1
|
Sato T, Oshi M, Huang JL, Chida K, Roy AM, Endo I, Takabe K. CD133 expression is associated with less DNA repair, better response to chemotherapy and survival in ER-positive/HER2-negative breast cancer. Breast Cancer Res Treat 2024; 208:415-427. [PMID: 39017815 DOI: 10.1007/s10549-024-07434-3] [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: 03/22/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE CD133, a cancer stem cells (CSC) marker, has been reported to be associated with treatment resistance and worse survival in triple-negative breast cancer (BC). However, the clinical relevance of CD133 expression in ER-positive/HER2-negative (ER + /HER2-) BC, the most abundant subtype, remains unknown. METHODS The BC cohorts from the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC, n = 1904) and The Cancer Genome Atlas (TCGA, n = 1065) were used to obtain biological variables and gene expression data. RESULTS Epithelial cells were the exclusive source of CD133 gene expression in a bulk BC. CD133-high ER + /HER2- BC was associated with CD24, NOTCH1, DLL1, and ALDH1A1 gene expressions, as well as with WNT/β-Catenin, Hedgehog, and Notch signaling pathways, all characteristic for CSC. Consistent with a CSC phenotype, CD133-low BC was enriched with gene sets related to cell proliferation, such as G2M Checkpoint, MYC Targets V1, E2F Targets, and Ki67 gene expression. CD133-low BC was also linked with enrichment of genes related to DNA repair, such as BRCA1, E2F1, E2F4, CDK1/2. On the other hand, CD133-high tumors had proinflammatory microenvironment, higher activity of immune cells, and higher expression of genes related to inflammation and immune response. Finally, CD133-high tumors had better pathological complete response after neoadjuvant chemotherapy in GSE25066 cohort and better disease-free survival and overall survival in both TCGA and METABRIC cohorts. CONCLUSION CD133-high ER + /HER2- BC was associated with CSC phenotype such as less cell proliferation and DNA repair, but also with enhanced inflammation, better response to neoadjuvant chemotherapy and better prognosis.
Collapse
Affiliation(s)
- Takumi Sato
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
- University of Tokyo Hospital, Tokyo, 113-8655, Japan
- National Hospital Organization Disaster Medical Center, Tokyo, 190-0014, Japan
| | - Masanori Oshi
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Jing Li Huang
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
| | - Kohei Chida
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA
- Department of Gastroenterological Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8562, Japan
| | - Arya Mariam Roy
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY, 14263, USA.
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, 14263, USA.
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8520, Japan.
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan.
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, 160-8402, Japan.
| |
Collapse
|
2
|
Leong VWS, Khan S, Sharma P, Wu S, Thomas RR, Li X, Singh SK, Lang FF, Yung AWK, Koul D. MGMT function determines the differential response of ATR inhibitors with DNA-damaging agents in glioma stem cells for GBM therapy. Neurooncol Adv 2024; 6:vdad165. [PMID: 38213834 PMCID: PMC10783493 DOI: 10.1093/noajnl/vdad165] [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: 01/13/2024] Open
Abstract
Background The most prevalent cancer treatments cause cell death through DNA damage. However, DNA damage response (DDR) repair pathways, initiated by tumor cells, can withstand the effects of anticancer drugs, providing justification for combining DDR inhibitors with DNA-damaging anticancer treatments. Methods Cell viability assays were performed with CellTiter-Glo assay. DNA damage was evaluated using Western blotting analysis. RNA-seq and single-cell level expression were used to identify the DDR signatures. In vivo, studies were conducted in mice to determine the effect of ATris on TMZ sensitization. Results We found a subpopulation of glioma sphere-forming cells (GSCs) with substantial synergism with temozolomide (TMZ) using a panel of 3 clinical-grade ataxia-telangiectasia- and Rad3-related kinase inhibitors (ATRis), (elimusertib, berzosertib, and ceralasertib). Interestingly, most synergistic cell lines had O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation, indicating that ATRi mainly benefits tumors with no MGMT repair. Further, TMZ activated the ATR-checkpoint kinase 1 (Chk1) axis in an MGMT-dependent way. TMZ caused ATR-dependent Chk1 phosphorylation and DNA double-strand breaks as shown by increased γH2AX. Increased DNA damage and decreased Chk1 phosphorylation were observed upon the addition of ATRis to TMZ in MGMT-methylated (MGMT-) GSCs. TMZ also improved sensitivity to ATRis in vivo, as shown by increased mouse survival with the TMZ and ATRi combination treatment. Conclusions This research provides a rationale for selectively targeting MGMT-methylated cells using ATRis and TMZ combination. Overall, we believe that MGMT methylation status in GBM could serve as a robust biomarker for patient selection for ATRi combined with TMZ.
Collapse
Affiliation(s)
- Vincent W S Leong
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sabbir Khan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Pratibha Sharma
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shaofang Wu
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Riya R Thomas
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xiaolong Li
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sanjay K Singh
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alfred W K Yung
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dimpy Koul
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
3
|
Zhang Y, Gu W, Shao Y. The therapeutic targets of N6-methyladenosine (m6A) modifications on tumor radioresistance. Discov Oncol 2023; 14:141. [PMID: 37522921 PMCID: PMC10390431 DOI: 10.1007/s12672-023-00759-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023] Open
Abstract
Radiation therapy is an important tool for malignant tumors, and its tolerance needs to be addressed. In recent years, several studies have shown that regulators of aberrant m6A methylation play an important role in the formation, development and invasion and metastasis of tumors. A large number of studies have confirmed aberrant m6A methylation as a new target for tumour therapy, but research on whether it can play a role in tumor sensitivity to radiotherapy has not been extensive and thorough enough. Recent studies have shown that all three major enzymes of m6A methylation have significant roles in radioresistance, and that the enzymes that play a role differ in different tumor types and by different mechanisms, including regulating tumor cell stemness, affecting DNA damage and repair, and controlling the cell cycle. Therefore, elucidating the mechanisms of m6A methylation in the radiotherapy of malignant tumors is essential to counteract radioresistance, improve the efficacy of radiotherapy, and even propose targeted treatment plans for specific tumors. The latest research progress on m6A methylation and radioresistance is reviewed in this article.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou, 213003, China
| | - Wendong Gu
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou, 213003, China.
| | - Yingjie Shao
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Changzhou, 213003, China.
| |
Collapse
|
4
|
Alejo S, Palacios B, Venkata PP, He Y, Li W, Johnson J, Chen Y, Jayamohan S, Pratap U, Clarke K, Zou Y, Lv Y, Weldon K, Viswanadhapalli S, Lai Z, Ye Z, Chen Y, Gilbert A, Suzuki T, Tekmal R, Zhao W, Zheng S, Vadlamudi R, Brenner A, Sareddy GR. Lysine-specific histone demethylase 1A (KDM1A/LSD1) inhibition attenuates DNA double-strand break repair and augments the efficacy of temozolomide in glioblastoma. Neuro Oncol 2023; 25:1249-1261. [PMID: 36652263 PMCID: PMC10326496 DOI: 10.1093/neuonc/noad018] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Efficient DNA repair in response to standard chemo and radiation therapies often contributes to glioblastoma (GBM) therapy resistance. Understanding the mechanisms of therapy resistance and identifying the drugs that enhance the therapeutic efficacy of standard therapies may extend the survival of GBM patients. In this study, we investigated the role of KDM1A/LSD1 in DNA double-strand break (DSB) repair and a combination of KDM1A inhibitor and temozolomide (TMZ) in vitro and in vivo using patient-derived glioma stem cells (GSCs). METHODS Brain bioavailability of the KDM1A inhibitor (NCD38) was established using LS-MS/MS. The effect of a combination of KDM1A knockdown or inhibition with TMZ was studied using cell viability and self-renewal assays. Mechanistic studies were conducted using CUT&Tag-seq, RNA-seq, RT-qPCR, western blot, homologous recombination (HR) and non-homologous end joining (NHEJ) reporter, immunofluorescence, and comet assays. Orthotopic murine models were used to study efficacy in vivo. RESULTS TCGA analysis showed KDM1A is highly expressed in TMZ-treated GBM patients. Knockdown or knockout or inhibition of KDM1A enhanced TMZ efficacy in reducing the viability and self-renewal of GSCs. Pharmacokinetic studies established that NCD38 readily crosses the blood-brain barrier. CUT&Tag-seq studies showed that KDM1A is enriched at the promoters of DNA repair genes and RNA-seq studies confirmed that KDM1A inhibition reduced their expression. Knockdown or inhibition of KDM1A attenuated HR and NHEJ-mediated DNA repair capacity and enhanced TMZ-mediated DNA damage. A combination of KDM1A knockdown or inhibition and TMZ treatment significantly enhanced the survival of tumor-bearing mice. CONCLUSIONS Our results provide evidence that KDM1A inhibition sensitizes GBM to TMZ via attenuation of DNA DSB repair pathways.
Collapse
Affiliation(s)
- Salvador Alejo
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Bridgitte E Palacios
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Prabhakar Pitta Venkata
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Yi He
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Wenjing Li
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Jessica D Johnson
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Yihong Chen
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P. R. China
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Sridharan Jayamohan
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Uday P Pratap
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Kyra Clarke
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Yi Zou
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Yingli Lv
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Korri Weldon
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Suryavathi Viswanadhapalli
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Zhao Lai
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Zhenqing Ye
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Yidong Chen
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P. R. China
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Andrea R Gilbert
- Department of Pathology and Laboratory Medicine, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Takayoshi Suzuki
- The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan
| | - Rajeshwar R Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Weixing Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Siyuan Zheng
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Audie L. Murphy South Texas Veterans Health Care System, San Antonio, Texas, 78229, USA
| | - Andrew J Brenner
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Department of Hematology & Oncology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| | - Gangadhara R Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas, 78229, USA
| |
Collapse
|
5
|
Li J, Song C, Gu J, Li C, Zang W, Shi L, Chen L, Zhu L, Zhou M, Wang T, Li H, Qi S, Lu Y. RBBP4 regulates the expression of the Mre11-Rad50-NBS1 (MRN) complex and promotes DNA double-strand break repair to mediate glioblastoma chemoradiotherapy resistance. Cancer Lett 2023; 557:216078. [PMID: 36736531 DOI: 10.1016/j.canlet.2023.216078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/27/2022] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
For treatment of glioblastoma (GBM), temozolomide (TMZ) and radiotherapy (RT) exert antitumor effects by inducing DNA double-strand breaks (DSBs), mainly via futile DNA mismatch repair (MMR) and inducing apoptosis. Here, we provide evidence that RBBP4 modulates glioblastoma resistance to chemotherapy and radiotherapy by recruiting transcription factors and epigenetic regulators that bind to their promoters to regulate the expression of the Mre11-Rad50-NBS1(MRN) complex and the level of DNA-DSB repair, which are closely associated with recovery from TMZ- and radiotherapy-induced DNA damage in U87MG and LN229 glioblastoma cells, which have negative MGMT expression. Disruption of RBBP4 induced GBM cell DNA damage and apoptosis in response to TMZ and radiotherapy and enhanced radiotherapy and chemotherapy sensitivity by the independent pathway of MGMT. These results displayed a possible chemo-radioresistant mechanism in MGMT negative GBM. In addition, the RBBP4-MRN complex regulation axis may provide an interesting target for developing therapy-sensitizing strategies for GBM.
Collapse
Affiliation(s)
- Junjie Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China; Nanfang Neurology Research Institution, Nanfang Hospital, Southern Medical University, Guangzhou, China; Nanfang Glioma Center, Guangzhou, China
| | - Chong Song
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Neurosurgery, The Central Hospital of Dalian University of Technology, Dalian, China
| | - Junwei Gu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China; The First People's Hospital of Xiushui County, Jiujiang, Jiangxi Province, China
| | - Chiyang Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenrui Zang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Linyong Shi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Chen
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liwen Zhu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Min Zhou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tong Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China; Nanfang Neurology Research Institution, Nanfang Hospital, Southern Medical University, Guangzhou, China; Nanfang Glioma Center, Guangzhou, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China; Nanfang Neurology Research Institution, Nanfang Hospital, Southern Medical University, Guangzhou, China; Nanfang Glioma Center, Guangzhou, China
| | - Yuntao Lu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China; Nanfang Neurology Research Institution, Nanfang Hospital, Southern Medical University, Guangzhou, China; Nanfang Glioma Center, Guangzhou, China.
| |
Collapse
|
6
|
Pavlova G, Belyashova A, Savchenko E, Panteleev D, Shamadykova D, Nikolaeva A, Pavlova S, Revishchin A, Golbin D, Potapov A, Antipina N, Golanov A. Reparative properties of human glioblastoma cells after single exposure to a wide range of X-ray doses. Front Oncol 2022; 12:912741. [PMID: 35992802 PMCID: PMC9386365 DOI: 10.3389/fonc.2022.912741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Radiation therapy induces double-stranded DNA breaks in tumor cells, which leads to their death. A fraction of glioblastoma cells repair such breaks and reinitiate tumor growth. It was necessary to identify the relationship between high radiation doses and the proliferative activity of glioblastoma cells, and to evaluate the contribution of DNA repair pathways, homologous recombination (HR), and nonhomologous end joining (NHEJ) to tumor-cell recovery. We demonstrated that the GO1 culture derived from glioblastoma cells from Patient G, who had previously been irradiated, proved to be less sensitive to radiation than the Sus\fP2 glioblastoma culture was from Patient S, who had not been exposed to radiation before. GO1 cell proliferation decreased with radiation dose, and MTT decreased to 35% after a single exposure to 125 Gγ. The proliferative potential of glioblastoma culture Sus\fP2 decreased to 35% after exposure to 5 Gγ. At low radiation doses, cell proliferation and the expression of RAD51 were decreased; at high doses, cell proliferation was correlated with Ku70 protein expression. Therefore, HR and NHEJ are involved in DNA break repair after exposure to different radiation doses. Low doses induce HR, while higher doses induce the faster but less accurate NHEJ pathway of double-stranded DNA break repair.
