repair or an increase in apoptosis, necrosis, or senescence. Instead sensitization appeared to be a consequence of the conversion of the cytoprotective autophagy induced by radiation alone to a novel cytostatic form of autophagy by the combination of 1,25-D3 or EB 1089 with radiation. While both pharmacological and genetic suppression of autophagy or inhibition of AMPK phosphorylation sensitized the NSCLC cells to radiation alone, inhibition of the cytostatic autophagy induced by the combination treatment reversed sensitization. Evidence for selectivity was provided by lack of radiosensitization in normal human bronchial cells and cardiomyocytes. Taken together, these studies have identified a unique cytostatic function of autophagy that appears to be mediated by VDR, TP53, and possibly AMPK in the promotion of an enhanced response to radiation by 1,25-D3 and EB 1089 in NSCLC.
Collapse
Key Words
- ACTB, actin, β
- AMPK, AMP activated protein kinase
- ANXA5, annexin A5
- ATG5, autophagy related 5
- AVO, acidic vesicular organelles
- BECN1, Beclin 1, autophagy-related
- Baf, bafilomycin A1
- FACS, fluorescence activating cell sorting
- GFP, green fluorescent protein; H2AFX/H2AX, H2A histone family, member X
- GLB, galactosidase
- MAP1LC3/LC3, microtubule-associated protein 1 light chain 3
- NSCLC
- NSCLC, non-small cell lung cancer
- PI, propidium iodide
- SQSTM1, sequestosome 1
- TP53, tumor protein p53
- VDR, vitamin D (1, 25-dihydroxyvitamin D3) receptor
- WT, wild-type; LC3 and LC3-II acronyms have been used to indicate the LC3B and LC3BII isoforms respectively
- autophagy
- cytoprotective
- cytostatic
- radiation
- β
Collapse
Affiliation(s)
- Khushboo Sharma
- a Department of Pharmacology and Pharmacology; Massey Cancer Center ; Virginia Commonwealth University ; Richmond , VA USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Sakai K, Kazama S, Nagai Y, Murono K, Tanaka T, Ishihara S, Sunami E, Tomida S, Nishio K, Watanabe T. Chemoradiation provides a physiological selective pressure that increases the expansion of aberrant TP53 tumor variants in residual rectal cancerous regions. Oncotarget 2015; 5:9641-9. [PMID: 25275295 PMCID: PMC4259426 DOI: 10.18632/oncotarget.2438] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Neoadjuvant chemoradiotherapy has been introduced in patients with surgically resected rectal cancer and reduced the local recurrence. Heterogeneity exists in rectal cancer, and we hypothesized that there are subclones resistant to chemoradiotherapy within the cancer mass. We performed DNA-targeted sequencing of pre- and post-treatment tumor tissues obtained from 20 rectal cancer patients who received chemoradiotherapy. The variant frequency of the mutant clones was compared between pre- and post-treatment samples of nine non-responder patients. RNA-targeted sequencing of 57 genes related to sensitivity to chemotherapy and radiotherapy was performed for the paired samples. Immunohistochemical analyses of p53 expression were also performed on the paired samples from the nine non-responder patients. DNA-sequencing detected frequent mutations of suppressor genes including TP53, APC and FBXW7 in the post-treatment samples of the nine non-responders. The frequency of TP53 mutations showed significant increases after chemoradiotherapy. RNA-targeted sequencing of 29 tumor tissues demonstrated that decreased expression of three genes and increased expression of four genes were detected in the post-treatment samples. Significantly increased expression of TP53 was observed in the post-treatment samples. Immunohistochemical staining for p53 revealed that increased p53 intensity scores were observed after chemoradiotherapy. These results suggest that the tumors with TP53 mutations tend to accumulate through chemoradiotherapy.
