1
|
Repolês BM, Rodrigues Ferreira WR, de Assis AV, Mendes IC, Morini FS, Gonçalves CS, Costa Catta-Preta CM, Kelley SO, Franco GR, Macedo AM, Mottram JC, Motta MCM, Fragoso SP, Machado CR. Transcription coupled repair occurrence in Trypanosoma cruzi mitochondria. Mitochondrion 2025; 83:102009. [PMID: 39993491 DOI: 10.1016/j.mito.2025.102009] [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: 09/30/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025]
Abstract
Although several proteins involved in DNA repair systems have been identified in the T. cruzi mitochondrion, limited information is available regarding the specific DNA repair mechanisms responsible for kinetoplast DNA (kDNA) maintenance. The kDNA, contained within a single mitochondrion, exhibits a highly complex replication mechanism compared to the mitochondrial DNA of other eukaryotes. The absence of additional mitochondria makes the proper maintenance of this single mitochondrion essential for parasite viability. Trypanosomatids possess a distinct set of proteins dedicated to kDNA organization and metabolism, known as kinetoplast-associated proteins (KAPs). Despite studies identifying the localization of these proteins, their functions remain largely unclear. Here, we demonstrate that TcKAP7 is involved in the repair of kDNA lesions induced by UV radiation and cisplatin. TcKAP7 mutant cells exhibited phenotypes similar to those observed in Angomonas deanei following the deletion of this gene. This monoxenic trypanosomatid colonizes the gastrointestinal tract of insects and possesses a kinetoplast with a distinct shape and kDNA topology compared to T. cruzi, making it a suitable comparative model in this study. Additionally, we observed that DNA damage can trigger distinct signaling pathways leading to cell death. Furthermore, we elucidated the involvement of CSB in this response, suggesting a potential interaction between TcKAP7 and CSB proteins in transcription-coupled DNA repair. The results presented here describe, for the first time, the mechanism of mitochondrial DNA repair in trypanosomatids following exposure to UV radiation and cisplatin.
Collapse
Affiliation(s)
- Bruno Marçal Repolês
- Laboratório de Genética Bioquímica, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brasil
| | - Wesley Roger Rodrigues Ferreira
- Laboratório de Genética Bioquímica, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brasil
| | - Antônio Vinicius de Assis
- Laboratório de Genética Bioquímica, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brasil
| | - Isabela Cecília Mendes
- Laboratório de Genética Bioquímica, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brasil
| | - Flávia Souza Morini
- Laboratório de Biologia Molecular e Sistêmica de Tripanossomatídeos, Instituto Carlos Chagas, Fundação Oswaldo Cruz, Curitiba, PR, Brasil
| | - Camila Silva Gonçalves
- Laboratório de Ultraestrutura Celular Hertha Meyer, Centro de Pesquisa em Medicina de Precisão (CPMP), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro-UFRJ, Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagens - Rio de Janeiro, RJ, Brazil
| | | | - Shana O Kelley
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Glória Regina Franco
- Laboratório de Genética Bioquímica, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brasil
| | - Andrea Mara Macedo
- Laboratório de Genética Bioquímica, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brasil
| | - Jeremy C Mottram
- Department of Biology, York Biomedical Research Institute, University of York, Wentworth Way, Heslington YorkYO10 5DD, UK
| | - Maria Cristina M Motta
- Laboratório de Ultraestrutura Celular Hertha Meyer, Centro de Pesquisa em Medicina de Precisão (CPMP), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro-UFRJ, Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagens - Rio de Janeiro, RJ, Brazil
| | - Stênio Perdigão Fragoso
- Laboratório de Biologia Molecular e Sistêmica de Tripanossomatídeos, Instituto Carlos Chagas, Fundação Oswaldo Cruz, Curitiba, PR, Brasil
| | - Carlos Renato Machado
- Laboratório de Genética Bioquímica, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brasil.
| |
Collapse
|
2
|
Ajay AK, Akinfolarin AA, Gifford CC, Sabbisetti VS, Bonventre JV. Breast cancer gene-1 (BRCA1) potentiates maladaptive repair after kidney injury. J Exp Med 2025; 222:e20231107. [PMID: 40152784 PMCID: PMC11951932 DOI: 10.1084/jem.20231107] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/17/2024] [Accepted: 12/06/2024] [Indexed: 03/29/2025] Open
Abstract
Maladaptive repair following kidney tubular injury leads to the development of interstitial fibrosis, a pathology common to chronic kidney diseases (CKD). Dysfunctional DNA damage response plays an important role in the progression of CKD. We found that BRCA1 expression was increased in the kidneys of patients with CKD and fibrotic kidneys of mice. Exon 11 deletion of Brca1 in proximal tubule cells (PTCs) of mice subjected to ischemic or nephrotoxic (aristolochic acid) injury resulted in a reduced number of senescent cells, as assessed by a decrease in phospho-histone H3, p16INK4a, RAD51 recruitment, G2/M cell cycle phase cells, GATA4, and senescence-associated β-galactosidase. There was less production of inflammatory profibrotic mediators and reduced kidney fibrosis. After cisplatin exposure in vitro, human PTCs with reduced BRCA1 had increased apoptosis, decreased RAD51 nuclear foci, and fewer cells in the G2/M cell cycle phase, with reduced IL-6 and sonic hedgehog production. Thus, BRCA1 regulates nonmalignant tissue responses to kidney injury, a role hitherto unrecognized.