Collapse
Affiliation(s)
- Galina Pavlova
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
- Department of Medical Genetics, Sechenov First Moscow State Medical University, Moscow, Russia
- *Correspondence: Galina Pavlova,
| | - Alexandra Belyashova
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| | - Ekaterina Savchenko
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| | - Dmitri Panteleev
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| | - Dzhirgala Shamadykova
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| | - Anna Nikolaeva
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| | - Svetlana Pavlova
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| | - Alexander Revishchin
- Laboratory of Neurogenetics and Genetics Development, Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| | - Denis Golbin
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| | - Alexander Potapov
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| | - Natalia Antipina
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| | - Andrey Golanov
- Nikolay Nilovich (N.N.) Burdenko National Medical Research Center of Neurosurgery (NMRCN), Moscow, Russia
| |
Collapse
|
7
|
Maksoud S. The DNA Double-Strand Break Repair in Glioma: Molecular Players and Therapeutic Strategies. Mol Neurobiol 2022; 59:5326-5365. [PMID: 35696013 DOI: 10.1007/s12035-022-02915-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 06/05/2022] [Indexed: 12/12/2022]
Abstract
Gliomas are the most frequent type of tumor in the central nervous system, which exhibit properties that make their treatment difficult, such as cellular infiltration, heterogeneity, and the presence of stem-like cells responsible for tumor recurrence. The response of this type of tumor to chemoradiotherapy is poor, possibly due to a higher repair activity of the genetic material, among other causes. The DNA double-strand breaks are an important type of lesion to the genetic material, which have the potential to trigger processes of cell death or cause gene aberrations that could promote tumorigenesis. This review describes how the different cellular elements regulate the formation of DNA double-strand breaks and their repair in gliomas, discussing the therapeutic potential of the induction of this type of lesion and the suppression of its repair as a control mechanism of brain tumorigenesis.
Collapse
Affiliation(s)
- Semer Maksoud
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
8
|
Dong Y, Xiong Y, Zhou D, Yao M, Wang X, Bi W, Zhang J. TRIM56 Reduces Radiosensitization of Human Glioblastoma by Regulating FOXM1-Mediated DNA Repair. Mol Neurobiol 2022; 59:5312-5325. [PMID: 35696011 DOI: 10.1007/s12035-022-02898-0] [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: 12/29/2021] [Accepted: 05/21/2022] [Indexed: 12/01/2022]
Abstract
Recurrent glioblastoma is characterized by resistance to radiotherapy or chemotherapy. In this study, we investigated the role of TRIM56 in radiosensitization and its potential underlying molecular mechanism. TRIM56 expression levels were measured in glioblastoma tissues and cell lines by immunohistochemical staining, western blot, and qRT-PCR. MTT assay, colony formation assay, and TUNEL assay were used to investigate the effect of TRIM56 on cell viability, cell proliferation, and cell apoptosis. Co-immunoprecipitation was used to clarify the interaction between TRIM56 and FOXM1. Finally, tumor xenograft experiments were performed to analyze the effect of TRIM56 on tumor growth in vivo. The expression of TRIM56 was significantly increased in glioblastoma tissues and cell lines and its expression was associated with poor prognosis of patients with glioblastoma. Moreover, TRIM56 reduced the radiosensitivity of glioblastoma cells and promoted DNA repairment. Mechanistically, TRIM56 promoted FOXM1 protein level, enhanced the stability of FOXM1 by de-ubiquitination, and promoted DNA damage repair through FOXM1 in glioblastoma cells. TRIM56 could reduce the radiosensitivity of glioblastoma in vivo. TRIM56 may suppress the radiosensitization of human glioblastoma by regulating FOXM1-mediated DNA repair. Targeting the TRIM56 may be an effective method to reverse radiotherapy-resistant in glioblastoma recurrent.
Collapse
Affiliation(s)
- Yun Dong
- School of Pharmacy and Food Sciences, Zhuhai College of Science and Technology, Zhuhai, 519040, Guangdong Province, China.,School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Nanshan District, No.1066, Xueyuan Road, Shenzhen City, 518055, Guangdong Province, China
| | - Yiping Xiong
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Nanshan District, No.1066, Xueyuan Road, Shenzhen City, 518055, Guangdong Province, China
| | - Duanyang Zhou
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Nanshan District, No.1066, Xueyuan Road, Shenzhen City, 518055, Guangdong Province, China
| | - Min Yao
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Nanshan District, No.1066, Xueyuan Road, Shenzhen City, 518055, Guangdong Province, China
| | - Xiao Wang
- Department of Pharmacy, Shenzhen People's Hospital, Shenzhen City, 815020, Guangdong Province, China
| | - Wenchuan Bi
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Nanshan District, No.1066, Xueyuan Road, Shenzhen City, 518055, Guangdong Province, China.
| | - Jian Zhang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Nanshan District, No.1066, Xueyuan Road, Shenzhen City, 518055, Guangdong Province, China.
| |
Collapse
|
9
|
Nageeb AM, Mohamed MM, Ezz El Arab LR, Khalifa MK, Swellam M. Next generation sequencing of BRCA genes in glioblastoma multiform Egyptian patients: a pilot study. Arch Physiol Biochem 2022; 128:809-817. [PMID: 32100578 DOI: 10.1080/13813455.2020.1729814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Germ line mutations of BRCA1 and BRCA2 were correlated with a variety of cancer Authors aimed to use next-generation sequencing (NGS) to detect BRCA1 and BRCA2 germ line mutations in glioblastoma multiform (GBM) Egyptian patients. MATERIALS AND METHODS Genomic DNA was extracted from six GBM cases, amplified using Ion AmpliSeq BRCA1 and BRCA2 panel. DNA libraries were pooled, barcoded and finally sequenced using Ion Torrent Personal Genome Machine sequencer. RESULTS BRCA1 the previously reported rs1799966, rs1799950, rs16941 were found in five cases and they are in a linkage disequilibrium forming two distinct haplotypes, which might support their role in cancer predisposition. Out of the 18 reported variants in BRCA2, three denovo mutations were detected which leads to frame shift. CONCLUSION Further studies on large number of GBM patients and control cases to determine BRCA1 and BRCA2 germline mutations and haplotypes; diagnostic and prognostic role are encouraged.
Collapse
Affiliation(s)
- Amira M Nageeb
- Biochemistry Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Dokki, Giza, Egypt
- High Throughput Molecular and Genetic Laboratory, Center for Excellences for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| | - Magdy M Mohamed
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Lobna R Ezz El Arab
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Menha Swellam
- Biochemistry Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Dokki, Giza, Egypt
- High Throughput Molecular and Genetic Laboratory, Center for Excellences for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
10
|
Maleki Dana P, Sadoughi F, Mirzaei H, Asemi Z, Yousefi B. DNA damage response and repair in the development and treatment of brain tumors. Eur J Pharmacol 2022; 924:174957. [DOI: 10.1016/j.ejphar.2022.174957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 04/03/2022] [Accepted: 04/11/2022] [Indexed: 11/03/2022]
|
11
|
ÇAĞLAR HO. Identification of Genes Related to DNA Repair Mechanism in Glioblastoma by Bioinformatics Methods. KOCAELI ÜNIVERSITESI SAĞLIK BILIMLERI DERGISI 2022. [DOI: 10.30934/kusbed.1003777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Objective: Aberrant expression of genes involved in DNA repair mechanisms (DRM) have been associated with radiation sensitivity of glioblastoma (GBM) cells. Identification of genes in DRM through bioinformatics methods may help identify potential novel therapeutic targets that can be used in GBM treatment. This study aims to identify genes that play a role in DRM in GBM using bioinformatics methods.
Methods: Genes associated with DRM were identified using the “Reactome” and “KEGG” databases. The mRNA expression profiles of DRM related genes were analyzed in the GEO GDS1813 and GDS2853 datasets including GBM tumor samples using the "Orange Canvas" software. Genetic changes of genes were identified in GBM TCGA cases using the cBioPortal database. The GEPIA2 was used to show the effect of altered expression profiles of these genes on patient survival.
Results: The mRNA expression profiles of ERCC6, FAN1, MBD4, PARP1 and UNG genes were found to be altered in GBM tumors. Mutations and copy number alterations for the identified genes were observed in TCGA GBM cases. The overall survival and disease-free survival of TCGA GBM patients were not significantly different between high and low expression groups.
Conclusion: ERCC6, PARP1 and UNG genes identified in the current study may be potential therapeutic targets that can increase the efficacy of radiotherapy in GBM in case of their suppression.
Collapse
|
12
|
Morrison C, Weterings E, Mahadevan D, Sanan A, Weinand M, Stea B. Expression Levels of RAD51 Inversely Correlate with Survival of Glioblastoma Patients. Cancers (Basel) 2021; 13:cancers13215358. [PMID: 34771522 PMCID: PMC8582387 DOI: 10.3390/cancers13215358] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/17/2021] [Accepted: 10/23/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Identifying prognostic and predictive biomarkers for glioblastoma (GBM), a primary brain tumor, is essential in improving patient survival. We utilized gene expression profiling to investigate a uniform population of GBM patients who had been treated with surgery and adjuvant radiation therapy versus normal brain tissue, and identified high RAD51 expression as a poor prognostic marker that is amenable to therapeutic intervention. This observation was confirmed utilizing a publicly available gene expression dataset in a cohort of GBM patients. Abstract Treatment failures of glioblastoma (GBM) occur within high-dose radiation fields. We hypothesized that this is due to increased capacity for DNA damage repair in GBM. We identified 24 adult GBM patients treated with maximal safe resection followed by radiation with concurrent and adjuvant temozolomide. The mRNA from patients was quantified using NanoString Technologies’ nCounter platform and compared with 12 non-neoplastic temporal lobe tissue samples as a control. Differential expression analysis identified seven DNA repair genes significantly upregulated in GBM tissues relative to controls (>4-fold difference, adjusted p values < 0.001). Among these seven genes, Cox proportional hazards models identified RAD51 to be associated with an increased risk of death (HR = 3.49; p = 0.03). Kaplan–Meier (KM) analysis showed that patients with high RAD51 expression had significantly shorter OS compared to low levels (median OS of 10.6 mo. vs 20.1 mo.; log-rank p = 0.03). Our findings were validated in a larger external dataset of 162 patients using publicly available gene expression data quantified by the same NanoString technology (median OS of 13.8 mo. vs. 17.4 mo; log-rank p = 0.006). Within this uniformly treated GBM population, RAD51, in the homologous recombination pathway, was overexpressed (vs. normal brain) and inversely correlated with OS. High RAD51 expression may be a prognostic biomarker and a therapeutic target in GBM.
Collapse
Affiliation(s)
- Christopher Morrison
- Department of Radiation Oncology, University of Arizona, Tucson, AZ 85719, USA; (C.M.); (E.W.)
| | - Eric Weterings
- Department of Radiation Oncology, University of Arizona, Tucson, AZ 85719, USA; (C.M.); (E.W.)
| | - Daruka Mahadevan
- Mays Cancer Center, University of Texas Health, San Antonio, TX 78229, USA;
| | - Abhay Sanan
- Center for Neurosciences, Tucson, AZ 85718, USA;
| | - Martin Weinand
- Department of Neurosurgery, University of Arizona College of Medicine, Tucson, AZ 85724, USA;
| | - Baldassarre Stea
- Department of Radiation Oncology, University of Arizona, Tucson, AZ 85719, USA; (C.M.); (E.W.)
- Correspondence: ; Tel.: +1-(520)-694-0861
| |
Collapse
|
13
|
The CD44high Subpopulation of Multifraction Irradiation-Surviving NSCLC Cells Exhibits Partial EMT-Program Activation and DNA Damage Response Depending on Their p53 Status. Int J Mol Sci 2021; 22:ijms22052369. [PMID: 33673439 PMCID: PMC7956695 DOI: 10.3390/ijms22052369] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/15/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Ionizing radiation (IR) is used for patients diagnosed with unresectable non-small cell lung cancer (NSCLC). However, radiotherapy remains largely palliative due to the survival of specific cell subpopulations. In the present study, the sublines of NSCLC cells, A549IR (p53wt) and H1299IR (p53null) survived multifraction X-ray radiation exposure (MFR) at a total dose of 60 Gy were investigated three weeks after the MFR course. We compared radiosensitivity (colony formation), expression of epithelial-mesenchymal transition (EMT) markers, migration activity, autophagy, and HR-dependent DNA double-strand break (DSB) repair in the bulk and entire CD44high/CD166high CSC-like populations of both parental and MFR survived NSCLC cells. We demonstrated that the p53 status affected: the pattern of expression of N-cadherin, E-cadherin, Vimentin, witnessing the appearance of EMT-like phenotype of MFR-surviving sublines; 1D confined migratory behavior (wound healing); the capability of an irradiated cell to continue to divide and form a colony of NSCLC cells before and after MFR; influencing the CD44/CD166 expression level in MFR-surviving NSCLC cells after additional single irradiation. Our data further emphasize the impact of p53 status on the decay of γH2AX foci and the associated efficacy of the DSB repair in NSCLC cells survived after MFR. We revealed that Rad51 protein might play a principal role in MFR-surviving of p53 null NSCLC cells promoting DNA DSB repair by homologous recombination (HR) pathway. The proportion of Rad51 + cells elevated in CD44high/CD166high population in MFR-surviving p53wt and p53null sublines and their parental cells. The p53wt ensures DNA-PK-mediated DSB repair for both parental and MFR-surviving cells irrespectively of a subsequent additional single irradiation. Whereas in the absence of p53, a dose-dependent increase of DNA-PK-mediated non-homologous end joining (NHEJ) occurred as an early post-irradiation response is more intensive in the CSC-like population MFR-surviving H1299IR, compared to their parental H1299 cells. Our study strictly observed a significantly higher content of LC3 + cells in the CD44high/CD166high populations of p53wt MFR-surviving cells, which enriched the CSC-like cells in contrast to their p53null counterparts. The additional 2 Gy and 5 Gy X-ray exposure leads to the dose-dependent increase in the proportion of LC3 + cells in CD44high/CD166high population of both parental p53wt and p53null, but not MFR-surviving NSCLC sublines. Our data indicated that autophagy is not necessarily associated with CSC-like cells’ radiosensitivity, emphasizing that careful assessment of other milestone processes (such as senescence and autophagy-p53-Zeb1 axis) of primary radiation responses may provide new potential targets modulated for therapeutic benefit through radiosensitizing cancer cells while rescuing normal tissue. Our findings also shed light on the intricate crosstalk between autophagy and the p53-related EMT, by which MFR-surviving cells might obtain an invasive phenotype and metastatic potential.