Collapse
|
31
|
Chakradeo S, Sharma K, Alhaddad A, Bakhshwin D, Le N, Harada H, Nakajima W, Yeudall WA, Torti SV, Torti FM, Gewirtz DA. Yet another function of p53--the switch that determines whether radiation-induced autophagy will be cytoprotective or nonprotective: implications for autophagy inhibition as a therapeutic strategy. Mol Pharmacol 2015; 87:803-14. [PMID: 25667224 PMCID: PMC4407732 DOI: 10.1124/mol.114.095273] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 02/09/2015] [Indexed: 01/28/2023] Open
Abstract
The influence of autophagy inhibition on radiation sensitivity was studied in human breast, head and neck, and non-small cell lung cancer cell lines, in cell lines that were either wild type or mutant/null in p53, and in cells where p53 was inducible or silenced. Whereas ionizing radiation promoted autophagy in all tumor cell lines studied, pharmacological inhibition of autophagy and/or genetic silencing of autophagy genes failed to influence sensitivity to radiation in p53 mutant Hs578t breast tumor cells, HN6 head and neck tumor cells, and H358 non-small cell lung cancer cells. The requirement for functional p53 in the promotion of cytoprotective autophagy by radiation was confirmed by the observation that radiation-induced autophagy was nonprotective in p53 null H1299 cells but was converted to the cytoprotective form with induction of p53. Conversely, whereas p53 wild-type HN30 head and neck cancer cells did show sensitization to radiation upon autophagy inhibition, HN30 cells in which p53 was knocked down using small hairpin RNA failed to be sensitized by pharmacological autophagy inhibition. Taken together, these findings indicate that radiation-induced autophagy can be either cytoprotective or nonprotective, a functional difference related to the presence or absence of function p53. Alternatively, these findings could be interpreted to suggest that whereas radiation can induce autophagy independent of p53 status, inhibition of autophagy promotes enhanced radiation sensitivity through a mechanism that requires functional p53. These observations are likely to have direct implications with respect to clinical efforts to modulate the response of malignancies to radiation through autophagy inhibition.
Collapse
Affiliation(s)
- Shweta Chakradeo
- Departments of Medicine and Pharmacology and Toxicology, Massey Cancer Center (S.C., K.S., A.A., D.B., N.L., D.A.G.), and Department of Oral and Craniofacial Molecular Biology, School of Dentistry (H.H., W.N., W.A.Y.), Virginia Commonwealth University, Richmond, Virginia; and Departments of Molecular Biology and Biophysics (S.V.T.) and Medicine (F.M.T.), University of Connecticut Health Sciences, Farmington, Connecticut
| | - Khushboo Sharma
- Departments of Medicine and Pharmacology and Toxicology, Massey Cancer Center (S.C., K.S., A.A., D.B., N.L., D.A.G.), and Department of Oral and Craniofacial Molecular Biology, School of Dentistry (H.H., W.N., W.A.Y.), Virginia Commonwealth University, Richmond, Virginia; and Departments of Molecular Biology and Biophysics (S.V.T.) and Medicine (F.M.T.), University of Connecticut Health Sciences, Farmington, Connecticut
| | - Aisha Alhaddad
- Departments of Medicine and Pharmacology and Toxicology, Massey Cancer Center (S.C., K.S., A.A., D.B., N.L., D.A.G.), and Department of Oral and Craniofacial Molecular Biology, School of Dentistry (H.H., W.N., W.A.Y.), Virginia Commonwealth University, Richmond, Virginia; and Departments of Molecular Biology and Biophysics (S.V.T.) and Medicine (F.M.T.), University of Connecticut Health Sciences, Farmington, Connecticut
| | - Duaa Bakhshwin
- Departments of Medicine and Pharmacology and Toxicology, Massey Cancer Center (S.C., K.S., A.A., D.B., N.L., D.A.G.), and Department of Oral and Craniofacial Molecular Biology, School of Dentistry (H.H., W.N., W.A.Y.), Virginia Commonwealth University, Richmond, Virginia; and Departments of Molecular Biology and Biophysics (S.V.T.) and Medicine (F.M.T.), University of Connecticut Health Sciences, Farmington, Connecticut
| | - Ngoc Le
- Departments of Medicine and Pharmacology and Toxicology, Massey Cancer Center (S.C., K.S., A.A., D.B., N.L., D.A.G.), and Department of Oral and Craniofacial Molecular Biology, School of Dentistry (H.H., W.N., W.A.Y.), Virginia Commonwealth University, Richmond, Virginia; and Departments of Molecular Biology and Biophysics (S.V.T.) and Medicine (F.M.T.), University of Connecticut Health Sciences, Farmington, Connecticut