Collapse
Affiliation(s)
- Amrendra K. Ajay
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Akinwande A. Akinfolarin
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Cody C. Gifford
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Venkata S. Sabbisetti
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph V. Bonventre
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
3
|
Kim HR, Hong JK, Kim Y, Choi JY. HEBP2 affects sensitivity to cisplatin and BCNU but not to paclitaxel in MDA-MB-231 breast cancer cells. Toxicol Res 2024; 40:561-569. [PMID: 39345749 PMCID: PMC11436541 DOI: 10.1007/s43188-024-00249-w] [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: 04/22/2024] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 10/01/2024] Open
Abstract
Breast cancer has the highest incidence of all cancer types in women. Triple-negative breast cancer (TNBC) accounts for 15% of all breast cancer cases and is the most aggressive type, with a poor prognosis and limited treatment. Treatment failure in patients is largely due to resistance to chemotherapy. In this study, we aimed to identify the novel factors contributing to chemoresistance in TNBC using cisplatin and 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU). We found that transactivation of the heme-binding protein 2 (HEBP2) gene was common in surviving colonies of cells after exposure to two types of chemotherapeutic agents, namely cisplatin and BCNU, from genome-scale transcriptional activation library screening in the TNBC cell line MDA-MB-231. Analysis of a public database (Proteogenomic Landscape of Breast Cancer, CPTAC) indicated that HEBP2 mRNA expression was elevated in TNBC tissues compared to that in non-TNBC tissues. HEBP2 facilitates necrotic cell death under oxidative stress; however, it is not yet known whether HEBP2 affects cancer cell survival following chemotherapy. Therefore, we investigated the effects of HEBP2 expression on the sensitivity to cisplatin and BCNU in MDA-MB-231 cells. Overexpression of HEBP2 significantly enhanced the viability of MDA-MB-231 cells in response to cisplatin and BCNU, but not methyl methanesulfonate (MMS) and paclitaxel. In contrast, CRISPR/Cas9-mediated HEBP2-knockout greatly reduced cell viability in response to cisplatin and BCNU, but not to MMS and paclitaxel, in MDA-MB-231 cells. Moreover, the exogenous introduction of HEBP2 restored the resistance of HEBP2-deficient cells to cisplatin and BCNU to wild-type levels. These findings suggest that HEBP2 may play a significant role in resistance to cisplatin and BCNU, which induce intrastrand and interstrand DNA crosslinks, but not to monoalkylating or microtubule-stabilizing agents in TNBC cells. The possibility exists that HEBP2 serves as a biomarker for predicting response or a therapeutic target for overcoming resistance to platinum-based and alkylating anticancer agents in TNBC.
Collapse
Affiliation(s)
- Hye Rim Kim
- Department of Pharmacology, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419 Republic of Korea
| | - Jin-Kyung Hong
- Department of Pharmacology, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419 Republic of Korea
| | - Yongsub Kim
- Department of Cell and Genetic Engineering, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505 Republic of Korea
| | - Jeong-Yun Choi
- Department of Pharmacology, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419 Republic of Korea
| |
Collapse
|
4
|
Abbouche L, Bythell-Douglas R, Deans AJ. FANCM branchpoint translocase: Master of traverse, reverse and adverse DNA repair. DNA Repair (Amst) 2024; 140:103701. [PMID: 38878565 DOI: 10.1016/j.dnarep.2024.103701] [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: 01/16/2024] [Revised: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 07/13/2024]
Abstract
FANCM is a multifunctional DNA repair enzyme that acts as a sensor and coordinator of replication stress responses, especially interstrand crosslink (ICL) repair mediated by the Fanconi anaemia (FA) pathway. Its specialised ability to bind and remodel branched DNA structures enables diverse genome maintenance activities. Through ATP-powered "branchpoint translocation", FANCM can promote fork reversal, facilitate replication traverse of ICLs, resolve deleterious R-loop structures, and restrain recombination. These remodelling functions also support a role as sensor of perturbed replication, eliciting checkpoint signalling and recruitment of downstream repair factors like the Fanconi anaemia FANCI:FANCD2 complex. Accordingly, FANCM deficiency causes chromosome fragility and cancer susceptibility. Other recent advances link FANCM to roles in gene editing efficiency and meiotic recombination, along with emerging synthetic lethal relationships, and targeting opportunities in ALT-positive cancers. Here we review key properties of FANCM's biochemical activities, with a particular focus on branchpoint translocation as a distinguishing characteristic.
Collapse
Affiliation(s)
- Lara Abbouche
- Genome Stability Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; Department of Medicine (St Vincent's), University of Melbourne, Fitzroy, VIC, Australia
| | - Rohan Bythell-Douglas
- Genome Stability Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Andrew J Deans
- Genome Stability Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; Department of Medicine (St Vincent's), University of Melbourne, Fitzroy, VIC, Australia.
| |
Collapse
|
5
|
Ahmed A, Kato N, Gautier J. Replication-Independent ICL Repair: From Chemotherapy to Cell Homeostasis. J Mol Biol 2024; 436:168618. [PMID: 38763228 PMCID: PMC11227339 DOI: 10.1016/j.jmb.2024.168618] [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: 03/18/2024] [Revised: 05/03/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Interstrand crosslinks (ICLs) are a type of covalent lesion that can prevent transcription and replication by inhibiting DNA strand separation and instead trigger cell death. ICL inducing compounds are commonly used as chemotherapies due to their effectiveness in inhibiting cell proliferation. Naturally occurring crosslinking agents formed from metabolic processes can also pose a challenge to genome stability especially in slowly or non-dividing cells. Cells maintain a variety of ICL repair mechanisms to cope with this stressor within and outside the S phase of the cell cycle. Here, we discuss the mechanisms of various replication-independent ICL repair pathways and how crosslink repair efficiency is tied to aging and disease.