Collapse
|
14
|
Kowalski-Chauvel A, Lacore MG, Arnauduc F, Delmas C, Toulas C, Cohen-Jonathan-Moyal E, Seva C. The m6A RNA Demethylase ALKBH5 Promotes Radioresistance and Invasion Capability of Glioma Stem Cells. Cancers (Basel) 2020; 13:cancers13010040. [PMID: 33375621 PMCID: PMC7795604 DOI: 10.3390/cancers13010040] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Glioblastoma stem cells (GBMSCs), which are particularly radio-resistant and invasive, are responsible for the high recurrence of glioblastoma (GBM). Therefore, there is a real need for a better understanding of the mechanisms involved in these processes and to identify new factors that might be targeted to radiosensitize GBMSC and decrease their invasive capability. Here, we report that the m6A RNA demethylase ALKBH5, which is overexpressed in GBMSCs, promotes their radioresistance by controlling the homologous repair. ALKBH5 was also involved in GBMSC invasion. These data suggest that ALKBH5 inhibition might be a novel approach to radiosensitize GBMSCs and to overcome their invasiveness. Abstract Recurrence of GBM is thought to be due to GBMSCs, which are particularly chemo-radioresistant and characterized by a high capacity to invade normal brain. Evidence is emerging that modulation of m6A RNA methylation plays an important role in tumor progression. However, the impact of this mRNA modification in GBM is poorly studied. We used patient-derived GBMSCs to demonstrate that high expression of the RNA demethylase, ALKBH5, increases radioresistance by regulating homologous recombination (HR). In cells downregulated for ALKBH5, we observed a decrease in GBMSC survival after irradiation likely due to a defect in DNA-damage repair. Indeed, we observed a decrease in the expression of several genes involved in the HR, including CHK1 and RAD51, as well as a persistence of γ-H2AX staining after IR. We also demonstrated in this study that ALKBH5 contributes to the aggressiveness of GBM by favoring the invasion of GBMSCs. Indeed, GBMSCs deficient for ALKBH5 exhibited a significant reduced invasion capability relative to control cells. Our data suggest that ALKBH5 is an attractive therapeutic target to overcome radioresistance and invasiveness of GBMSCs.
Collapse
Affiliation(s)
- Aline Kowalski-Chauvel
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
| | - Marie Géraldine Lacore
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
| | - Florent Arnauduc
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
| | - Caroline Delmas
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
- IUCT-Oncopole Toulouse, 31000 Tolouse, France
| | - Christine Toulas
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
- IUCT-Oncopole Toulouse, 31000 Tolouse, France
| | - Elizabeth Cohen-Jonathan-Moyal
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
- IUCT-Oncopole Toulouse, 31000 Tolouse, France
| | - Catherine Seva
- INSERM UMR.1037-Cancer Research Center of Toulouse (CRCT)/University Paul Sabatier Toulouse III, 31100 Toulouse, France; (A.K.-C.); (M.G.L.); (F.A.); (C.D.); (C.T.); (E.C.-J.-M.)
- Correspondence: ; Tel.: +33-(5)82741604
| |
Collapse
|
15
|
Chien JCY, Tabet E, Pinkham K, da Hora CC, Chang JCY, Lin S, Badr CE, Lai CPK. A multiplexed bioluminescent reporter for sensitive and non-invasive tracking of DNA double strand break repair dynamics in vitro and in vivo. Nucleic Acids Res 2020; 48:e100. [PMID: 32797168 PMCID: PMC7515717 DOI: 10.1093/nar/gkaa669] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/29/2020] [Accepted: 07/31/2020] [Indexed: 12/31/2022] Open
Abstract
Tracking DNA double strand break (DSB) repair is paramount for the understanding and therapeutic development of various diseases including cancers. Herein, we describe a multiplexed bioluminescent repair reporter (BLRR) for non-invasive monitoring of DSB repair pathways in living cells and animals. The BLRR approach employs secreted Gaussia and Vargula luciferases to simultaneously detect homology-directed repair (HDR) and non-homologous end joining (NHEJ), respectively. BLRR data are consistent with next-generation sequencing results for reporting HDR (R2 = 0.9722) and NHEJ (R2 = 0.919) events. Moreover, BLRR analysis allows longitudinal tracking of HDR and NHEJ activities in cells, and enables detection of DSB repairs in xenografted tumours in vivo. Using the BLRR system, we observed a significant difference in the efficiency of CRISPR/Cas9-mediated editing with guide RNAs only 1-10 bp apart. Moreover, BLRR analysis detected altered dynamics for DSB repair induced by small-molecule modulators. Finally, we discovered HDR-suppressing functions of anticancer cardiac glycosides in human glioblastomas and glioma cancer stem-like cells via inhibition of DNA repair protein RAD51 homolog 1 (RAD51). The BLRR method provides a highly sensitive platform to simultaneously and longitudinally track HDR and NHEJ dynamics that is sufficiently versatile for elucidating the physiology and therapeutic development of DSB repair.
Collapse
Affiliation(s)
| | - Elie Tabet
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA.,Department of Biomedical Engineering, University of South Dakota, 4800 N. Career Ave, Suite 221, Sioux Falls, Vermillion, SD 57069, USA
| | - Kelsey Pinkham
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Cintia Carla da Hora
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA.,Neuroscience Program, Harvard Medical School, Boston, MA 02115, USA
| | - Jason Cheng-Yu Chang
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan.,Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Steven Lin
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan.,Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Christian E Badr
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA.,Neuroscience Program, Harvard Medical School, Boston, MA 02115, USA
| | - Charles Pin-Kuang Lai
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 10617, Taiwan.,Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan.,Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
16
|
Das PK, Zahan T, Abdur Rakib M, Khanam JA, Pillai S, Islam F. Natural Compounds Targeting Cancer Stem Cells: A Promising Resource for Chemotherapy. Anticancer Agents Med Chem 2020; 19:1796-1808. [PMID: 31272363 DOI: 10.2174/1871520619666190704111714] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/04/2019] [Accepted: 05/20/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cancer Stem Cells (CSCs) are the subpopulation of cancer cells which are directly involved in drug resistance, metastases to distant organ and cancer recurrence. METHODS A systematic literature search was conducted through various electronic databases including, Pubmed, Scopus, Google scholar using the keywords "cancer stem cells" and "natural compounds" in the present study. Articles published between 1999 and 2019 were reviewed. All the expositions concerning CSCs associated cancer pathogenesis and therapy resistance, as well as targeting these properties of CSCs by natural compounds were selected for the current study. RESULTS Natural compounds have always been thought as a rich source of biologically active principles, which target aberrantly activated signaling pathways and other modalities of CSCs, while tethering painful side effects commonly involved in the first-line and second-line chemo-radiotherapies. In this review, we have described the key signaling pathways activated in CSCs to maintain their survival and highlighted how natural compounds interrupt these signaling pathways to minimize therapy resistance, pathogenesis and cancer recurrence properties of CSCs, thereby providing useful strategies to treat cancer or aid in cancer therapy improvement. Like normal stem cells, CSCs rely on different signaling pathways and other properties for their maintenance. Therefore, the success of cancer treatment depends on the development of proper anti-neoplastic drugs capable of intercepting those signaling pathways as well as other properties of CSCs in order to eradicate this evasive subpopulation of cancer cells. CONCLUSION Compounds of natural origin might act as an outstanding source to design novel therapies against cancer stem cells.
Collapse
Affiliation(s)
- Plabon K Das
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Tasnim Zahan
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Md Abdur Rakib
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Jahan A Khanam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Suja Pillai
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Farhadul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh.,School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
17
|
Hembram KC, Dash SR, Das B, Sethy C, Chatterjee S, Bindhani BK, Kundu CN. Quinacrine Based Gold Hybrid Nanoparticles Caused Apoptosis through Modulating Replication Fork in Oral Cancer Stem Cells. Mol Pharm 2020; 17:2463-2472. [PMID: 32407635 DOI: 10.1021/acs.molpharmaceut.0c00197] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The presence of cancer stem cells (CSCs) in the tumor microenvironment is responsible for the development of chemoresistance and recurrence of cancer. Our previous investigation revealed the anticancer mechanism of quinacrine-based silver and gold hybrid nanoparticles (QAgNP and QAuNP) in oral cancer cells, but to avoid cancer recurrence, it is important to study the effect of these nanoparticles (NPs) on CSCs. Here, we developed an in vitro CSCs model using SCC-9 oral cancer cells and validated via FACS analysis. Then, 40-60% of cells were found to be CD44+/CD133+ and CD24-. QAuNP showed excellent anti-CSC growth potential against SCC-9-cancer stem like cells (IC50 = 0.4 μg/mL) with the down-regulation of representative CSC markers. Prolonged exposure of QAuNP induced the S-phase arrest and caused re-replication shown by the extended G2/M population and apoptosis to SCC-9-CSC like cells. Up-regulation of BAX, PARP cleavage, and simultaneous down-regulation of Bcl-xL in prolonged treatment to CSCs suggested that the majority of the cells have undergone apoptosis. QAuNP treatment also caused a loss in DNA repair in CSCs. Mostly, the base excision repair (BER) components (Fen-1, DNA ligase-1, Pol-β, RPA, etc.) were significantly down-regulated after QAuNP treatment, which suggested its action against DNA repair machinery. The replication fork maintenance-related proteins, RAD 51 and BRCA-2, were also deregulated. Very surprisingly, depletion of WRN (an interacting partner for Pre-RC and Fen-1) and a significant increase in expression of fork-degrading nuclease MRE-11 in 96 h treated NPs were observed. Results suggest QAuNP treatment caused excessive DNA damage and re-replication mediated replication stress (RS) and stalling of the replication fork. Inhibition of BER components hinders the flap clearance activity of Fen-1, and it further caused RS and stopped DNA synthesis. Overall, QAuNP treatment led to irreparable replication fork movement, and the stalled replication fork might have degraded by MRE-11, which ultimately results in apoptosis and the death of the CSCs.
Collapse
Affiliation(s)
- Krushna Chandra Hembram
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Campus-11, Patia, Bhubaneswar, Odisha 751024, India
| | - Somya Ranjan Dash
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Campus-11, Patia, Bhubaneswar, Odisha 751024, India
| | - Biswajit Das
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Campus-11, Patia, Bhubaneswar, Odisha 751024, India
| | - Chinmayee Sethy
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Campus-11, Patia, Bhubaneswar, Odisha 751024, India
| | - Subhajit Chatterjee
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Campus-11, Patia, Bhubaneswar, Odisha 751024, India
| | - Birendra Kumar Bindhani
- Plant Biotechnology and Nanotechnology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Campus-11, Patia, Bhubaneswar, Odisha 751024, India
| | - Chanakya Nath Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Campus-11, Patia, Bhubaneswar, Odisha 751024, India
| |
Collapse
|
18
|
Role of Rad51 and DNA repair in cancer: A molecular perspective. Pharmacol Ther 2020; 208:107492. [PMID: 32001312 DOI: 10.1016/j.pharmthera.2020.107492] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/13/2020] [Accepted: 01/22/2020] [Indexed: 12/24/2022]
Abstract
The maintenance of genome integrity is essential for any organism survival and for the inheritance of traits to offspring. To the purpose, cells have developed a complex DNA repair system to defend the genetic information against both endogenous and exogenous sources of damage. Accordingly, multiple repair pathways can be aroused from the diverse forms of DNA lesions, which can be effective per se or via crosstalk with others to complete the whole DNA repair process. Deficiencies in DNA healing resulting in faulty repair and/or prolonged DNA damage can lead to genes mutations, chromosome rearrangements, genomic instability, and finally carcinogenesis and/or cancer progression. Although it might seem paradoxical, at the same time such defects in DNA repair pathways may have therapeutic implications for potential clinical practice. Here we provide an overview of the main DNA repair pathways, with special focus on the role played by homologous repair and the RAD51 recombinase protein in the cellular DNA damage response. We next discuss the recombinase structure and function per se and in combination with all its principal mediators and regulators. Finally, we conclude with an analysis of the manifold roles that RAD51 plays in carcinogenesis, cancer progression and anticancer drug resistance, and conclude this work with a survey of the most promising therapeutic strategies aimed at targeting RAD51 in experimental oncology.