| | - Hisashi Harada
- Departments of Medicine and Pharmacology and Toxicology, Massey Cancer Center (S.C., K.S., A.A., D.B., N.L., D.A.G.), and Department of Oral and Craniofacial Molecular Biology, School of Dentistry (H.H., W.N., W.A.Y.), Virginia Commonwealth University, Richmond, Virginia; and Departments of Molecular Biology and Biophysics (S.V.T.) and Medicine (F.M.T.), University of Connecticut Health Sciences, Farmington, Connecticut
| | - Wataru Nakajima
- Departments of Medicine and Pharmacology and Toxicology, Massey Cancer Center (S.C., K.S., A.A., D.B., N.L., D.A.G.), and Department of Oral and Craniofacial Molecular Biology, School of Dentistry (H.H., W.N., W.A.Y.), Virginia Commonwealth University, Richmond, Virginia; and Departments of Molecular Biology and Biophysics (S.V.T.) and Medicine (F.M.T.), University of Connecticut Health Sciences, Farmington, Connecticut
| | - W Andrew Yeudall
- Departments of Medicine and Pharmacology and Toxicology, Massey Cancer Center (S.C., K.S., A.A., D.B., N.L., D.A.G.), and Department of Oral and Craniofacial Molecular Biology, School of Dentistry (H.H., W.N., W.A.Y.), Virginia Commonwealth University, Richmond, Virginia; and Departments of Molecular Biology and Biophysics (S.V.T.) and Medicine (F.M.T.), University of Connecticut Health Sciences, Farmington, Connecticut
| | - Suzy V Torti
- Departments of Medicine and Pharmacology and Toxicology, Massey Cancer Center (S.C., K.S., A.A., D.B., N.L., D.A.G.), and Department of Oral and Craniofacial Molecular Biology, School of Dentistry (H.H., W.N., W.A.Y.), Virginia Commonwealth University, Richmond, Virginia; and Departments of Molecular Biology and Biophysics (S.V.T.) and Medicine (F.M.T.), University of Connecticut Health Sciences, Farmington, Connecticut
| | - Frank M Torti
- Departments of Medicine and Pharmacology and Toxicology, Massey Cancer Center (S.C., K.S., A.A., D.B., N.L., D.A.G.), and Department of Oral and Craniofacial Molecular Biology, School of Dentistry (H.H., W.N., W.A.Y.), Virginia Commonwealth University, Richmond, Virginia; and Departments of Molecular Biology and Biophysics (S.V.T.) and Medicine (F.M.T.), University of Connecticut Health Sciences, Farmington, Connecticut
| | - David A Gewirtz
- Departments of Medicine and Pharmacology and Toxicology, Massey Cancer Center (S.C., K.S., A.A., D.B., N.L., D.A.G.), and Department of Oral and Craniofacial Molecular Biology, School of Dentistry (H.H., W.N., W.A.Y.), Virginia Commonwealth University, Richmond, Virginia; and Departments of Molecular Biology and Biophysics (S.V.T.) and Medicine (F.M.T.), University of Connecticut Health Sciences, Farmington, Connecticut
| |
Collapse
|
32
|
p53-mediated autophagic regulation: A prospective strategy for cancer therapy. Cancer Lett 2015; 363:101-7. [PMID: 25896632 DOI: 10.1016/j.canlet.2015.04.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/13/2015] [Accepted: 04/14/2015] [Indexed: 12/25/2022]
Abstract
Autophagy is a major catabolic process that degrades and recycles cytosolic components in autophagosomes, which fuse with lysosomes. This process enables starving cells to sustain their energy requirements and metabolic states, thus facilitating their survival, especially in cancer pathogenesis. The regulation of autophagy is quite intricate. It involves a series of signaling cascades including p53, known as the best-characterized tumor suppressor protein. Recent reports have indicated that p53 plays dual roles in regulating autophagy depending on its subcellular localization. Nuclear p53 facilitates autophagy by transactivating its target genes, whereas cytoplasmic p53 mainly inhibits autophagy through extranuclear, transcription-independent mechanisms. The relationship between autophagy and neoplasia is complicated. It may be intrinsically associated with the functional status of p53, but this is not clearly elucidated. This review focuses on the role of p53 as a master regulator of autophagy. We conclude that the contextual role of autophagy in cancer, which could be switched by p53 status, is expected to be developed into a new anticancer therapeutic approach.