Collapse
Affiliation(s)
- Arooba Ahmed
- Institute for Cancer Genetics, Columbia University Vagelos, College of Physicians and Surgeons, New York, NY, USA
| | - Niyo Kato
- Institute for Cancer Genetics, Columbia University Vagelos, College of Physicians and Surgeons, New York, NY, USA
| | - Jean Gautier
- Institute for Cancer Genetics, Columbia University Vagelos, College of Physicians and Surgeons, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos, College of Physicians and Surgeons, New York, NY, USA; Department of Genetics and Development, Columbia University Vagelos, College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
6
|
Wufuer R, Liu K, Feng J, Wang M, Hu S, Chen F, Lin S, Zhang Y. Distinct mechanisms by which Nrf1 and Nrf2 as drug targets contribute to the anticancer efficacy of cisplatin on hepatoma cells. Free Radic Biol Med 2024; 213:488-511. [PMID: 38278308 DOI: 10.1016/j.freeradbiomed.2024.01.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/23/2023] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
Cisplatin (cis-Dichlorodiamineplatinum[II], CDDP) is generally accepted as a platinum-based alkylating agent type of the DNA-damaging anticancer drug, which is widely administrated in clinical treatment of many solid tumors. The pharmacological effect of CDDP is mainly achieved by replacing the chloride ion (Cl-) in its structure with H2O to form active substances with the strong electrophilic properties and then react with any nucleophilic molecules, primarily leading to genomic DNA damage and subsequent cell death. In this process, those target genes driven by the consensus electrophilic and/or antioxidant response elements (EpREs/AREs) in their promoter regions are also activated or repressed by CDDP. Thereby, we here examined the expression profiling of such genes regulated by two principal antioxidant transcription factors Nrf1 and Nrf2 (both encoded by Nfe2l1 and Nfe2l2, respectively) in diverse cellular signaling responses to this intervention. The results demonstrated distinct cellular metabolisms, molecular pathways and signaling response mechanisms by which Nrf1 and Nrf2 as the drug targets differentially contribute to the anticancer efficacy of CDDP on hepatoma cells and xenograft tumor mice. Interestingly, the role of Nrf1, rather than Nrf2, is required for the anticancer effect of CDDP, to suppress malignant behavior of HepG2 cells by differentially monitoring multi-hierarchical signaling to gene regulatory networks. To our surprise, it was found there exists a closer relationship of Nrf1α than Nrf2 with DNA repair, but the hyperactive Nrf2 in Nrf1α-∕- cells manifests a strong correlation with its resistance to CDDP, albeit their mechanistic details remain elusive.
Collapse
Affiliation(s)
- Reziyamu Wufuer
- Bioengineering College and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China.
| | - Keli Liu
- Bioengineering College and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China.
| | - Jing Feng
- Bioengineering College and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402260, China.
| | - Meng Wang
- Bioengineering College and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China.
| | - Shaofan Hu
- Bioengineering College and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China.
| | - Feilong Chen
- Bioengineering College and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402260, China.
| | - Shanshan Lin
- Bioengineering College and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China.
| | - Yiguo Zhang
- Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China.
| |
Collapse
|
7
|
Omar NE, Elewa H. Cisplatin-induced ototoxicity: a novel approach to an ancient problem. Pharmacogenet Genomics 2023; 33:111-115. [PMID: 37068004 DOI: 10.1097/fpc.0000000000000497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
With the scarcity of pharmacological otoprotective agents against cisplatin-induced ototoxicity (CIO), researchers find themselves compelled to look at and navigate all possible strategies to identify ways to prevent CIO. One of these promising strategies is pharmacogenomic implementation. This strategy aims for identifying and detecting high-risk genetic variants to tailor cisplatin therapy to reach the best survival outcomes with the least risk of ototoxicity.
Collapse
Affiliation(s)
- Nabil E Omar
- Pharmacy Department, National Center for Cancer Care and Research, Hamad Medical Corporation
- Clinical and Population Health Research, College of Pharmacy, Qatar University, Doha, Qatar
| | - Hazem Elewa
- Clinical and Population Health Research, College of Pharmacy, Qatar University, Doha, Qatar
| |
Collapse
|
8
|
Huang Z, Chen Y, Chen R, Zhou B, Wang Y, Hong L, Wang Y, Wang J, Xu X, Huang Z, Chen W. HPV Enhances HNSCC Chemosensitization by Inhibiting SERPINB3 Expression to Disrupt the Fanconi Anemia Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 10:e2202437. [PMID: 36382555 PMCID: PMC9811475 DOI: 10.1002/advs.202202437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 09/06/2022] [Indexed: 06/16/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the most common malignant tumor of the head and neck, and the prognosis of patients is poor due to chemotherapeutic resistance. Interestingly, patients with HNSCC induced by human papillomavirus (HPV) infection are more sensitive to chemotherapy and display a better prognosis than HPV-negative patients. The biological relevance of HPV infection and the mechanism underlying chemosensitivity to cisplatin remain unknown. Herein, SERPINB3 is identified as an important target for regulation of cisplatin sensitivity by HPV-E6/E7 in HNSCC. Downregulation of SERPINB3 inhibits cisplatin-induced DNA damage repair and enhances the cytotoxicity of cisplatin. In detail, decreasing SERPINB3 expression reduces the USP1-mediated deubiquitination of FANCD2-FANCI in the Fanconi anemia pathway, thereby interfering with cisplatin-induced DNA interstrand crosslinks repair and further contributing to HNSCC cell apoptosis. To translate this finding, pH-responsive nanoparticles are used to deliver SERPINB3 small interfering RNA in combination with cisplatin, and this treatment successfully reverses cisplatin chemotherapeutic resistance in a patient-derived xenograft model from HPV-negative HNSCC. Taken together, these findings suggest that targeting SERPINB3 based on HPV-positive HNSCC is a potential strategy to overcome cisplatin resistance in HPV-negative HNSCC and improves the prognosis of this disease.
Collapse
Affiliation(s)
- Zixian Huang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Yongju Chen
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Rui Chen
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Bin Zhou
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Yongqiang Wang
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Lei Hong
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Yuepeng Wang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Jianguang Wang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Xiaoding Xu
- Medical Research CenterSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Zhiquan Huang
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| | - Weiliang Chen
- Department of Oral and Maxillofacial SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510120P. R. China
| |
Collapse
|
9
|
Seah JWK, Lee JXT, Li Y, Pullarkat SA, Tan NS, Leung PH. Chelating Phosphine-N-Heterocyclic Carbene Platinum Complexes via Catalytic Asymmetric Hydrophosphination and Their Cytotoxicity Toward MKN74 and MCF7 Cancer Cell Lines. Inorg Chem 2021; 60:17276-17287. [PMID: 34709031 DOI: 10.1021/acs.inorgchem.1c02625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A series of activated vinyl azoles was hydrophosphinated in the presence of a chiral palladacycle catalyst under mild conditions to give enantioenriched phosphine azoles with moderate enantioselectivities and yields. The racemic phosphine azoles were transformed into eleven novel chelating phosphine-N-heterocyclic carbene (NHC) platinum complexes. The drug efficacies of nine selected phosphine-NHC platinum(II) chlorides in two cancer cell lines (MKN74 and MCF7) were evaluated, and two were found to exhibit activities comparable to that of cisplatin.