Collapse
|
19
|
Abad E, Graifer D, Lyakhovich A. DNA damage response and resistance of cancer stem cells. Cancer Lett 2020; 474:106-117. [PMID: 31968219 DOI: 10.1016/j.canlet.2020.01.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/20/2022]
Abstract
The cancer stem cell (CSC) model defines tumors as hierarchically organized entities, containing a small population of tumorigenic CSC, or tumour-initiating cells, placed at the apex of this hierarchy. These cells may share common qualities with chemo- and radio-resistant cancer cells and contribute to self-renewal activities resulting in tumour formation, maintenance, growth and metastasis. Yet, it remains obscure what self-defense mechanisms are utilized by these cells against the chemotherapeutic drugs or radiotherapy. Recently, attention has been focused on the pivotal role of the DNA damage response (DDR) in tumorigenesis. In line with this note, an increased DDR that prevents CSC and chemoresistant cells from genotoxic pressure of chemotherapeutic drugs or radiation may be responsible for cancer metastasis. In this review, we focus on the current knowledge concerning the role of DDR in CSC and resistant cancer cells and describe the existing opportunities of re-sensitizing such cells to modulate therapeutic treatment effects.
Collapse
Affiliation(s)
- Etna Abad
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Alex Lyakhovich
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia; Vall D'Hebron Institut de Recerca, 08035, Barcelona, Spain.
| |
Collapse
|
20
|
Zou X, Zhu C, Zhang L, Zhang Y, Fu F, Chen Y, Zhou J. MicroRNA-708 Suppresses Cell Proliferation and Enhances Chemosensitivity of Cervical Cancer Cells to cDDP by Negatively Targeting Timeless. Onco Targets Ther 2020; 13:225-235. [PMID: 32021269 PMCID: PMC6966141 DOI: 10.2147/ott.s227015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/22/2019] [Indexed: 01/10/2023] Open
Abstract
Purpose Cervical cancer is the fourth most common cause of cancer-associated mortality in women worldwide. Previous studies have reported that microRNAs (miRNAs) are involved in multiple biological aspects of cancer progression by regulating gene expression. Here, we investigated the role of microRNA-708 (miR-708) in cervical cancer. Methods The expression levels of miR-708 in cervical cancer tissues and paired-normal cervical tissues were tested by quantitative polymerase chain reaction (qPCR). The interaction between miR-708 and Timeless was identified by bioinformatics method, dual-luciferase reporter assay, and Western blotting. The effects of over-expression of miR-708 on cell proliferation and cisplatin sensitivity were determined by Cell Counting Kit-8 (CCK-8) and colony formation assay. Cell cycle and apoptosis were analyzed by flow cytometry. DNA damage induced by over-expression of miR-708 was determined by comet assay. Expression levels of the genes involved in repair of DNA damage were analyzed by Western blotting. Results MiR-708 was down-regulated in cervical cancer tissues compared with paired-normal cervical tissues. By bioinformatics method, Western blotting, and dual-luciferase reporter assay, we found that Timeless was a direct target of miR-708. Furthermore, miR-708 suppressed cellular viability, colony formation, promoted apoptosis, and induced DNA damage levels. MiR-708 also enhanced chemosensitivity of cervical cancer cells to cDDP via impairing the ATR/CHK1 signaling pathway. Conclusion We conclude that miR-708 suppresses cell proliferation, facilitates cisplatin efficacy, and impairs DNA repair pathway in cervical cancer cells. These results demonstrate that miR-708 might be a candidate therapeutic target for future cervical cancer therapy.
Collapse
Affiliation(s)
- Xinwei Zou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Chenjie Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Lin Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yi Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Fengqing Fu
- Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
21
|
Sousa JFD, Serafim RB, Freitas LMD, Fontana CR, Valente V. DNA repair genes in astrocytoma tumorigenesis, progression and therapy resistance. Genet Mol Biol 2019; 43:e20190066. [PMID: 31930277 PMCID: PMC7198033 DOI: 10.1590/1678-4685-gmb-2019-0066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/21/2019] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma (GBM) is the most common and malignant type of primary brain tumor,
showing rapid development and resistance to therapies. On average, patients
survive 14.6 months after diagnosis and less than 5% survive five years or more.
Several pieces of evidence have suggested that the DNA damage signaling and
repair activities are directly correlated with GBM phenotype and exhibit
opposite functions in cancer establishment and progression. The functions of
these pathways appear to present a dual role in tumorigenesis and cancer
progression. Activation and/or overexpression of ATRX, ATM and RAD51 genes were
extensively characterized as barriers for GBM initiation, but paradoxically the
exacerbated activity of these genes was further associated with cancer
progression to more aggressive stages. Excessive amounts of other DNA repair
proteins, namely HJURP, EXO1, NEIL3, BRCA2, and BRIP, have also been connected
to proliferative competence, resistance and poor prognosis. This scenario
suggests that these networks help tumor cells to manage replicative stress and
treatment-induced damage, diminishing genome instability and conferring therapy
resistance. Finally, in this review we address promising new drugs and
therapeutic approaches with potential to improve patient survival. However,
despite all technological advances, the prognosis is still dismal and further
research is needed to dissect such complex mechanisms.
Collapse
Affiliation(s)
- Juliana Ferreira de Sousa
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, U.S.A
| | - Rodolfo Bortolozo Serafim
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Laura Marise de Freitas
- Universidade de São Paulo, Instituto de Química, Departamento de Bioquímica, São Paulo, SP, Brazil
| | - Carla Raquel Fontana
- Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas, Araraquara, SP, Brazil
| | - Valeria Valente
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Universidade de São Paulo, Ribeirão Preto, SP, Brazil.,Universidade Estadual Paulista "Júlio de Mesquita Filho" (UNESP), Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas, Araraquara, SP, Brazil.,Centro de Terapia Celular (CEPID-FAPESP), Ribeirão Preto, SP, Brazil
| |
Collapse
|
22
|
Ding J, Wu S, Zhang C, Garyali A, Martinez-Ledesma E, Gao F, Pokkulandra A, Li X, Bristow C, Carugo A, Koul D, Yung WKA. BRCA1 identified as a modulator of temozolomide resistance in P53 wild-type GBM using a high-throughput shRNA-based synthetic lethality screening. Am J Cancer Res 2019; 9:2428-2441. [PMID: 31815044 PMCID: PMC6895442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/12/2019] [Indexed: 06/10/2023] Open
Abstract
Glioblastoma multiforme (GBM), the most common type of primary brain tumor, is universally fatal, with a median survival duration ranging from 12-15 months despite maximum treatment efforts. Temozolomide (TMZ) is the current standard of care for GBM patients; however patients usually develop resistance to TMZ and limits its benefit. The identification of novel synergistic targets in GBM will lead to the development of new targeted drugs, which could be combined with broad-spectrum cytotoxic agents. In this study, we used a high-throughput synthetic lethality screen with a pooled short hairpin DNA repair library, in combination with TMZ, to identify targets that will enhance TMZ-induced antitumor effects. Using an unbiased bioinformatical analysis, we identified BRCA1 as a potential promising candidate gene that induced synthetic lethality with TMZ in glioma sphere-forming cells (GSCs). BRCA1 knockdown resulted in antitumor activity with TMZ in P53 wild-type GSCs but not in P53 mutant GSCs. TMZ treatment induced a DNA damage repair response; the activation of BRCA1 DNA repair pathway targets and knockdown of BRCA1, together with TMZ, led to increased DNA damage and cell death in P53 wild-type GSCs. Our study identified BRCA1 as a potential target that sensitizes TMZ-induced cell death in P53 wild-type GBM, suggesting that the combined inhibition of BRCA1 and TMZ treatment will be a successful targeted therapy for GBM patients.
Collapse
Affiliation(s)
- Jie Ding
- Department of Neuro-Oncology, Brain Tumor Center, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Shaofang Wu
- Department of Neuro-Oncology, Brain Tumor Center, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Chen Zhang
- Department of Neuro-Oncology, Brain Tumor Center, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Arnav Garyali
- Department of Neuro-Oncology, Brain Tumor Center, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Emmanuel Martinez-Ledesma
- Department of Neuro-Oncology, Brain Tumor Center, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la SaludMonterrey, Nuevo Leon, Mexico
| | - Feng Gao
- Department of Neuro-Oncology, Brain Tumor Center, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Adarsha Pokkulandra
- Department of Neuro-Oncology, Brain Tumor Center, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Xiaolong Li
- Department of Neuro-Oncology, Brain Tumor Center, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Christopher Bristow
- Department of Applied Cancer Science, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Alessandro Carugo
- Department of Applied Cancer Science, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - Dimpy Koul
- Department of Neuro-Oncology, Brain Tumor Center, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| | - WK Alfred Yung
- Department of Neuro-Oncology, Brain Tumor Center, The University of Texas MD Anderson Cancer CenterHouston, Texas, USA
| |
Collapse
|
23
|
Yang M, Tian X, Fan Z, Yu W, Li Z, Zhou J, Zhang W, Liang A. Targeting RAD51 enhances chemosensitivity of adult T‑cell leukemia‑lymphoma cells by reducing DNA double‑strand break repair. Oncol Rep 2019; 42:2426-2434. [PMID: 31638261 PMCID: PMC6859462 DOI: 10.3892/or.2019.7384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 01/09/2019] [Indexed: 12/19/2022] Open
Abstract
RAD51, is a key homologous recombination protein that repairs DNA damage and maintains gene diversity and stability. Previous studies have demonstrated that the over‑expression of RAD51 is associated with chemotherapy resistance of tumor cells to chemotherapy, and enhanced activity of DNA damage repair (DDR) systems contributes to resistance of adult T‑cell leukemia‑lymphoma (ATL) resistance to chemotherapy. Thus, targeting RAD51 is a potential strategy for the sensitization of ATL cells to chemotherapeutic drugs by inducing DNA damage. In general, cells can repair minor DNA damage through DDR; however, serious DNA damage may cause cell toxicity in cells which cannot be restored. In the present, down regulation of RAD51 by shRNA and imatinib sensitized Jurkat cells to etoposide by decreasing the activity of homologous recombination (HR). We found that the suppression of RAD51 by shRNA inhibited tumor cells proliferation and enhanced apoptosis of Jurkat cells after etoposide treatment. Importantly, downregulation of RAD51 by imatinib obviously increased the apoptosis of Jurkat cell after etoposide treatment. These results demonstrated that RAD51 may be of great value to as a novel target for the clinical treatment of adult T‑cell leukemia‑lymphoma (ATL), and it may improve the survival of leukemia patients.
Collapse
Affiliation(s)
- Meng Yang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R China
| | - Xiaoxue Tian
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R China
| | - Zhuoyi Fan
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R China
| | - Wenlei Yu
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R China
| | - Zheng Li
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R China
| | - Jie Zhou
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R China
| | - Wenjun Zhang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R China
| | - Aibin Liang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R China
| |
Collapse
|
24
|
Berardinelli F, Tanori M, Muoio D, Buccarelli M, di Masi A, Leone S, Ricci-Vitiani L, Pallini R, Mancuso M, Antoccia A. G-quadruplex ligand RHPS4 radiosensitizes glioblastoma xenograft in vivo through a differential targeting of bulky differentiated- and stem-cancer cells. J Exp Clin Cancer Res 2019; 38:311. [PMID: 31311580 PMCID: PMC6636127 DOI: 10.1186/s13046-019-1293-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 06/25/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Glioblastoma is the most aggressive and most lethal primary brain tumor in the adulthood. Current standard therapies are not curative and novel therapeutic options are urgently required. Present knowledge suggests that the continued glioblastoma growth and recurrence is determined by glioblastoma stem-like cells (GSCs), which display self-renewal, tumorigenic potential, and increased radio- and chemo-resistance. The G-quadruplex ligand RHPS4 displays in vitro radiosensitizing effect in GBM radioresistant cells through the targeting and dysfunctionalization of telomeres but RHPS4 and Ionizing Radiation (IR) combined treatment efficacy in vivo has not been explored so far. METHODS RHPS4 and IR combined effects were tested in vivo in a heterotopic mice xenograft model and in vitro in stem-like cells derived from U251MG and from four GBM patients. Cell growth assays, cytogenetic analysis, immunoblotting, gene expression and cytofluorimetric analysis were performed in order to characterize the response of differentiated and stem-like cells to RHPS4 and IR in single and combined treatments. RESULTS RHPS4 administration and IR exposure is very effective in blocking tumor growth in vivo up to 65 days. The tumor volume reduction and the long-term tumor control suggested the targeting of the stem cell compartment. Interestingly, RHPS4 treatment was able to strongly reduce cell proliferation in GSCs but, unexpectedly, did not synergize with IR. Lack of radiosensitization was supported by the GSCs telomeric-resistance observed as the total absence of telomere-involving chromosomal aberrations. Remarkably, RHPS4 treatment determined a strong reduction of CHK1 and RAD51 proteins and transcript levels suggesting that the inhibition of GSCs growth is determined by the impairment of the replication stress (RS) response and DNA repair. CONCLUSIONS We propose that the potent antiproliferative effect of RHPS4 in GSCs is not determined by telomeric dysfunction but is achieved by the induction of RS and by the concomitant depletion of CHK1 and RAD51, leading to DNA damage and cell death. These data open to novel therapeutic options for the targeting of GSCs, indicating that the combined inhibition of cell-cycle checkpoints and DNA repair proteins provides the most effective means to overcome resistance of GSC to genotoxic insults.