Collapse
|
33
|
Zhong R, Xu H, Chen G, Zhao G, Gao Y, Liu X, Ma S, Dong L. The role of hypoxia-inducible factor-1α in radiation-induced autophagic cell death in breast cancer cells. Tumour Biol 2015; 36:7077-83. [PMID: 25874499 DOI: 10.1007/s13277-015-3425-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 04/06/2015] [Indexed: 12/21/2022] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a major effector in cell survival response to hypoxia, while the roles of HIF-1α in radiation-induced autophagy are still unclear in breast cancer cells. Human breast cancer carcinoma MCF-7 cells were stably transfected with pSUPER-shRNA against human HIF-1α or a scrambled sequence with no homology to mammalian genes, named as pSUPER-HIF-1α and pSUPER-SC, respectively. Cell Counting Kit-8 (CCK-8) assay and colony formation assay were used to detect cell viability, Western blot was used to detect protein expression, monodansylcadaverine (MDC) staining was used to analyze autophagy, and Hoechts/PI staining was used to assess apoptosis. Ionizing radiation (IR) and cobalt chloride (CoCl2) could induce HIF-1α expression and increase the microtubule-associated protein 1 light chain 3 (MAPLC3)-II/MAPLC3-I ratio, especially in radiation + CoCl2 group. After the silencing of HIF-1α, the radiosensitivity of MCF-7 cells increased and the autophagy level decreased in response to DNA damage induced by ionizing radiation, but there was no influence on IR-induced apoptosis. HIF-1α silencing also increased the expression of phospho-Akt, mTOR, and P70S6K and activated the mTOR signals significantly. Hypoxia can induce autophagy and also improve the IR-induced autophagy via the suppression of Akt/mTOR/P70S6K pathway, which consequently lead to radioresistance.
Collapse
Affiliation(s)
- Rui Zhong
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, 130021, China
| | - Huiying Xu
- Department of Ultrasound, The 1st Hospitals Affiliated to Jilin University, Changchun, 130021, China
| | - Ge Chen
- Department of Stomatology, The 1st Hospitals Affiliated to Jilin University, Changchun, 130021, China
| | - Gang Zhao
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, 130021, China
| | - Yan Gao
- Department of Radiation Oncology, The 1st Hospitals Affiliated to Jilin University, Changchun, 130021, China
| | - Xiaodong Liu
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, 130021, China.,Department of Radiation Oncology, The 1st Hospitals Affiliated to Jilin University, Changchun, 130021, China
| | - Shumei Ma
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, 130021, China. .,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3T 3T2, Canada.
| | - Lihua Dong
- Department of Radiation Oncology, The 1st Hospitals Affiliated to Jilin University, Changchun, 130021, China.
| |
Collapse
|
34
|
Xiao J, Zhang T, Xu D, Wang H, Cai Y, Jin T, Liu M, Jin M, Wu K, Yuan J. FBXL20-mediated Vps34 ubiquitination as a p53 controlled checkpoint in regulating autophagy and receptor degradation. Genes Dev 2015; 29:184-96. [PMID: 25593308 PMCID: PMC4298137 DOI: 10.1101/gad.252528.114] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vps34, the catalytic subunit in the class III phosphatidylinositol 3 kinase complexes, mediates the production of PtdIns3P, a key intracellular lipid involved in regulating autophagy and receptor degradation. Xiao et al. show that DNA damage-activated mitotic arrest and CDK activation lead to the phosphorylation of Vps34. This provides a signal to promote Vps34 ubiquitination and proteasomal degradation mediated by FBXL20, leading to inhibition of autophagy and receptor endocytosis. Importantly, they also find that expression of FBXL20 is regulated by p53-dependent transcription. Vacuolar protein-sorting 34 (Vps34), the catalytic subunit in the class III PtdIns3 (phosphatidylinositol 3) kinase complexes, mediates the production of PtdIns3P, a key intracellular lipid involved in regulating autophagy and receptor degradation. However, the signal transduction pathways by which extracellular signals regulate Vps34 complexes and the downstream cellular mechanisms are not well understood. Here we show that DNA damage-activated mitotic arrest and CDK activation lead to the phosphorylation of Vps34, which provides a signal to promote its ubiquitination and proteasomal degradation mediated by FBXL20 (an F-box protein) and the associated Skp1 (S-phase kinase-associated protein-1)–Cullin1 complex, leading to inhibition of autophagy and receptor endocytosis. Furthermore, we show that the expression of FBXL20 is regulated by p53-dependent transcription. Our study provides a molecular pathway by which DNA damage regulates Vps34 complexes and its downstream mechanisms, including autophagy and receptor endocytosis, through SCF (Skp1–Cul1–F-box)-mediated ubiquitination and degradation. Since the expression of FBXL20 is regulated by p53-dependent transcription, the control of Vps34 ubiquitination and proteasomal degradation by FBXL20 and the associated SCF complex expression provides a novel checkpoint for p53 to regulate autophagy and receptor degradation in DNA damage response.