Collapse
Affiliation(s)
- Jeffery Wee Kiong Seah
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | - Jeannie Xue Ting Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
| | - Yongxin Li
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | - Sumod A Pullarkat
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Pak-Hing Leung
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
| |
Collapse
|
10
|
Zhang J, Ma Y, Hu K, Feng Y, Chen S, Yang X, Fong-Chuen Loo J, Zhang H, Yin F, Li Z. Surface Coordination of Black Phosphorus with Modified Cisplatin. Bioconjug Chem 2019; 30:1658-1664. [PMID: 31070357 DOI: 10.1021/acs.bioconjchem.9b00128] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Black phosphorus (BP) is a two-dimensional (2D) nanomaterial with high charge-carrier mobility, a tunable direct bandgap, and a unique in-plane anisotropic structure; however, the easiness of BP oxidation into P xO y species in ambient conditions largely limits its applications. In this study, modified cisplatin-Pt-NO3 [Pt(NH3)2(NO3)2] is used for surface coordination with BP nanosheets to generate Pt@BP, which maintains the surface morphology and properties of BP nanosheets for more than 24 h in ambient conditions. In addition, Pt@BP interacts with DNA both in vitro and in cell. Pt@BP shows a good cellular uptake rate and significantly increases the drug sensitivity of cisplatin-resistant cancer cell lines (A2780 and HepG2) compared with unmodified cisplatin. Our study is the first attempt to stabilize bare BP with cationic cisplatin species, and the generated Pt@BP could be used for potential synergistic photothermal/chemotherapy of cisplatin-resistant cancer.
Collapse
Affiliation(s)
- Jianing Zhang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Yue Ma
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Kuan Hu
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Yuan Feng
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Si Chen
- SZU-NUS Collaborative Innovation Center for Optoelectronic Science and Technology and Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province College of Optoelectronic Engineering , Shenzhen University , Shenzhen 518060 , China
| | - Xiaoyang Yang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Jacky Fong-Chuen Loo
- Department of Biomedical Engineering , The Chinese University of Hong Kong , Hong Kong , SAR 999077 , China
| | - Han Zhang
- SZU-NUS Collaborative Innovation Center for Optoelectronic Science and Technology and Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province College of Optoelectronic Engineering , Shenzhen University , Shenzhen 518060 , China
| | - Feng Yin
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| | - Zigang Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology , Peking University Shenzhen Graduate School , Shenzhen 518055 , China
| |
Collapse
|
11
|
Ghosh S. Cisplatin: The first metal based anticancer drug. Bioorg Chem 2019; 88:102925. [PMID: 31003078 DOI: 10.1016/j.bioorg.2019.102925] [Citation(s) in RCA: 1065] [Impact Index Per Article: 177.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 03/30/2019] [Accepted: 04/10/2019] [Indexed: 12/17/2022]
Abstract
Cisplatin or (SP-4-2)-diamminedichloridoplatinum(II) is one of the most potential and widely used drugs for the treatment of various solid cancers such as testicular, ovarian, head and neck, bladder, lung, cervical cancer, melanoma, lymphomas and several others. Cisplatin exerts anticancer activity via multiple mechanisms but its most acceptable mechanism involves generation of DNA lesions by interacting with purine bases on DNA followed by activation of several signal transduction pathways which finally lead to apoptosis. However, side effects and drug resistance are the two inherent challenges of cisplatin which limit its application and effectiveness. Reduction of drug accumulation inside cancer cells, inactivation of drug by reacting with glutathione and metallothioneins and faster repairing of DNA lesions are responsible for cisplatin resistance. To minimize cisplatin side effects and resistance, combination therapies are used and have proven more effective to defect cancers. This article highlights a systematic description on cisplatin which includes a brief history, synthesis, action mechanism, resistance, uses, side effects and modulation of side effects. It also briefly describes development of platinum drugs from very small cisplatin complex to very large next generation nanocarriers conjugated platinum complexes.
Collapse
Affiliation(s)
- Sumit Ghosh
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India.