Collapse
Affiliation(s)
| | - M. Tanori
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l’Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - D. Muoio
- Department of Science, University Roma Tre, Rome, Italy
| | - M. Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - A. di Masi
- Department of Science, University Roma Tre, Rome, Italy
| | - S. Leone
- Department of Science, University Roma Tre, Rome, Italy
| | - L. Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - R. Pallini
- Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - M. Mancuso
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l’Energia e lo Sviluppo Economico Sostenibile (ENEA), Rome, Italy
| | - A. Antoccia
- Department of Science, University Roma Tre, Rome, Italy
| |
Collapse
|
25
|
Jiang M, Zhuang Y, Zu WC, Jiao L, Richard SA, Zhang S. Overexpression of EPAC2 reduces the invasion of glioma cells via MMP-2. Oncol Lett 2019; 17:5080-5086. [PMID: 31186720 PMCID: PMC6507491 DOI: 10.3892/ol.2019.10200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/26/2019] [Indexed: 01/15/2023] Open
Abstract
Exchange proteins directly activated by cAMP (EPACs) are crucial cyclic adenosine 3′,5′-monophosphate- determined signaling pathway intercessors, which are associated with the pathogenesis of neurological disorders and numerous human diseases. To the best of our knowledge, the role of EPAC2 signaling via matrix metalloproteinase 2 (MMP-2) in the pathogenesis of glioma has not been studied. Therefore, the present study focused on the role of EPAC2 in glioma, and assessed the invasiveness of human glioma cell lines following EPAC2 overexpression. Expression levels of EPAC2 in normal brain tissues and clinical glioma specimens were detected by western blotting. An EPAC2 overexpression vector was transfected into U251 and U87 cell lines to increase the expression levels of EPAC2. Expression levels of MMP-2 were detected by western blotting, and the invasive abilities of glioma cells were detected by a Transwell assay. EPAC2 was relatively highly expressed in normal brain tissue, while EPAC2 expression was significantly decreased in clinical glioma specimens (P<0.01). In vitro transfection of EPAC2 overexpression vector significantly reduced the MMP-2 protein levels of glioma cells, and, at the same time, the invasive cell number was significantly decreased in a Transwell assay. The present study demonstrated that MMP-2 regulation via EPAC2 overexpression is a novel promising therapeutic route in malignant types of glioma.
Collapse
Affiliation(s)
- Ming Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China.,Department of Neurosurgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Yan Zhuang
- Department of Neurosurgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Wang-Cun Zu
- Department of Neurosurgery, Northern Jiangsu People's Hospital, Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| | - Lei Jiao
- Department of Neurosurgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Seidu A Richard
- Department of Neurosurgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China.,Department of Immunology, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Department of Medicine, Princefield University, P.O. Box MA 128, Ho, Volta Region, Ghana
| | - Shiming Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
26
|
Sun H, Fan G, Deng C, Wu L. miR‐4429 sensitized cervical cancer cells to irradiation by targeting RAD51. J Cell Physiol 2019; 235:185-193. [PMID: 31190335 DOI: 10.1002/jcp.28957] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Hongbo Sun
- Department of Gynecology and Obstetrics Beijing ChuiYangLiu Hospital Beijing China
| | - Guimei Fan
- Department of Gynecology and Obstetrics Shanxian Central Hospital Heze Shandong China
| | - Chunxia Deng
- Department of Gynecology and Obstetrics Beijing ChuiYangLiu Hospital Beijing China
| | - Lin Wu
- Department of Gynecology and Obstetrics Xi'an XD Group Hospital Xi'an Shaanxi China
| |
Collapse
|
27
|
Liu Y, Ji W, Shergalis A, Xu J, Delaney AM, Calcaterra A, Pal A, Ljungman M, Neamati N, Rehemtulla A. Activation of the Unfolded Protein Response via Inhibition of Protein Disulfide Isomerase Decreases the Capacity for DNA Repair to Sensitize Glioblastoma to Radiotherapy. Cancer Res 2019; 79:2923-2932. [PMID: 30996048 DOI: 10.1158/0008-5472.can-18-2540] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 03/08/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022]
Abstract
Patients with glioblastoma multiforme (GBM) survive on average 12 to 14 months after diagnosis despite surgical resection followed by radiotheraphy and temozolomide therapy. Intrinsic or acquired resistance to chemo- and radiotherapy is common and contributes to a high rate of recurrence. To investigate the therapeutic potential of protein disulfide isomerase (PDI) as a target to overcome resistance to chemoradiation, we developed a GBM tumor model wherein conditional genetic ablation of prolyl 4-hydroxylase subunit beta (P4HB), the gene that encodes PDI, can be accomplished. Loss of PDI expression induced the unfolded protein response (UPR) and decreased cell survival in two independent GBM models. Nascent RNA Bru-seq analysis of PDI-depleted cells revealed a decrease in transcription of genes involved in DNA repair and cell-cycle regulation. Activation of the UPR also led to a robust decrease in RAD51 protein expression as a result of its ubiquitination-mediated proteosomal degradation. Clonogenic survival assays demonstrated enhanced killing of GBM cells in response to a combination of PDI knockdown and ionizing radiation (IR) compared with either modality alone, which correlated with a decreased capacity to repair IR-induced DNA damage. Synergistic tumor control was also observed with the combination of PDI inhibition and IR in a mouse xenograft model compared with either single agent alone. These findings provide a strong rationale for the development of PDI inhibitors and their use in combination with DNA damage-inducing, standard-of-care therapies such as IR. SIGNIFICANCE: These findings identify PDIA1 as a therapeutic target in GBM by demonstrating efficacy of its inhibition in combination with radiotherapy through a novel mechanism involving downregulation of DNA repair genes.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/11/2923/F1.large.jpg.
Collapse
Affiliation(s)
- Yajing Liu
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Wenbin Ji
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Andrea Shergalis
- Department of Medicinal Chemistry, College of Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Jiaqi Xu
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan.,Weill Cornell Graduate School of Medical Sciences, New York, New York
| | - Amy M Delaney
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Andrew Calcaterra
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Anupama Pal
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Mats Ljungman
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan.,Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Alnawaz Rehemtulla
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan.
| |
Collapse
|
28
|
Pinkham K, Park DJ, Hashemiaghdam A, Kirov AB, Adam I, Rosiak K, da Hora CC, Teng J, Cheah PS, Carvalho L, Ganguli-Indra G, Kelly A, Indra AK, Badr CE. Stearoyl CoA Desaturase Is Essential for Regulation of Endoplasmic Reticulum Homeostasis and Tumor Growth in Glioblastoma Cancer Stem Cells. Stem Cell Reports 2019; 12:712-727. [PMID: 30930246 PMCID: PMC6450460 DOI: 10.1016/j.stemcr.2019.02.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/23/2019] [Accepted: 02/26/2019] [Indexed: 12/20/2022] Open
Abstract
Inherent plasticity and various survival cues allow glioblastoma stem-like cells (GSCs) to survive and proliferate under intrinsic and extrinsic stress conditions. Here, we report that GSCs depend on the adaptive activation of ER stress and subsequent activation of lipogenesis and particularly stearoyl CoA desaturase (SCD1), which promotes ER homeostasis, cytoprotection, and tumor initiation. Pharmacological targeting of SCD1 is particularly toxic due to the accumulation of saturated fatty acids, which exacerbates ER stress, triggers apoptosis, impairs RAD51-mediated DNA repair, and achieves a remarkable therapeutic outcome with 25%-100% cure rate in xenograft mouse models. Mechanistically, divergent cell fates under varying levels of ER stress are primarily controlled by the ER sensor IRE1, which either promotes SCD1 transcriptional activation or converts to apoptotic signaling when SCD1 activity is impaired. Taken together, the dependence of GSCs on fatty acid desaturation presents an exploitable vulnerability to target glioblastoma.
Collapse
Affiliation(s)
- Kelsey Pinkham
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - David Jaehyun Park
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Arsalan Hashemiaghdam
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Aleksandar B Kirov
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Isam Adam
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Kamila Rosiak
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Cintia C da Hora
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Jian Teng
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Pike See Cheah
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Seri Kembangan, Selangor 43400, Malaysia
| | - Litia Carvalho
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA
| | - Gitali Ganguli-Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Avalon Kelly
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Christian E Badr
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02129, USA.
| |
Collapse
|
29
|
Ratnayake G, Bain AL, Fletcher N, Howard CB, Khanna KK, Thurecht KJ. RNA interference to enhance radiation therapy: Targeting the DNA damage response. Cancer Lett 2018; 439:14-23. [PMID: 30240587 DOI: 10.1016/j.canlet.2018.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/28/2018] [Accepted: 09/06/2018] [Indexed: 10/28/2022]
Abstract
RNA interference (RNAi) therapy is an emerging class of biopharmaceutical that has immense potential in cancer medicine. RNAi medicines are based on synthetic oligonucleotides that can suppress a target protein in tumour cells with high specificity. This review explores the attractive prospect of using RNAi as a radiosensitiser by targeting the DNA damage response. There are a multitude of molecular targets involved in the detection and repair of DNA damage that are suitable for this purpose. Recent developments in delivery technologies such nanoparticle carriers and conjugation strategies have allowed RNAi therapeutics to enter clinical trials in the treatment of cancer. With further progress, RNAi targeting of the DNA damage response may hold great promise in guiding radiation oncology into the era of precision medicine.
Collapse
Affiliation(s)
- G Ratnayake
- Centre of Advanced Imaging, University of Queensland, Australia; Australian Institute of Bioengineering and Nanotechnology, University of Queensland, Australia; QIMR Berghofer Medical Research Institute, Australia; Royal Brisbane and Women's Hospital, Australia.
| | - A L Bain
- QIMR Berghofer Medical Research Institute, Australia
| | - N Fletcher
- Centre of Advanced Imaging, University of Queensland, Australia; Australian Institute of Bioengineering and Nanotechnology, University of Queensland, Australia
| | - C B Howard
- Centre of Advanced Imaging, University of Queensland, Australia; Australian Institute of Bioengineering and Nanotechnology, University of Queensland, Australia
| | - K K Khanna
- QIMR Berghofer Medical Research Institute, Australia
| | - K J Thurecht
- Centre of Advanced Imaging, University of Queensland, Australia; Australian Institute of Bioengineering and Nanotechnology, University of Queensland, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australia
| |
Collapse
|
30
|
Ingham MA, McGuinness JE, Kalinsky K, Schwartz GK. Exceptional Response to Dacarbazine in Uterine Leiomyosarcoma With Homozygous BRCA2 Deletion Highlights the Role of Homologous Recombination in Response to DNA Damage From Alkylating Agents. JCO Precis Oncol 2018; 2:1-6. [DOI: 10.1200/po.18.00131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Matthew A. Ingham
- All authors: Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| | - Julia E. McGuinness
- All authors: Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| | - Kevin Kalinsky
- All authors: Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| | - Gary K. Schwartz
- All authors: Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| |
Collapse
|
31
|
Tachon G, Cortes U, Guichet PO, Rivet P, Balbous A, Masliantsev K, Berger A, Boissonnade O, Wager M, Karayan-Tapon L. Cell Cycle Changes after Glioblastoma Stem Cell Irradiation: The Major Role of RAD51. Int J Mol Sci 2018; 19:ijms19103018. [PMID: 30282933 PMCID: PMC6213228 DOI: 10.3390/ijms19103018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/01/2018] [Accepted: 10/01/2018] [Indexed: 12/26/2022] Open
Abstract
“Glioma Stem Cells” (GSCs) are known to play a role in glioblastoma (GBM) recurrence. Homologous recombination (HR) defects and cell cycle checkpoint abnormalities can contribute concurrently to the radioresistance of GSCs. DNA repair protein RAD51 homolog 1 (RAD51) is a crucial protein for HR and its inhibition has been shown to sensitize GSCs to irradiation. The aim of this study was to examine the consequences of ionizing radiation (IR) for cell cycle progression in GSCs. In addition, we intended to assess the potential effect of RAD51 inhibition on cell cycle progression. Five radiosensitive GSC lines and five GSC lines that were previously characterized as radioresistant were exposed to 4Gy IR, and cell cycle analysis was done by fluorescence-activated cell sorting (FACS) at 24, 48, 72, and 96 h with or without RAD51 inhibitor. Following 4Gy IR, all GSC lines presented a significant increase in G2 phase at 24 h, which was maintained over 72 h. In the presence of RAD51 inhibitor, radioresistant GSCs showed delayed G2 arrest post-irradiation for up to 48 h. This study demonstrates that all GSCs can promote G2 arrest in response to radiation-induced DNA damage. However, following RAD51 inhibition, the cell cycle checkpoint response differed. This study contributes to the characterization of the radioresistance mechanisms of GSCs, thereby supporting the rationale of targeting RAD51-dependent repair pathways in view of radiosensitizing GSCs.
Collapse
Affiliation(s)
- Gaelle Tachon
- Laboratoire de Neurosciences Expérimentales et Cliniques (LNEC), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1084, Université de Poitiers, F-86073 Poitiers, France.
- Département de Cancérologie Biologique, Centre Hospitalo-Universitaire de Poitiers, F-86021 Poitiers, France.
- Faculté de Médecine-Pharmacie, Université de Poitiers, F-86021 Poitiers, France.
| | - Ulrich Cortes
- Département de Cancérologie Biologique, Centre Hospitalo-Universitaire de Poitiers, F-86021 Poitiers, France.
| | - Pierre-Olivier Guichet
- Laboratoire de Neurosciences Expérimentales et Cliniques (LNEC), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1084, Université de Poitiers, F-86073 Poitiers, France.
- Département de Cancérologie Biologique, Centre Hospitalo-Universitaire de Poitiers, F-86021 Poitiers, France.
| | - Pierre Rivet
- Département de Cancérologie Biologique, Centre Hospitalo-Universitaire de Poitiers, F-86021 Poitiers, France.
| | - Anais Balbous
- Laboratoire de Neurosciences Expérimentales et Cliniques (LNEC), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1084, Université de Poitiers, F-86073 Poitiers, France.