Collapse
Affiliation(s)
- Juan Xiao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Tao Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Daichao Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Huibing Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yu Cai
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Taijie Jin
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Min Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Mingzhi Jin
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Kejia Wu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Junying Yuan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
35
|
Gewirtz DA. The autophagic response to radiation: relevance for radiation sensitization in cancer therapy. Radiat Res 2014; 182:363-7. [PMID: 25184372 DOI: 10.1667/rr13774.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation almost uniformly promotes autophagy in tumor cells. While radiation-induced autophagy often serves as a protective function in cell culture studies, it is currently uncertain to what extent autophagy might be induced by radiation in human malignancies; it is furthermore unknown whether autophagy induced by radiation can or should be suppressed for therapeutic benefit. Current clinical trials combining chemotherapeutic drugs or radiation therapy with chloroquine or hydroxychloroquine as autophagy inhibitors may be premature without the benefit of stratification to identify patients whose malignancies might be susceptible to autophagy inhibition as a therapeutic strategy. In addition, there are also concerns as to whether chloroquine and hydroxychloroquine, the agents currently in use, have the capacity to suppress autophagy when administered systemically at tolerable doses. Finally, any agent that actually has the appropriate pharmacokinetic profile to function as a systemic autophagy inhibitor may collaterally disrupt the homeostatic function of autophagy in normal cells.
Collapse
Affiliation(s)
- David A Gewirtz
- Department of Pharmacology, Toxicology and Medicine, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
36
|
Cytotoxic autophagy in cancer therapy. Int J Mol Sci 2014; 15:10034-51. [PMID: 24905404 PMCID: PMC4100138 DOI: 10.3390/ijms150610034] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 04/17/2014] [Accepted: 05/19/2014] [Indexed: 01/01/2023] Open
Abstract
Autophagy is a process of cellular self-digestion, whereby the cell degrades subcellular materials in order to generate energy and metabolic precursors in order to prolong survival, classically under conditions of nutrient deprivation. Autophagy can also involve the degradation of damaged or aged organelles, and misfolded or damaged proteins to eliminate these components that might otherwise be deleterious to cellular survival. Consequently, autophagy has generally been considered a prosurvival response. Many, if not most chemotherapeutic drugs and radiation also promote autophagy, which is generally considered a cytoprotective response, in that its inhibition frequently promotes apoptotic cells death. Furthermore, it has been shown that conventional chemotherapeutic drugs and radiation alone rarely induce a form of autophagy that leads to cell death. However, there are multiple examples in the literature where newer chemotherapeutic agents, drug combinations or drugs in combination with radiation promote autophagic cell death. This review will describe autophagic cell death induced in breast tumor cells, lung cancer cells as well as glioblastoma, demonstrating that it cannot be concluded that stress induced autophagy is, of necessity, cytoprotective in function.