| |
Collapse
|
12
|
Reilly NM, Yard BD, Pittman DL. Homologous Recombination-Mediated DNA Repair and Implications for Clinical Treatment of Repair Defective Cancers. Methods Mol Biol 2019; 1999:3-29. [PMID: 31127567 DOI: 10.1007/978-1-4939-9500-4_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Double-strand DNA breaks (DSBs) are generated by ionizing radiation and as intermediates during the processing of DNA, such as repair of interstrand cross-links and collapsed replication forks. These potentially deleterious DSBs are repaired primarily by the homologous recombination (HR) and nonhomologous end joining (NHEJ) DNA repair pathways. HR utilizes a homologous template to accurately restore damaged DNA, whereas NHEJ utilizes microhomology to join breaks in close proximity. The pathway available for DSB repair is dependent upon the cell cycle stage; for example, HR primarily functions during the S/G2 stages while NHEJ can repair DSBs at any cell cycle stage. Posttranslational modifications (PTMs) promote activity of specific pathways and subpathways through enzyme activation and precisely timed protein recruitment and degradation. This chapter provides an overview of PTMs occurring during DSB repair. In addition, clinical phenotypes associated with HR-defective cancers, such as mutational signatures used to predict response to poly(ADP-ribose) polymerase inhibitors, are discussed. Understanding these processes will provide insight into mechanisms of genome maintenance and likely identify targets and new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Nicole M Reilly
- Fondazione Piemontese per la Ricerca sul Cancro ONLUS, Candiolo, Italy
| | - Brian D Yard
- Department of Translational Hematology and Oncology Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Douglas L Pittman
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
13
|
Bjelogrlić S, Todorović TR, Cvijetić I, Rodić MV, Vujčić M, Marković S, Araškov J, Janović B, Emhemmed F, Muller CD, Filipović NR. A novel binuclear hydrazone-based Cd(II) complex is a strong pro-apoptotic inducer with significant activity against 2D and 3D pancreatic cancer stem cells. J Inorg Biochem 2018; 190:45-66. [PMID: 30352315 DOI: 10.1016/j.jinorgbio.2018.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/16/2018] [Accepted: 10/03/2018] [Indexed: 11/29/2022]
Abstract
A novel binuclear Cd complex (1) with hydrazone-based ligand was prepared and characterized by spectroscopy and single crystal X-ray diffraction techniques. Complex 1 reveals a strong pro-apoptotic activity in both human, mammary adenocarcinoma cells (MCF-7) and pancreatic AsPC-1 cancer stem cells (CSCs). While apoptosis undergoes mostly caspase-independent, 1 stimulates the activation of intrinsic pathway with noteworthy down regulation of caspase-8 activity in respect to non-treated controls. Distribution of cells over mitotic division indicates that 1 caused DNA damage in both cell lines, which is confirmed in DNA interaction studies. Compared to 1, cisplatin (CDDP) does not achieve cell death in 2D cultured AsPC-1 cells, while induces different pattern of cell cycle changes and caspase activation in 2D cultured MCF-7 cells, implying that these two compounds do not share similar mechanism of action. Additionally, 1 acts as a powerful inducer of mitochondrial superoxide production with dissipated trans-membrane potential in the majority of the treated cells already after 6 h of incubation. On 3D tumors, 1 displays a superior activity against CSC model, and at 100 μM induces disintegration of spheroids within 2 days of incubation. Fluorescence spectroscopy, along with molecular docking show that compound 1 binds to the minor groove of DNA. Compound 1 binds to the human serum albumin (HSA) showing that the HSA can effectively transport and store 1 in the human body. Thus, our current study strongly supports further investigations on antitumor activity of 1 as a drug candidate for the treatment of highly resistant pancreatic cancer.
Collapse
Affiliation(s)
- Snežana Bjelogrlić
- National Cancer Research Center of Serbia, Pasterova 14, Belgrade, Serbia; Institut Pluridisciplinaire Hubert Curien, UMR 7178 CNRS Université de Strasbourg, 67401 Illkirch, France
| | - Tamara R Todorović
- Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, Belgrade, Serbia
| | - Ilija Cvijetić
- Innovation Center of the Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, Belgrade, Serbia
| | - Marko V Rodić
- Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 3, Novi Sad, Serbia
| | - Miroslava Vujčić
- Institute of Chemistry, Technology and Metallurgy, University of Belgrade, Njegoševa 12, Belgrade, Serbia
| | - Sanja Marković
- Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, Belgrade, Serbia
| | - Jovana Araškov
- Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, Belgrade, Serbia
| | - Barbara Janović
- Institute of Chemistry, Technology and Metallurgy, University of Belgrade, Njegoševa 12, Belgrade, Serbia
| | - Fathi Emhemmed
- Institut Pluridisciplinaire Hubert Curien, UMR 7178 CNRS Université de Strasbourg, 67401 Illkirch, France
| | - Christian D Muller
- Institut Pluridisciplinaire Hubert Curien, UMR 7178 CNRS Université de Strasbourg, 67401 Illkirch, France
| | - Nenad R Filipović
- Faculty of Agriculture, University of Belgrade, Nemanjina 6, Belgrade, Serbia.
| |
Collapse
|
14
|
Jakubowski M, Łakomska I, Sitkowski J, Wiśniewska J. Dicarboxylato platinum(ii) complexes containing dimethyl sulfoxide and triazolopyrimidine as potential anticancer agents: synthesis, structural and biological studies in solution. NEW J CHEM 2018. [DOI: 10.1039/c8nj01199k] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
N,O,S-donors platinum(ii) complexes were synthesized and well characterized. We demonstrate that modification of coordination sphere by insertion of dmso molecule and bulky triazolopyrimidine ligand is good direction for the design effective less toxic platinum(ii) complexes.
Collapse
Affiliation(s)
- Mateusz Jakubowski
- Faculty of Chemistry
- Nicolaus Copernicus University in Toruń
- 87-100 Toruń
- Poland
| | - Iwona Łakomska
- Faculty of Chemistry
- Nicolaus Copernicus University in Toruń
- 87-100 Toruń
- Poland
| | - Jerzy Sitkowski
- National Institutes of Medicines
- 00-725 Warszawa
- Poland
- Institutes of Organic Chemistry
- Polish Academic of Science
| | - Joanna Wiśniewska
- Faculty of Chemistry
- Nicolaus Copernicus University in Toruń
- 87-100 Toruń
- Poland
| |
Collapse
|
15
|
Lopez-Martinez D, Liang CC, Cohn MA. Cellular response to DNA interstrand crosslinks: the Fanconi anemia pathway. Cell Mol Life Sci 2016; 73:3097-114. [PMID: 27094386 PMCID: PMC4951507 DOI: 10.1007/s00018-016-2218-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 04/04/2016] [Accepted: 04/05/2016] [Indexed: 12/22/2022]
Abstract
Interstrand crosslinks (ICLs) are a highly toxic form of DNA damage. ICLs can interfere with vital biological processes requiring separation of the two DNA strands, such as replication and transcription. If ICLs are left unrepaired, it can lead to mutations, chromosome breakage and mitotic catastrophe. The Fanconi anemia (FA) pathway can repair this type of DNA lesion, ensuring genomic stability. In this review, we will provide an overview of the cellular response to ICLs. First, we will discuss the origin of ICLs, comparing various endogenous and exogenous sources. Second, we will describe FA proteins as well as FA-related proteins involved in ICL repair, and the post-translational modifications that regulate these proteins. Finally, we will review the process of how ICLs are repaired by both replication-dependent and replication-independent mechanisms.