- Département de Cancérologie Biologique, Centre Hospitalo-Universitaire de Poitiers, F-86021 Poitiers, France.
| | - Konstantin Masliantsev
- Laboratoire de Neurosciences Expérimentales et Cliniques (LNEC), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1084, Université de Poitiers, F-86073 Poitiers, France.
- Département de Cancérologie Biologique, Centre Hospitalo-Universitaire de Poitiers, F-86021 Poitiers, France.
- Faculté de Médecine-Pharmacie, Université de Poitiers, F-86021 Poitiers, France.
| | - Antoine Berger
- Département d'Oncologie Radiothérapie, Centre Hospitalo-Universitaire de Poitiers, F-86021 Poitiers, France.
| | - Odile Boissonnade
- Département d'Oncologie Radiothérapie, Centre Hospitalo-Universitaire de Poitiers, F-86021 Poitiers, France.
| | - Michel Wager
- Laboratoire de Neurosciences Expérimentales et Cliniques (LNEC), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1084, Université de Poitiers, F-86073 Poitiers, France.
- Faculté de Médecine-Pharmacie, Université de Poitiers, F-86021 Poitiers, France.
- Département de Neurochirurgie, Centre Hospitalo-Universitaire de Poitiers, F-86021 Poitiers, France.
| | - Lucie Karayan-Tapon
- Laboratoire de Neurosciences Expérimentales et Cliniques (LNEC), Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1084, Université de Poitiers, F-86073 Poitiers, France.
- Département de Cancérologie Biologique, Centre Hospitalo-Universitaire de Poitiers, F-86021 Poitiers, France.
- Faculté de Médecine-Pharmacie, Université de Poitiers, F-86021 Poitiers, France.
| |
Collapse
|
32
|
Campbell MC, Pontiggia L, Russell AY, Schwarting R, Camacho J, Jasmin JF, Mercier I. CAPER as a therapeutic target for triple negative breast cancer. Oncotarget 2018; 9:30340-30354. [PMID: 30100993 PMCID: PMC6084388 DOI: 10.18632/oncotarget.25719] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/13/2018] [Indexed: 12/31/2022] Open
Abstract
Breast cancers (BCas) that lack expression of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) are referred to as triple negative breast cancers (TNBCs) and have the poorest clinical outcome. Once these aggressive tumors progress to distant organs, the median survival decreases to 12 months. With endocrine therapies being ineffective in this BCa subtype, highly toxic chemo- and radiation therapies are the only options. A better understanding of the functional role(s) of molecular targets contributing to TNBC progression could help in the design and development of new treatments that are more targeted with less toxicity. CAPER (Co-activator of AP-1 and ER) is a nuclear transcriptional co-activator that was recently involved in ER-positive BCa progression, however its role in hormone-independent cancers remains unknown. Our current report demonstrates that CAPER expression is upregulated in human TNBC specimens compared to normal breast tissue and that its selective downregulation through a lentiviral-mediated shRNA knockdown approach resulted in decreased cell numbers in MDA-MB-231 and BT549 TNBC cell lines without affecting the growth of non-tumorigenic cell line MCF-10A. Concordant with these observations, CAPER knockdown was also associated with a decrease in DNA repair proteins leading to a marked increase in apoptosis, through caspase-3/7 activation without any changes in cell cycle. Collectively, we propose CAPER as an important signaling molecule in the development of TNBC linked to DNA repair mechanisms, which could lead to new therapeutic modalities for the treatment of this aggressive cancer.
Collapse
Affiliation(s)
- Mallory C Campbell
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA 19104, USA
| | - Laura Pontiggia
- Department of Mathematics, Physics and Statistics, Misher College of Arts and Sciences, University of the Sciences, Philadelphia, PA 19104, USA
| | - Ashley Y Russell
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA 19104, USA
| | - Roland Schwarting
- Department of Pathology, Cooper University Hospital, Camden, NJ 08103, USA
| | - Jeanette Camacho
- Department of Pathology, Cooper University Hospital, Camden, NJ 08103, USA
| | - Jean-Francois Jasmin
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA 19104, USA
| | - Isabelle Mercier
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA 19104, USA.,Program in Personalized Medicine and Targeted Therapeutics, University of the Sciences, Philadelphia, PA 19104, USA
| |
Collapse
|
33
|
Reduced expression of DNA repair genes and chemosensitivity in 1p19q codeleted lower-grade gliomas. J Neurooncol 2018; 139:563-571. [PMID: 29923053 DOI: 10.1007/s11060-018-2915-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/27/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Lower-grade gliomas (LGGs, defined as WHO grades II and III) with 1p19q codeletion have increased chemosensitivity when compared to LGGs without 1p19q codeletion, but the mechanism is currently unknown. METHODS RNAseq data from 515 LGG patients in the Cancer Genome Atlas (TCGA) were analyzed to compare the effect of expression of the 9 DNA repair genes located on chromosome arms 1p and 19q on progression free survival (PFS) and overall survival (OS) between patients who received chemotherapy and those who did not. Chemosensitivity of cells with DNA repair genes knocked down was tested using MTS cell proliferation assay in HS683 cell line and U251 cell line. RESULTS The expression of 9 DNA repair genes on 1p and 19q was significantly lower in 1p19q-codeleted tumors (n = 175) than in tumors without the codeletion (n = 337) (p < 0.001). In LGG patients who received chemotherapy, lower expression of LIG1, POLD1, PNKP, RAD54L and MUTYH was associated with longer PFS and OS. This difference between chemotherapy and non-chemotherapy groups in the association of gene expression with survival was not observed in non-DNA repair genes located on chromosome arms 1p and 19q. MTS assays showed that knockdown of DNA repair genes LIG1, POLD1, PNKP, RAD54L and MUTYH significantly inhibited recovery in response to temozolomide when compared with control group (p < 0.001). CONCLUSIONS Our results suggest that reduced expression of DNA repair genes on chromosome arms 1p and 19q may account for the increased chemosensitivity of LGGs with 1p19q codeletion.
Collapse
|
34
|
Liu X, Li P, Hirayama R, Niu Y, Liu X, Chen W, Jin X, Zhang P, Ye F, Zhao T, Liu B, Li Q. Genistein sensitizes glioblastoma cells to carbon ions via inhibiting DNA-PKcs phosphorylation and subsequently repressing NHEJ and delaying HR repair pathways. Radiother Oncol 2018; 129:84-94. [PMID: 29685705 DOI: 10.1016/j.radonc.2018.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 01/22/2018] [Accepted: 04/04/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND PURPOSE Previously, we found genistein could sensitize cancer cells to low linear energy transfer (LET) X-rays via inhibiting DNA-PKcs activities. Especially, high-LET heavy ion produces more DNA double strand breaks (DSBs) than low-LET radiation. Thus, the study was designed to investigate the detailed molecular mechanisms of genistein on sensitizing cancer cells to heavy ions. MATERIALS AND METHODS Human glioblastoma (GBM) cell lines with or without genistein pre-treatment were irradiated with high-LET carbon ions. Cell survival was determined with colony formation assay. DNA DSBs were evaluated by means of detecting γ-H2AX foci and immuno-blotting DSB repair proteins, cell apoptosis was detected using Annexin V and PI staining. The interaction of genistein with DNA-PKcs activation site was estimated by molecular docking in the autodock software. RESULTS Genistein sensitized DNA-PKcs proficient GBM cells to high-LET carbon ions via delaying the clearance of γ-H2AX foci. Genistein was physically bound to DNA-PKcs and functionally inhibited the phosphorylation of DNA-PKcs. Consequently, the non-homologous end joining (NHEJ) repair of DSBs was inhibited and the homologous recombination (HR) repair was delayed by genistein, thereby leading to an increase in apoptosis in DNA-PKcs proficient GBM cells after irradiation. CONCLUSION Our study demonstrated that genistein holds promise as a radiosensitizer for enhancing the efficacy of carbon ion radiotherapy against DNA-PKcs proficient GBM via inhibiting DNA-PKcs phosphorylation and subsequently repressing NHEJ and delaying HR repair pathways.
Collapse
Affiliation(s)
- Xiongxiong Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China
| | - Ping Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China
| | - Ryoichi Hirayama
- Research Center for Charged Particle Therapy, National Institute of Radiological Sciences, Chiba, Japan
| | - Yuzhen Niu
- Shandong Provincial Research Center for Bioinformatic Engineering and Technique, School of Life Sciences, Shandong University of Technology, Zibo, China
| | - Xinguo Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China
| | - Pengcheng Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China; University of Chinese Academy of Sciences, Beijing, China
| | - Fei Ye
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China
| | - Ting Zhao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China
| | - Bingtao Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China; University of Chinese Academy of Sciences, Beijing, China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, China.
| |
Collapse
|
35
|
Christmann M, Diesler K, Majhen D, Steigerwald C, Berte N, Freund H, Stojanović N, Kaina B, Osmak M, Ambriović-Ristov A, Tomicic MT. Integrin αVβ3 silencing sensitizes malignant glioma cells to temozolomide by suppression of homologous recombination repair. Oncotarget 2018; 8:27754-27771. [PMID: 27487141 PMCID: PMC5438606 DOI: 10.18632/oncotarget.10897] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/19/2016] [Indexed: 12/20/2022] Open
Abstract
Integrins have been suggested as possible targets in anticancer therapy. Here we show that knockdown of integrins αVβ3, αVβ5, α3β1 and α4β1 and pharmacological inhibition using a cyclo-RGD integrin αVβ3/αVβ5 antagonist sensitized multiple high-grade glioma cell lines to temozolomide (TMZ)-induced cytotoxicity. The greatest effect was observed in LN229 cells upon integrin β3 silencing, which led to inhibition of the FAK/Src/Akt/NFκB signaling pathway and increased formation of γH2AX foci. The integrin β3 knockdown led to the proteasomal degradation of Rad51, reduction of Rad51 foci and reduced repair of TMZ-induced DNA double-strand breaks by impairing homologous recombination efficiency. The down-regulation of β3 in Rad51 knockdown (LN229-Rad51kd) cells neither further sensitized them to TMZ nor increased the number of γH2AX foci, confirming causality between β3 silencing and Rad51 reduction. RIP1 was found cleaved and IκBα significantly less degraded in β3-silenced/TMZ-exposed cells, indicating inactivation of NFκB signaling. The anti-apoptotic proteins Bcl-xL, survivin and XIAP were proteasomally degraded and caspase-3/−2 cleaved. Increased H2AX phosphorylation, caspase-3 cleavage, reduced Rad51 and RIP1 expression, as well as sustained IκBα expression were also observed in mouse glioma xenografts treated with the cyclo-RGD inhibitor and TMZ, confirming the molecular mechanism in vivo. Our data indicates that β3 silencing in glioma cells represents a promising strategy to sensitize high-grade gliomas to TMZ therapy.
Collapse
Affiliation(s)
- Markus Christmann
- Department of Toxicology, University Medical Center Mainz, D-55131 Mainz, Germany
| | - Kathrin Diesler
- Department of Toxicology, University Medical Center Mainz, D-55131 Mainz, Germany
| | - Dragomira Majhen
- Laboratory for Cell Biology and Signaling, Division of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | | | - Nancy Berte
- Department of Toxicology, University Medical Center Mainz, D-55131 Mainz, Germany
| | - Halima Freund
- Department of Toxicology, University Medical Center Mainz, D-55131 Mainz, Germany
| | - Nikolina Stojanović
- Laboratory for Cell Biology and Signaling, Division of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Bernd Kaina
- Department of Toxicology, University Medical Center Mainz, D-55131 Mainz, Germany
| | - Maja Osmak
- Laboratory for Cell Biology and Signaling, Division of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Andreja Ambriović-Ristov
- Laboratory for Cell Biology and Signaling, Division of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Maja T Tomicic
- Department of Toxicology, University Medical Center Mainz, D-55131 Mainz, Germany
| |
Collapse
|
36
|
Li J, Ji X, Wang H. Targeting Long Noncoding RNA HMMR-AS1 Suppresses and Radiosensitizes Glioblastoma. Neoplasia 2018; 20:456-466. [PMID: 29574252 PMCID: PMC5915996 DOI: 10.1016/j.neo.2018.02.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/11/2018] [Accepted: 02/14/2018] [Indexed: 01/07/2023] Open
Abstract
Emergent evidences revealed that long noncoding RNAs (lncRNAs) participate in neoplastic progression. HMMR is an oncogene that is highly expressed in glioblastoma (GBM) and supports GBM growth. Whether lncRNAs regulate HMMR in GBM remains unknown. Herein, we identify that an HMMR antisense lncRNA, HMMR-AS1, is hyperexpressed in GBM cell lines and stabilizes HMMR mRNA. Knockdown of HMMR-AS1 reduces HMMR expression; inhibits cell migration, invasion, and mesenchymal phenotypes; and suppresses GBM cell growth both in vitro and in vivo. Moreover, knockdown of HMMR-AS1 radiosensitizes GBM by reducing DNA repair proteins ATM, RAD51, and BMI1. Our data demonstrate a mechanism of sense-antisense interference between HMMR and HMMR-AS1 in GBM and suggest that targeting HMMR-AS1 is a potential strategy for GBM treatment.