Collapse
|
37
|
Sharma K, Goehe R, Beckta JM, Valerie K, Gewirtz DA. Autophagy and radiosensitization in cancer. EXCLI JOURNAL 2014; 13:178-91. [PMID: 26417252 PMCID: PMC4464266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 02/19/2014] [Indexed: 11/15/2022]
Abstract
Autophagy is a natural self-degradative process by which cells eliminate misfolded proteins and damaged organelles. Autophagy has been shown to have multiple functions in tumor cells that may be dependent on the tumor type and the treatment conditions. Autophagy can have a cytoprotective role and be thought of as a survival mechanism or be cytotoxic in nature and mediate cell death. Radiation, one of the primary treatments for many different types of cancer, almost uniformly promotes autophagy in tumor cells. While autophagy produced in response to radiation is often considered to be cytoprotective, radiation-induced autophagy has also been shown to mediate susceptibility to radiation. This review addresses the complexity of autophagy in response to radiation treatment in three different cancer models, specifically lung cancer, breast cancer and glioblastoma. A deeper understanding of the different roles played by autophagy in response to radiation should facilitate the development of approaches for enhancing the therapeutic utility of radiation by providing strategies for combination treatment with unique radiosensitizers as well as preventing the initiation of strategies which are likely to attenuate the effectiveness of radiation therapy.
Collapse
Affiliation(s)
- Khushboo Sharma
- Virginia Commonwealth University, Departments of Pharmacology and Toxicology
| | - Rachel Goehe
- Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, 20892
| | - Jason M. Beckta
- Virginia Commonwealth University, Department of Radiation Oncology
| | - Kristoffer Valerie
- Virginia Commonwealth University, Department of Radiation Oncology
- Massey Cancer Center
| | - David A. Gewirtz
- Virginia Commonwealth University, Departments of Pharmacology and Toxicology
- Massey Cancer Center
- Virginia Commonwealth University, Department of Medicine
| |
Collapse
|
38
|
Elbaz HA, Lee I, Antwih DA, Liu J, Hüttemann M, Zielske SP. Epicatechin stimulates mitochondrial activity and selectively sensitizes cancer cells to radiation. PLoS One 2014; 9:e88322. [PMID: 24516636 PMCID: PMC3916420 DOI: 10.1371/journal.pone.0088322] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 01/13/2014] [Indexed: 12/27/2022] Open
Abstract
Radiotherapy is the treatment of choice for solid tumors including pancreatic cancer, but the effectiveness of treatment is limited by radiation resistance. Resistance to chemotherapy or radiotherapy is associated with reduced mitochondrial respiration and drugs that stimulate mitochondrial respiration may decrease radiation resistance. The objectives of this study were to evaluate the potential of (-)-epicatechin to stimulate mitochondrial respiration in cancer cells and to selectively sensitize cancer cells to radiation. We investigated the natural compound (-)-epicatechin for effects on mitochondrial respiration and radiation resistance of pancreatic and glioblastoma cancer cells using a Clark type oxygen electrode, clonogenic survival assays, and Western blot analyses. (-)-Epicatechin stimulated mitochondrial respiration and oxygen consumption in Panc-1 cells. Human normal fibroblasts were not affected. (-)-Epicatechin sensitized Panc-1, U87, and MIA PaCa-2 cells with an average radiation enhancement factor (REF) of 1.7, 1.5, and 1.2, respectively. (-)-Epicatechin did not sensitize normal fibroblast cells to ionizing radiation with a REF of 0.9, suggesting cancer cell selectivity. (-)-Epicatechin enhanced Chk2 phosphorylation and p21 induction when combined with radiation in cancer, but not normal, cells. Taken together, (-)-epicatechin radiosensitized cancer cells, but not normal cells, and may be a promising candidate for pancreatic cancer treatment when combined with radiation.