Collapse
Affiliation(s)
- David Lopez-Martinez
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Chih-Chao Liang
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Martin A Cohn
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
| |
Collapse
|
16
|
Li L, Guo W, Wu K, Zhao Y, Luo Q, Zhang Q, Liu J, Xiong S, Wang F. Identification of binding sites of cisplatin to human copper chaperone protein Cox17 by high-resolution FT-ICR-MS. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2016; 30 Suppl 1:168-172. [PMID: 27539433 DOI: 10.1002/rcm.7645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
RATIONALE Cox17 is a key copper chaperone protein responsible for delivery of cuprous ions to mitochondria and has been demonstrated to be involved in the anticancer action of cisplatin. However, the binding sites of the drug to the protein have not yet been directly identified. METHODS The recombinant protein apo-Cox172s-s , the functional state of Cox17 transferring Cu(I), was reacted with an excess of cisplatin to produce platinated Cox17 adducts, of which the platination sites were identified by high-resolution Fourier transform ion cyclotron tandem mass spectrometry (FT-ICR-MS/MS) through electron capture dissociation (ECD). RESULTS Primary FT-ICR-MS showed that mono-platinated Cox17 adducts were the main products, and top-down MS/MS results indicated that cisplatin bound to the Cys26 or Cys27 residue which is the binding site of cuprous ions in apo-Cox172s-s . CONCLUSIONS This is the first report for identification of the main binding sites of cisplatin to Cox17 by top-down high-resolution mass spectrometry, providing direct evidence for the competitive coordination with Cox17 of cisplatin and cuprous ions. These findings will also be helpful to understand further how Cox17 facilitates cisplatin accumulation in mitochondria, and how cisplatin disturbs the transportation of cuprous ions. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Lijie Li
- School of Chemical and Environmental Engineering, China University of Mining and Technology, Beijing, 100083, China
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Wei Guo
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Kui Wu
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yao Zhao
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Qun Luo
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Qingwu Zhang
- School of Chemical and Environmental Engineering, China University of Mining and Technology, Beijing, 100083, China
| | - Jianan Liu
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Shaoxiang Xiong
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Fuyi Wang
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
17
|
Nguyen TTT, Chua JKK, Seah KS, Koo SH, Yee JY, Yang EG, Lim KK, Pang SYW, Yuen A, Zhang L, Ang WH, Dymock B, Lee EJD, Chen ES. Predicting chemotherapeutic drug combinations through gene network profiling. Sci Rep 2016; 6:18658. [PMID: 26791325 PMCID: PMC4726371 DOI: 10.1038/srep18658] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 11/23/2015] [Indexed: 12/29/2022] Open
Abstract
Contemporary chemotherapeutic treatments incorporate the use of several agents in combination. However, selecting the most appropriate drugs for such therapy is not necessarily an easy or straightforward task. Here, we describe a targeted approach that can facilitate the reliable selection of chemotherapeutic drug combinations through the interrogation of drug-resistance gene networks. Our method employed single-cell eukaryote fission yeast (Schizosaccharomyces pombe) as a model of proliferating cells to delineate a drug resistance gene network using a synthetic lethality workflow. Using the results of a previous unbiased screen, we assessed the genetic overlap of doxorubicin with six other drugs harboring varied mechanisms of action. Using this fission yeast model, drug-specific ontological sub-classifications were identified through the computation of relative hypersensitivities. We found that human gastric adenocarcinoma cells can be sensitized to doxorubicin by concomitant treatment with cisplatin, an intra-DNA strand crosslinking agent, and suberoylanilide hydroxamic acid, a histone deacetylase inhibitor. Our findings point to the utility of fission yeast as a model and the differential targeting of a conserved gene interaction network when screening for successful chemotherapeutic drug combinations for human cells.
Collapse
Affiliation(s)
- Thi Thuy Trang Nguyen
- Department of Biochemistry, National University of Singapore, Singapore.,National University Health System (NUHS), Singapore
| | - Jacqueline Kia Kee Chua
- Department of Biochemistry, National University of Singapore, Singapore.,Department of Chemistry, Faculty of Science, National University of Singapore, Singapore
| | - Kwi Shan Seah
- Department of Biochemistry, National University of Singapore, Singapore.,National University Health System (NUHS), Singapore
| | - Seok Hwee Koo
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Changi General Hospital, Ministry of Health, Singapore
| | - Jie Yin Yee
- National University Health System (NUHS), Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Eugene Guorong Yang
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Kim Kiat Lim
- Department of Biochemistry, National University of Singapore, Singapore.,National University Health System (NUHS), Singapore
| | | | - Audrey Yuen
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore
| | - Louxin Zhang
- Department of Mathematics, Faculty of Science, National University of Singapore, Singapore
| | - Wee Han Ang
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore.,Department of Chemistry, Faculty of Science, National University of Singapore, Singapore
| | - Brian Dymock
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Edmund Jon Deoon Lee
- National University Health System (NUHS), Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ee Sin Chen
- Department of Biochemistry, National University of Singapore, Singapore.,National University Health System (NUHS), Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
18
|
Filipović NR, Bjelogrlić S, Portalone G, Pelliccia S, Silvestri R, Klisurić O, Senćanski M, Stanković D, Todorović TR, Muller CD. Pro-apoptotic and pro-differentiation induction by 8-quinolinecarboxaldehyde selenosemicarbazone and its Co(iii) complex in human cancer cell lines. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00199h] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The ligand initiated reprogramming of cancer stem cells phenotype in AsPC-1 cells. The complex digested plasmid DNA which might be the cause of its cytotoxic activity.