Collapse
Affiliation(s)
- Junyang Li
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Xiangjun Ji
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
37
|
Inhibition of RAD51 by siRNA and Resveratrol Sensitizes Cancer Stem Cells Derived from HeLa Cell Cultures to Apoptosis. Stem Cells Int 2018; 2018:2493869. [PMID: 29681946 PMCID: PMC5846439 DOI: 10.1155/2018/2493869] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 07/13/2017] [Accepted: 07/27/2017] [Indexed: 12/28/2022] Open
Abstract
Cervical cancer is the second most frequent tumor type in women worldwide with cases developing clinical recurrence, metastasis, and chemoresistance. The cancer stem cells (CSC) may be implicated in tumor resistance to therapy. RESveratrol (RES), a natural compound, is an antioxidant with multiple beneficial activities. We previously determined that the expression of RAD51 is decreased by RES. The aim of our study was to examine molecular mechanism by which CSC from HeLa cultures exhibit chemoresistance. We hypothesized CSC repair more efficiently DNA breaks and that RAD51 plays an important role in this mechanism. We found that CSC, derived from cervical cancer cell lines, overexpress RAD51 and are less sensitive to Etoposide (VP16). We inhibited RAD51 in CSC-enriched cultures using RES or siRNA against RAD51 messenger RNA and observed a decrease in cell viability and induction of apoptosis when treated simultaneously with VP16. In addition, we found that inhibition of RAD51 expression using RES also sensitizes CSC to VP16 treatment. Our results suggest that resveratrol is effective to sensitize cervical CSC because of RAD51 inhibition, targeting high RAD51 expressing CD49f-positive cells, which supports the possible therapeutic application of RES as a novel agent to treat cancer.
Collapse
|
38
|
RAD51 Is a Selective DNA Repair Target to Radiosensitize Glioma Stem Cells. Stem Cell Reports 2017; 8:125-139. [PMID: 28076755 PMCID: PMC5233453 DOI: 10.1016/j.stemcr.2016.12.005] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 12/05/2016] [Accepted: 12/05/2016] [Indexed: 11/27/2022] Open
Abstract
Patients with glioblastoma die from local relapse despite surgery and high-dose radiotherapy. Resistance to radiotherapy is thought to be due to efficient DNA double-strand break (DSB) repair in stem-like cells able to survive DNA damage and repopulate the tumor. We used clinical samples and patient-derived glioblastoma stem cells (GSCs) to confirm that the DSB repair protein RAD51 is highly expressed in GSCs, which are reliant on RAD51-dependent DSB repair after radiation. RAD51 expression and RAD51 foci numbers fall when these cells move toward astrocytic differentiation. In GSCs, the small-molecule RAD51 inhibitors RI-1 and B02 prevent RAD51 focus formation, reduce DNA DSB repair, and cause significant radiosensitization. We further demonstrate that treatment with these agents combined with radiation promotes loss of stem cells defined by SOX2 expression. This indicates that RAD51-dependent repair represents an effective and specific target in GSCs. RAD51 is overexpressed in glioma stem cells RAD51 expression levels fall when GSCs are differentiated RAD51 inhibitors abrogate DNA repair leading to radiosensitization in GSCs RAD51 inhibition + XR removes SOX2-expressing cells and abolishes clonogenicity
Collapse
|
39
|
Mueck K, Rebholz S, Harati MD, Rodemann HP, Toulany M. Akt1 Stimulates Homologous Recombination Repair of DNA Double-Strand Breaks in a Rad51-Dependent Manner. Int J Mol Sci 2017; 18:E2473. [PMID: 29156644 PMCID: PMC5713439 DOI: 10.3390/ijms18112473] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/03/2017] [Accepted: 11/03/2017] [Indexed: 12/28/2022] Open
Abstract
Akt1 is known to promote non-homologous end-joining (NHEJ)-mediated DNA double-strand break (DSB) repair by stimulation of DNA-PKcs. In the present study, we investigated the effect of Akt1 on homologous recombination (HR)-dependent repair of radiation-induced DSBs in non-small cell lung cancer (NSCLC) cells A549 and H460. Akt1-knockdown (Akt1-KD) significantly reduced Rad51 protein level, Rad51 foci formation and its colocalization with γH2AX foci after irradiation. Moreover, Akt1-KD decreased clonogenicity after treatment with Mitomycin C and HR repair, as tested by an HR-reporter assay. Double knockdown of Akt1 and Rad51 did not lead to a further decrease in HR compared to the single knockdown of Rad51. Consequently, Akt1-KD significantly increased the number of residual DSBs after irradiation partially independent of the kinase activity of DNA-PKcs. Likewise, the number of residual BRCA1 foci, indicating unsuccessful HR events, also significantly increased in the irradiated cells after Akt1-KD. Together, the results of the study indicate that Akt1 seems to be a regulatory component in the HR repair of DSBs in a Rad51-dependent manner. Thus, based on this novel role of Akt1 in HR and the previously described role of Akt1 in NHEJ, we propose that targeting Akt1 could be an effective approach to selectively improve the killing of tumor cells by DSB-inducing cytotoxic agents, such as ionizing radiation.
Collapse
Affiliation(s)
- Katharina Mueck
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tuebingen, 72076 Tuebingen, Germany.
- German Cancer Consortium (DKTK), Partner site Tuebingen, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Simone Rebholz
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tuebingen, 72076 Tuebingen, Germany.
- German Cancer Consortium (DKTK), Partner site Tuebingen, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Mozhgan Dehghan Harati
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tuebingen, 72076 Tuebingen, Germany.
- German Cancer Consortium (DKTK), Partner site Tuebingen, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - H Peter Rodemann
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tuebingen, 72076 Tuebingen, Germany.
- German Cancer Consortium (DKTK), Partner site Tuebingen, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Mahmoud Toulany
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tuebingen, 72076 Tuebingen, Germany.
- German Cancer Consortium (DKTK), Partner site Tuebingen, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
40
|
Wang S, Liu X, Qiao T, Zhang Q. Radiosensitization by CpG ODN7909 in an epidermoid laryngeal carcinoma Hep-2 cell line. J Int Med Res 2017; 45:2009-2022. [PMID: 29239279 PMCID: PMC5805225 DOI: 10.1177/0300060517728634] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Objective To evaluate the radiosensitivity effect of CpG oligodeoxyribonucleotide (ODN)
7909 on human epidermoid cancer strain-2 (Hep-2) cells in
vitro and discuss the potential for improved radiotherapy
treatment in patients with laryngeal squamous cell carcinoma. Methods Toll-like receptor (TLR) 9 expression was assessed in Hep-2
cells using Western blots and reverse transcription polymerase chain
reaction. Cell Counting Kit-8 was used to detect Hep-2 cell viability at 24
and 48 h following treatment with different CpG ODN7909 concentrations.
Cellular colonization was evaluated using microscopy. Cell cycle
distribution and apoptosis rate was determined with flow cytometry.
Interleukin (IL)-12 and tumour necrosis factor (TNF)-α concentrations were
detected by enzyme-linked immunosorbent assay. Results Hep-2 cells were found to express TLR9, and CpG ODN7909
treatment suppressed Hep-2 cell viability in a dose- and time-dependent
manner. Cell survival curve analyses revealed a sensitivity enhancement
ratio of the mean death dose of 1.225 for CpG ODN7909 plus irradiation
versus irradiation alone. Furthermore, the population of Gap 2/mitotic-phase
cells, apoptosis rate and secreted IL-12 and TNF-α levels were significantly
increased in Hep-2 cells treated with CpG ODN7909 plus irradiation versus IR
alone. Conclusion CpG ODN7909 enhanced the radiosensitivity of Hep-2 cells in
vitro.
Collapse
Affiliation(s)
| | | | | | - Qi Zhang
- Department of Oncology, Jinshan Hospital, Medical Centre of Fudan University, Jinshan District, Shanghai, China
| |
Collapse
|
41
|
Annovazzi L, Mellai M, Schiffer D. Chemotherapeutic Drugs: DNA Damage and Repair in Glioblastoma. Cancers (Basel) 2017; 9:E57. [PMID: 28587121 PMCID: PMC5483876 DOI: 10.3390/cancers9060057] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/10/2017] [Accepted: 05/22/2017] [Indexed: 11/16/2022] Open
Abstract
Despite improvements in therapeutic strategies, glioblastoma (GB) remains one of the most lethal cancers. The presence of the blood-brain barrier, the infiltrative nature of the tumor and several resistance mechanisms account for the failure of current treatments. Distinct DNA repair pathways can neutralize the cytotoxicity of chemo- and radio-therapeutic agents, driving resistance and tumor relapse. It seems that a subpopulation of stem-like cells, indicated as glioma stem cells (GSCs), is responsible for tumor initiation, maintenance and recurrence and they appear to be more resistant owing to their enhanced DNA repair capacity. Recently, attention has been focused on the pivotal role of the DNA damage response (DDR) in tumorigenesis and in the modulation of therapeutic treatment effects. In this review, we try to summarize the knowledge concerning the main molecular mechanisms involved in the removal of genotoxic lesions caused by alkylating agents, emphasizing the role of GSCs. Beside their increased DNA repair capacity in comparison with non-stem tumor cells, GSCs show a constitutive checkpoint expression that enables them to survive to treatments in a quiescent, non-proliferative state. The targeted inhibition of checkpoint/repair factors of DDR can contribute to eradicate the GSC population and can have a great potential therapeutic impact aiming at sensitizing malignant gliomas to treatments, improving the overall survival of patients.
Collapse
Affiliation(s)
- Laura Annovazzi
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100 Vercelli, Italy.
| | - Marta Mellai
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100 Vercelli, Italy.
| | - Davide Schiffer
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100 Vercelli, Italy.
| |
Collapse
|
42
|
Seidu RA, Wu M, Su Z, Xu H. Paradoxical Role of High Mobility Group Box 1 in Glioma: A Suppressor or a Promoter? Oncol Rev 2017; 11:325. [PMID: 28382190 PMCID: PMC5364998 DOI: 10.4081/oncol.2017.325] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/13/2017] [Accepted: 02/27/2017] [Indexed: 01/08/2023] Open
Abstract
Gliomas represent 60% of primary intracranial brain tumors and 80% of all malignant types, with highest morbidity and mortality worldwide. Although glioma has been extensively studied, the molecular mechanisms underlying its pathology remain poorly understood. Clarification of the molecular mechanisms involved in their development and/or treatment resistance is highly required. High mobility group box 1 protein (HMGB1) is a nuclear protein that can also act as an extracellular trigger of inflammation, proliferation and migration, through receptor for advanced glycation end products and toll like receptors in a number of cancers including gliomas. It is known that excessive release of HMGB1 in cancer leads to unlimited replicative potential, ability to develop blood vessels (angiogenesis), evasion of programmed cell death (apoptosis), self-sufficiency in growth signals, insensitivity to inhibitors of growth, inflammation, tissue invasion and metastasis. In this review we explore the mechanisms by which HMGB1 regulates apoptosis and autophagy in glioma. We also looked at how HMGB1 mediates glioma regression and promotes angiogenesis as well as possible signaling pathways with an attempt to provide potential therapeutic targets for the treatment of glioma.
Collapse
Affiliation(s)
- Richard A. Seidu
- Department of Neurosurgery, Affiliated Hospital of Jiangsu University (Jiangbin Hospital), Zhenjiang, China
- Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Min Wu
- Department of Neurosurgery, Affiliated Hospital of Jiangsu University (Jiangbin Hospital), Zhenjiang, China
| | - Zhaoliang Su
- Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Huaxi Xu
- Department of Immunology, Jiangsu University, Zhenjiang, China
| |
Collapse
|
43
|
Regulation of DNA Alkylation Damage Repair: Lessons and Therapeutic Opportunities. Trends Biochem Sci 2016; 42:206-218. [PMID: 27816326 DOI: 10.1016/j.tibs.2016.10.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/03/2016] [Accepted: 10/11/2016] [Indexed: 12/15/2022]
Abstract
Alkylation chemotherapy is one of the most widely used systemic therapies for cancer. While somewhat effective, clinical responses and toxicities of these agents are highly variable. A major contributing factor for this variability is the numerous distinct lesions that are created upon alkylation damage. These adducts activate multiple repair pathways. There is mounting evidence that the individual pathways function cooperatively, suggesting that coordinated regulation of alkylation repair is critical to prevent toxicity. Furthermore, some alkylating agents produce adducts that overlap with newly discovered methylation marks, making it difficult to distinguish between bona fide damaged bases and so-called 'epigenetic' adducts. Here, we discuss new efforts aimed at deciphering the mechanisms that regulate these repair pathways, emphasizing their implications for cancer chemotherapy.