Collapse
Affiliation(s)
- Hosam A. Elbaz
- Department of Radiation Oncology, Wayne State University, Detroit, Michigan, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
- Wayne State University and Karmanos Cancer Institute, Detroit, Michigan, United States of America
| | - Icksoo Lee
- College of Medicine, Dankook University, Cheonan-si, Chungcheongnam-do, Republic of Korea
| | - Deborah A. Antwih
- Department of Radiation Oncology, Wayne State University, Detroit, Michigan, United States of America
- Wayne State University and Karmanos Cancer Institute, Detroit, Michigan, United States of America
| | - Jenney Liu
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
- Wayne State University and Karmanos Cancer Institute, Detroit, Michigan, United States of America
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
- Cardiovascular Research Institute, Wayne State University, Detroit, Michigan, United States of America
- Wayne State University and Karmanos Cancer Institute, Detroit, Michigan, United States of America
| | - Steven P. Zielske
- Department of Radiation Oncology, Wayne State University, Detroit, Michigan, United States of America
- Wayne State University and Karmanos Cancer Institute, Detroit, Michigan, United States of America
| |
Collapse
|
39
|
Gallegos CE, Michelin S, Trasci SB, Lobos EA, Dubner D, Carosella ED. HLA-G1 increases the radiosensitivity of human tumoral cells. Cell Immunol 2014; 287:106-11. [PMID: 24487034 DOI: 10.1016/j.cellimm.2014.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 01/05/2014] [Accepted: 01/07/2014] [Indexed: 11/30/2022]
Abstract
Different molecules regulate the response of tumoral tissues to ionizing radiation. The objective of this work was to determine if HLA-G1 expression modulates the radiosensitivity of human tumoral cell lines. To this end, human melanoma M8 and human erythroleukemia K562 cell lines, with their correspondent HLA-G1 negative and positive variants, were gamma irradiated and the survival frequency was determined by clonogenic assay. The survival fraction of HLA-G1 expressing cells was around 60% of HLA-G1 negative cells. The generation of acidic vesicular organelles was higher in HLA-G1 positive cells. Apoptosis levels showed statistically significant differences only in K562 cells, whereas the variation in G2/M cycle progression was only significant in M8 cells. In addition, irradiation diminished cell-surface HLA-G1 and increased soluble HLA-G1 levels. Soluble HLA-G1 has no influence on cell survival in any cell line. In summary, we could demonstrate that HLA-G1 confers higher radiosensitivity to HLA-G1 expressing cells.
Collapse
Affiliation(s)
- Cristina E Gallegos
- Radiopathology Laboratory, Autoridad Regulatoria Nuclear (ARN), Buenos Aires, Argentina; Toxicology Laboratory, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Severino Michelin
- Radiopathology Laboratory, Autoridad Regulatoria Nuclear (ARN), Buenos Aires, Argentina.
| | - Sofía Baffa Trasci
- Radiopathology Laboratory, Autoridad Regulatoria Nuclear (ARN), Buenos Aires, Argentina
| | | | - Diana Dubner
- Radiopathology Laboratory, Autoridad Regulatoria Nuclear (ARN), Buenos Aires, Argentina
| | - Edgardo D Carosella
- Commissariat a l'Energie Atomique et aux Energies Alternatives (CEA), Institute of Emerging Diseases and Innovative Therapies (iMETI), Research Division in Hematology and Immunology (SRHI), Paris, France; University Paris Diderot, Sorbonne Paris Cité, UMR E-5 Institut Universitaire d'Hematologie, Saint-Louis Hospital, Paris, France
| |
Collapse
|
40
|
Tchelebi L, Ashamalla H, Graves PR. Mutant p53 and the response to chemotherapy and radiation. Subcell Biochem 2014; 85:133-59. [PMID: 25201193 DOI: 10.1007/978-94-017-9211-0_8] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In addition to playing roles in the genesis and progression of cancer, mutant p53 also appears to play a significant role in the response to cancer therapy. In response to chemotherapy and radiation, two mainstays of cancer treatment, most cancer cells harboring p53 mutations show a reduced sensitivity compared to cells lacking p53 or those with wild type p53. However, there are also many instances where mutant p53 has shown no effect or enhances cellular sensitivity to chemotherapy and radiation. Similar to the in vitro cellular studies, the majority of clinical studies show a correlation between the presence of mutant p53 in patient tumors and adverse outcomes following treatment with chemotherapy agents or radiation in comparison to tumors with wild-type p53. However, it still remains unclear whether the presence of mutant p53 in tumors can serve as a reliable prognostic factor and aid in treatment planning. Thus, as genomic analysis of patient tumors becomes more cost effective, the role of mutant p53 in tumor responses from cancer therapy ultimately needs to be addressed. This chapter will discuss current mechanisms of how p53 mutations affect cellular responses to chemotherapy and radiation and discuss patient outcomes based on p53 status.
Collapse
Affiliation(s)
- Leila Tchelebi
- Department of Radiation Oncology, New York Methodist Hospital, 506 6th Street, Brooklyn, NY, 11215, USA
| | | | | |
Collapse
|