Collapse
Affiliation(s)
| | | | | | - Sveva Pelliccia
- Dipartimento di Chimica e Tecnologie del Farmaco
- Sapienza Universita di Roma
- I-00185 Roma
- Italy
| | - Romano Silvestri
- Dipartimento di Chimica e Tecnologie del Farmaco
- Sapienza Universita di Roma
- I-00185 Roma
- Italy
| | - Olivera Klisurić
- Department of Physics, Faculty of Sciences
- University of Novi Sad
- Novi Sad
- Serbia
| | - Milan Senćanski
- Center for Multidisciplinary Research
- Institute of Nuclear Sciences “Vinča”
- University of Belgrade
- Belgrade
- Serbia
| | - Dalibor Stanković
- Innovation Center of the Faculty of Chemistry
- University of Belgrade
- Belgrade
- Serbia
| | | | - Christian D. Muller
- Institut Pluridisciplinaire Hubert Curien
- UMR 7178 CNRS Université de Strasbourg
- 67401 Illkirch
- France
| |
Collapse
|
19
|
Filipović NR, Bjelogrlić S, Todorović TR, Blagojević VA, Muller CD, Marinković A, Vujčić M, Janović B, Malešević AS, Begović N, Senćanski M, Minić DM. Ni(ii) complex with bishydrazone ligand: synthesis, characterization, DNA binding studies and pro-apoptotic and pro-differentiation induction in human cancerous cell lines. RSC Adv 2016. [DOI: 10.1039/c6ra24604d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
A new Ni(ii) complex, [Ni(L)(H2O)] (1), with diethyl 3,3′-(2,2′-(1,1′-(pyridine-2,6-diyl)bis(ethan-1-yl-1-ylidene))bis(hydrazin-1-yl-2-ylidene))bis(3-oxopropanoate) ligand (H2L) was synthesized as a potential chemotherapeutic agent.
Collapse
Affiliation(s)
| | | | | | | | - Christian D. Muller
- Institut Pluridisciplinaire Hubert Curien
- UMR 7178 CNRS Université de Strasbourg
- 67401 Illkirch
- France
| | | | - Miroslava Vujčić
- Institute of Chemistry, Technology and Metallurgy
- University of Belgrade
- 11000 Belgrade
- Serbia
| | - Barbara Janović
- Institute of Chemistry, Technology and Metallurgy
- University of Belgrade
- 11000 Belgrade
- Serbia
| | | | - Nebojša Begović
- Institute of General and Physical Chemistry
- 11000 Belgrade
- Serbia
| | - Milan Senćanski
- Center for Multidisciplinary Research
- Institute of Nuclear Sciences ”Vinča”
- University of Belgrade
- 11000 Belgrade
- Serbia
| | - Dragica M. Minić
- Faculty of Physical Chemistry
- University of Belgrade
- 11000 Belgrade
- Serbia
| |
Collapse
|
20
|
Bergamo A, Sava G. Linking the future of anticancer metal-complexes to the therapy of tumour metastases. Chem Soc Rev 2015; 44:8818-35. [DOI: 10.1039/c5cs00134j] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cancer chemotherapy is almost always applied to patients with one or more diagnosed metastases and is expected to impact these lesions, thus providing significant benefits to the patient.
Collapse
Affiliation(s)
| | - Gianni Sava
- Callerio Foundation Onlus
- 34127 Trieste
- Italy
- Department of Life Sciences
- University of Trieste
| |
Collapse
|
21
|
Lando DY, Chang CL, Fridman AS, Grigoryan IE, Galyuk EN, Hsueh YW, Hu CK. Comparative thermal and thermodynamic study of DNA chemically modified with antitumor drug cisplatin and its inactive analog transplatin. J Inorg Biochem 2014; 137:85-93. [PMID: 24831492 DOI: 10.1016/j.jinorgbio.2014.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 04/14/2014] [Accepted: 04/15/2014] [Indexed: 12/11/2022]
Abstract
Antitumor activity of cisplatin is exerted by covalent binding to DNA. For comparison, studies of cisplatin-DNA complexes often employ the very similar but inactive transplatin. In this work, thermal and thermodynamic properties of DNA complexes with these compounds were studied using differential scanning calorimetry (DSC) and computer modeling. DSC demonstrates that cisplatin decreases thermal stability (melting temperature, Tm) of long DNA, and transplatin increases it. At the same time, both compounds decrease the enthalpy and entropy of the helix-coil transition, and the impact of transplatin is much higher. From Pt/nucleotide molar ratio rb=0.001, both compounds destroy the fine structure of DSC profile and increase the temperature melting range (ΔT). For cisplatin and transplatin, the dependences δTm vs rb differ in sign, while δΔT vs rb are positive for both compounds. The change in the parameter δΔT vs rb demonstrates the GC specificity in the location of DNA distortions. Our experimental results and calculations show that 1) in contrast to [Pt(dien)Cl]Cl, monofunctional adducts formed by transplatin decrease the thermal stability of long DNA at [Na(+)]>30mM; 2) interstrand crosslinks of cisplatin and transplatin only slightly increase Tm; 3) the difference in thermal stability of DNA complexes with cisplatin vs DNA complexes with transplatin mainly arises from the different thermodynamic properties of their intrastrand crosslinks. This type of crosslink appears to be responsible for the antitumor activity of cisplatin. At any [Na(+)] from interval 10-210mM, cisplatin and transplatin intrastrand crosslinks give rise to destabilization and stabilization, respectively.
Collapse
Affiliation(s)
- Dmitri Y Lando
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, 220141 Minsk, Belarus.
| | - Chun-Ling Chang
- Institute of Physics, Academia Sinica, Nankang, Taipei 11529, Taiwan
| | - Alexander S Fridman
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, 220141 Minsk, Belarus
| | | | - Elena N Galyuk
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, 220141 Minsk, Belarus
| | - Ya-Wei Hsueh
- Department of Physics, National Central University, Chungli 32001, Taiwan
| | - Chin-Kun Hu
- Institute of Physics, Academia Sinica, Nankang, Taipei 11529, Taiwan.
| |
Collapse
|
22
|
Abstract
Platinum-based chemotherapeutic drugs such as cisplatin, carboplatin and oxaliplatin are widely applied for the treatment of various types of tumours. Over the last few decades, a large variety of Pt(II) and Pt(IV) complexes have been developed to improve the applicability in a wider spectrum of cancers, increase their therapeutic window and reduce the dose-limiting side effects. Photodynamic therapy (PDT), which is the administration of a photosensitiser followed by visible light activation, is a promising route to avoid damage to healthy cells and the surrounding tissue. Transition metal complexes as photochemotherapeutic agents are an attractive option for further development in the field of photoactivated chemotherapy (PACT). These complexes exhibit different numbers and types of excited states which are easily accessible upon light irradiation, subsequently giving rise to the formation of various photoproducts that can enable a distinct mode of action. Platinum-diazido complexes are promising candidates for PACT due to the low cytotoxicity when irradiated with visible light. This review summarises the mode of action of current platinum anticancer drugs with cisplatin as a lead example and the development of non-conventional Pt(II) complexes. Background information regarding PDT the photophysical and photochemical properties of metal complexes is provided, as well as notable examples of photoactivated metal complexes with biological activity. Particular emphasis is placed on recent developments on platinum photoactivated drugs.