Collapse
|
44
|
Balbous A, Cortes U, Guilloteau K, Rivet P, Pinel B, Duchesne M, Godet J, Boissonnade O, Wager M, Bensadoun RJ, Chomel JC, Karayan-Tapon L. A radiosensitizing effect of RAD51 inhibition in glioblastoma stem-like cells. BMC Cancer 2016; 16:604. [PMID: 27495836 PMCID: PMC4974671 DOI: 10.1186/s12885-016-2647-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 07/28/2016] [Indexed: 06/29/2024] Open
Abstract
Background Radioresistant glioblastoma stem cells (GSCs) contribute to tumor recurrence and identification of the molecular targets involved in radioresistance mechanisms is likely to enhance therapeutic efficacy. This study analyzed the DNA damage response following ionizing radiation (IR) in 10 GSC lines derived from patients. Methods DNA damage was quantified by Comet assay and DNA repair effectors were assessed by Low Density Array. The effect of RAD51 inhibitor, RI-1, was evaluated by comet and annexin V assays. Results While all GSC lines displayed efficient DNA repair machinery following ionizing radiation, our results demonstrated heterogeneous responses within two distinct groups showing different intrinsic radioresistance, up to 4Gy for group 1 and up to 8Gy for group 2. Radioresistant cell group 2 (comprising 5 out of 10 GSCs) showed significantly higher RAD51 expression after IR. In these cells, inhibition of RAD51 prevented DNA repair up to 180 min after IR and induced apoptosis. In addition, RAD51 protein expression in glioblastoma seems to be associated with poor progression-free survival. Conclusion These results underscore the importance of RAD51 in radioresistance of GSCs. RAD51 inhibition could be a therapeutic strategy helping to treat a significant number of glioblastoma, in combination with radiotherapy. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2647-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anaïs Balbous
- INSERM1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, F-86021, France.,Université de Poitiers, U1084, Poitiers, F-86022, France.,CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, F-86021, France
| | - Ulrich Cortes
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, F-86021, France
| | - Karline Guilloteau
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, F-86021, France
| | - Pierre Rivet
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, F-86021, France
| | - Baptiste Pinel
- CHU de Poitiers, Service d'Oncologie Radiotherapique, Poitiers, F86021, France
| | - Mathilde Duchesne
- CHU de Poitiers, Service d'Anatomo-cytopathologie, Poitiers, F86021, France
| | - Julie Godet
- CHU de Poitiers, Service d'Anatomo-cytopathologie, Poitiers, F86021, France
| | - Odile Boissonnade
- CHU de Poitiers, Service d'Oncologie Radiotherapique, Poitiers, F86021, France
| | - Michel Wager
- CHU de Poitiers, Service de Neurochirurgie, Poitiers, F86021, France
| | - René Jean Bensadoun
- CHU de Poitiers, Service d'Oncologie Radiotherapique, Poitiers, F86021, France
| | - Jean-Claude Chomel
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, F-86021, France
| | - Lucie Karayan-Tapon
- INSERM1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, F-86021, France. .,Université de Poitiers, U1084, Poitiers, F-86022, France. .,CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, F-86021, France.
| |
Collapse
|
45
|
Erasimus H, Gobin M, Niclou S, Van Dyck E. DNA repair mechanisms and their clinical impact in glioblastoma. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 769:19-35. [PMID: 27543314 DOI: 10.1016/j.mrrev.2016.05.005] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/04/2016] [Indexed: 12/18/2022]
Abstract
Despite surgical resection and genotoxic treatment with ionizing radiation and the DNA alkylating agent temozolomide, glioblastoma remains one of the most lethal cancers, due in great part to the action of DNA repair mechanisms that drive resistance and tumor relapse. Understanding the molecular details of these mechanisms and identifying potential pharmacological targets have emerged as vital tasks to improve treatment. In this review, we introduce the various cellular systems and animal models that are used in studies of DNA repair in glioblastoma. We summarize recent progress in our knowledge of the pathways and factors involved in the removal of DNA lesions induced by ionizing radiation and temozolomide. We introduce the therapeutic strategies relying on DNA repair inhibitors that are currently being tested in vitro or in clinical trials, and present the challenges raised by drug delivery across the blood brain barrier as well as new opportunities in this field. Finally, we review the genetic and epigenetic alterations that help shape the DNA repair makeup of glioblastoma cells, and discuss their potential therapeutic impact and implications for personalized therapy.
Collapse
Affiliation(s)
- Hélène Erasimus
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (LIH), 84 Val Fleuri, L-1526 Luxembourg, Luxembourg
| | - Matthieu Gobin
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (LIH), 84 Val Fleuri, L-1526 Luxembourg, Luxembourg
| | - Simone Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (LIH), 84 Val Fleuri, L-1526 Luxembourg, Luxembourg
| | - Eric Van Dyck
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (LIH), 84 Val Fleuri, L-1526 Luxembourg, Luxembourg.
| |
Collapse
|
46
|
Kaminsky N, Bihari O, Kanner S, Barzilai A. Connecting Malfunctioning Glial Cells and Brain Degenerative Disorders. GENOMICS, PROTEOMICS & BIOINFORMATICS 2016; 14:155-165. [PMID: 27245308 PMCID: PMC4936608 DOI: 10.1016/j.gpb.2016.04.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 12/19/2022]
Abstract
The DNA damage response (DDR) is a complex biological system activated by different types of DNA damage. Mutations in certain components of the DDR machinery can lead to genomic instability disorders that culminate in tissue degeneration, premature aging, and various types of cancers. Intriguingly, malfunctioning DDR plays a role in the etiology of late onset brain degenerative disorders such as Parkinson's, Alzheimer's, and Huntington's diseases. For many years, brain degenerative disorders were thought to result from aberrant neural death. Here we discuss the evidence that supports our novel hypothesis that brain degenerative diseases involve dysfunction of glial cells (astrocytes, microglia, and oligodendrocytes). Impairment in the functionality of glial cells results in pathological neuro-glial interactions that, in turn, generate a "hostile" environment that impairs the functionality of neuronal cells. These events can lead to systematic neural demise on a scale that appears to be proportional to the severity of the neurological deficit.
Collapse
Affiliation(s)
- Natalie Kaminsky
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ofer Bihari
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sivan Kanner
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Ari Barzilai
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel.
| |
Collapse
|
47
|
Yant L, Bomblies K. Genome management and mismanagement--cell-level opportunities and challenges of whole-genome duplication. Genes Dev 2016; 29:2405-19. [PMID: 26637526 PMCID: PMC4691946 DOI: 10.1101/gad.271072.115] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Whole-genome duplication (WGD) doubles the DNA content in the nucleus and leads to polyploidy. In this review, Yant and Bomblies discuss both the adaptive potential and problems associated with WGD, focusing primarily on cellular effects. Whole-genome duplication (WGD) doubles the DNA content in the nucleus and leads to polyploidy. In whole-organism polyploids, WGD has been implicated in adaptability and the evolution of increased genome complexity, but polyploidy can also arise in somatic cells of otherwise diploid plants and animals, where it plays important roles in development and likely environmental responses. As with whole organisms, WGD can also promote adaptability and diversity in proliferating cell lineages, although whether WGD is beneficial is clearly context-dependent. WGD is also sometimes associated with aging and disease and may be a facilitator of dangerous genetic and karyotypic diversity in tumorigenesis. Scaling changes can affect cell physiology, but problems associated with WGD in large part seem to arise from problems with chromosome segregation in polyploid cells. Here we discuss both the adaptive potential and problems associated with WGD, focusing primarily on cellular effects. We see value in recognizing polyploidy as a key player in generating diversity in development and cell lineage evolution, with intriguing parallels across kingdoms.
Collapse
Affiliation(s)
- Levi Yant
- John Innes Centre, Colney, Norwich NR4 7UH, United Kingdom
| | | |
Collapse
|
48
|
Kitange GJ, Mladek AC, Schroeder MA, Pokorny JC, Carlson BL, Zhang Y, Nair AA, Lee JH, Yan H, Decker PA, Zhang Z, Sarkaria JN. Retinoblastoma Binding Protein 4 Modulates Temozolomide Sensitivity in Glioblastoma by Regulating DNA Repair Proteins. Cell Rep 2016; 14:2587-98. [PMID: 26972001 DOI: 10.1016/j.celrep.2016.02.045] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 12/22/2015] [Accepted: 02/04/2016] [Indexed: 01/18/2023] Open
Abstract
Here we provide evidence that RBBP4 modulates temozolomide (TMZ) sensitivity through coordinate regulation of two key DNA repair genes critical for recovery from TMZ-induced DNA damage: methylguanine-DNA-methyltransferase (MGMT) and RAD51. Disruption of RBBP4 enhanced TMZ sensitivity, induced synthetic lethality to PARP inhibition, and increased DNA damage signaling in response to TMZ. Moreover, RBBP4 silencing enhanced TMZ-induced H2AX phosphorylation and apoptosis in GBM cells. Intriguingly, RBBP4 knockdown suppressed the expression of MGMT, RAD51, and other genes in association with decreased promoter H3K9 acetylation (H3K9Ac) and increased H3K9 tri-methylation (H3K9me3). Consistent with these data, RBBP4 interacts with CBP/p300 to form a chromatin-modifying complex that binds within the promoter of MGMT, RAD51, and perhaps other genes. Globally, RBBP4 positively and negatively regulates genes involved in critical cellular functions including tumorigenesis. The RBBP4/CBP/p300 complex may provide an interesting target for developing therapy-sensitizing strategies for GBM and other tumors.
Collapse
Affiliation(s)
- Gaspar J Kitange
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Ann C Mladek
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Mark A Schroeder
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jenny C Pokorny
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Brett L Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Yuji Zhang
- Department of Biostatistics and Bioinformatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Asha A Nair
- Department of Biostatistics and Bioinformatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jeong-Heon Lee
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Huihuang Yan
- Department of Biostatistics and Bioinformatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Paul A Decker
- Department of Biostatistics and Bioinformatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhiguo Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
49
|
Xipell E, Aragón T, Martínez-Velez N, Vera B, Idoate MA, Martínez-Irujo JJ, Garzón AG, Gonzalez-Huarriz M, Acanda AM, Jones C, Lang FF, Fueyo J, Gomez-Manzano C, Alonso MM. Endoplasmic reticulum stress-inducing drugs sensitize glioma cells to temozolomide through downregulation of MGMT, MPG, and Rad51. Neuro Oncol 2016; 18:1109-19. [PMID: 26951384 DOI: 10.1093/neuonc/now022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 01/29/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Endoplasmic reticulum (ER) stress results from protein misfolding imbalance and has been postulated as a therapeutic strategy. ER stress activates the unfolded protein response which leads to a complex cellular response, including the upregulation of aberrant protein degradation in the ER, with the goal of resolving that stress. O(6)-methylguanine DNA methyltransferase (MGMT), N-methylpurine DNA glycosylase (MPG), and Rad51 are DNA damage repair proteins that mediate resistance to temozolomide in glioblastoma. In this work we sought to evaluate whether ER stress-inducing drugs were able to downmodulate DNA damage repair proteins and become candidates to combine with temozolomide. METHODS MTT assays were performed to evaluate the cytotoxicity of the treatments. The expression of proteins was evaluated using western blot and immunofluorescence. In vivo studies were performed using 2 orthotopic glioblastoma models in nude mice to evaluate the efficacy of the treatments. All statistical tests were 2-sided. RESULTS Treatment of glioblastoma cells with ER stress-inducing drugs leads to downregulation of MGMT, MPG, and Rad51. Inhibition of ER stress through pharmacological treatment resulted in rescue of MGMT, MPG, and Rad51 protein levels. Moreover, treatment of glioblastoma cells with salinomycin, an ER stress-inducing drug, and temozolomide resulted in enhanced DNA damage and a synergistic antitumor effect in vitro. Of importance, treatment with salinomycin/temozolomide resulted in a significant antiglioma effect in 2 aggressive orthotopic intracranial brain tumor models. CONCLUSIONS These findings provide a strong rationale for combining temozolomide with ER stress-inducing drugs as an alternative therapeutic strategy for glioblastoma.
Collapse
Affiliation(s)
- Enric Xipell
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Tomás Aragón
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Naiara Martínez-Velez
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Beatriz Vera
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Miguel Angel Idoate
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Juan José Martínez-Irujo
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Antonia García Garzón
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Marisol Gonzalez-Huarriz
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Arlet M Acanda
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Chris Jones
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Frederick F Lang
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Juan Fueyo
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Candelaria Gomez-Manzano
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| | - Marta M Alonso
- Department of Medical Oncology, University Hospital of Navarra, Pamplona, Navarra, Spain (E.X., N.M.-V, B.V., M.G.-H, A.M.A, M.M.A); Department of Hepatology, Foundation for Applied Medical Research, Pamplona, Navarra, Spain (T.A.); Department of Pathology, University Hospital of Navarra, Pamplona, Navarra, Spain (M.A.I); Department of Biochemistry, University of Navarra, Pamplona, Navarra, Spain (J.J.M.-I, A.G.G); Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London, UK (C.J.); Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas (F.F.L, C.G.-M., J.F.)
| |
Collapse
|
50
|
Chai KM, Wang CY, Liaw HJ, Fang KM, Yang CS, Tzeng SF. Downregulation of BRCA1-BRCA2-containing complex subunit 3 sensitizes glioma cells to temozolomide. Oncotarget 2015; 5:10901-15. [PMID: 25337721 PMCID: PMC4279418 DOI: 10.18632/oncotarget.2543] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 09/29/2014] [Indexed: 12/19/2022] Open
Abstract
We previously found that BRCA1-BRCA2-containing complex subunit 3 (BRCC3) was highly expressed in tumorigenic rat glioma cells. However, the functional role of BRCC3 in human glioma cells remains to be characterized. This study indicated that the upregulation of BRCC3 expression was induced in two human malignant glioblastoma U251 and A172 cell lines following exposure to the alkylating agent, temozolomide (TMZ). Homologous recombination (HR)-dependent DNA repair-associated genes (i.e. BRCA1, BRCA2, RAD51 and FANCD2) were also increased in U251 and A172 cells after treatment with TMZ. BRCC3 gene knockdown through lentivirus-mediated gene knockdown approach not only significantly reduced the clonogenic and migratory abilities of U251 and A172 cells, but also enhanced their sensitization to TMZ. The increase in phosphorylated H2AX foci (γH2AX) formation, an indicator of DNA damage, persisted in TMZ-treated glioma cells with stable knockdown BRCC3 expression, suggesting that BRCC3 gene deficiency is associated with DNA repair impairment. In summary, we demonstrate that by inducing DNA repair, BRCC3 renders glioma cells resistant to TMZ. The findings point to BRCC3 as a potential target for treatment of alkylating drug-resistant glioma.
Collapse
Affiliation(s)
- Kit Man Chai
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan City, 70101, Taiwan
| | - Chih-Yen Wang
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan City, 70101, Taiwan
| | - Hung-Jiun Liaw
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan City, 70101, Taiwan
| | - Kuan-Min Fang
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan City, 70101, Taiwan
| | - Chung-Shi Yang
- Center for Nanomedicine Research, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Shun-Fen Tzeng
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan City, 70101, Taiwan
| |
Collapse
|