Collapse
|
23
|
Johnson D, Hastwell PW, Walmsley RM. The involvement of WT1 in the regulation of GADD45a in response to genotoxic stress. Mutagenesis 2013; 28:393-9. [PMID: 23476008 DOI: 10.1093/mutage/get015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Expression of the human GADD45a gene is increased in TK6 cells exposed to mutagens, clastogens and aneugens. It is known to be regulated through both p53-dependent and p53-independent pathways and WT1 has been implicated in both cases. This article reports an investigation into the effect that mutations in the WT1 and p53 response elements of the gene have on GADD45a expression. This was conducted in both p53 wild-type (TK6) and mutant (WI-L2-NS) human B lymphoblastoid cell lines. Gene expression was monitored using a GADD45a-green fluorescent protein reporter assay. Mutant cell lines were exposed to the mechanistically diverse genotoxins methyl methanesulphonate, cisplatin and mitomycin C (direct acting), hydroxyurea, aphidicolin and 5'fluorouracil (inhibitors of nucleotide/DNA synthesis) and benomyl (aneugen). In all cases, the induction of the reporter was reduced in the mutants compared with wild-type. These results provide experimental evidence for the implied role of WT1 in both p53-dependent and p53-independent pathways of GADD45a regulation and further insight into the mechanism of GADD45a induction by genotoxins.
Collapse
Affiliation(s)
- Donna Johnson
- Harper Adams University College, Newport, Shropshire TF10 8NB, UK
| | | | | |
Collapse
|
24
|
Lando DY, Galyuk EN, Chang CL, Hu CK. Temporal behavior of DNA thermal stability in the presence of platinum compounds. Role of monofunctional and bifunctional adducts. J Inorg Biochem 2012; 117:164-70. [DOI: 10.1016/j.jinorgbio.2012.08.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 08/24/2012] [Accepted: 08/28/2012] [Indexed: 10/27/2022]
|
25
|
Mowaka S, Ziehe M, Mohamed D, Hochkirch U, Thomale J, Linscheid MW. Structures of oxaliplatin-oligonucleotide adducts from DNA. JOURNAL OF MASS SPECTROMETRY : JMS 2012; 47:1282-1293. [PMID: 23019159 DOI: 10.1002/jms.3080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Oxaliplatin, [(1R,2R)-cyclohexane-1,2-diamine](ethanedioato-O,O')platinum(II) shows a great efficiency against colorectal cancer. Although the mode of action of oxaliplatin is not yet understood, it is commonly accepted that binding of oxaliplatin to DNA prevents DNA synthesis and alters protein to DNA binding. In order to elucidate the modified DNA-protein interaction and thus to understand the mechanisms leading to cellular misinterpretation of DNA information and apoptosis, we have identified the preferential binding sites and the dynamics of the oxaliplatin-DNA intrastrand and interstrand adducts at the oligomer level using high-performance liquid chromatography/electrospray ionization-tandem mass spectrometry (HPLC/ESI-MS/MS) and HPLC/inductively coupled plasma-MS for quantitative studies. We used a combination of benzonase, alkaline phosphatase and Nuclease S1 for digestion. This digestion procedure allows the study of platinated oligomeric nucleotides and more complex interstrand adducts. The digestion products were mostly chromatographically separated and characterized using HPLC/ESI-ion trap MS/MS experiments. We could show that the adducts to guanine and adenine are quite dynamic; that is, the ratios are changing for several days. In addition, the resulting adducts provide evidence for the action of the digesting enzymes and indicate that the adduct spectrum at the oligomeric level is different to that at the commonly studies dinucleotide level.
Collapse
Affiliation(s)
- Shereen Mowaka
- Humboldt-Universitaet zu Berlin, Department of Chemistry, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Short-term therapeutic effects of transcatheter arterial chemoembolization using miriplatin–lipiodol suspension for hepatocellular carcinoma. Jpn J Radiol 2012; 30:735-42. [DOI: 10.1007/s11604-012-0116-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 08/06/2012] [Indexed: 01/13/2023]
|
27
|
|
28
|
|
29
|
Li H, Zhao Y, Phillips HIA, Qi Y, Lin TY, Sadler PJ, O’Connor PB. Mass spectrometry evidence for cisplatin as a protein cross-linking reagent. Anal Chem 2011; 83:5369-76. [PMID: 21591778 PMCID: PMC3131505 DOI: 10.1021/ac200861k] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cisplatin is a potent anticancer drug, which functions by cross-linking adjacent DNA guanine residues. However within 1 day of injection, 65-98% of the platinum in the blood plasma is protein-bound. It is generally accepted that cisplatin binds to methionine and histidine residues, but what is often underappreciated is that platinum from cisplatin has a 2+ charge and can form up to four bonds. Thus, it has the potential to function as a cross-linker. In this report, the cross-linking ability of cisplatin is demonstrated by Fourier transform ion cyclotron resonance (FTICR) mass spectrometry (MS) with the use of standard peptides, the 16.8 kDa protein calmodulin (CaM), but was unsuccessful for the 64 kDa protein hemoglobin. The high resolution and mass accuracy of FTICR MS along with the high degree of fragmentation of large peptides afforded by collisionally activated dissociation (CAD) and electron capture dissociation (ECD) are shown to be a valuable means of characterizing cross-linking sites. Cisplatin is different from current cross-linking reagents by targeting new functional groups, thioethers, and imidazoles groups, which provides complementarity with existing cross-linkers. In addition, platinum(II) inherently has two positive charges which enhance the detection of cross-linked products. Higher charge states not only promote the detection of cross-linking products with less purification but result in more comprehensive MS/MS fragmentation and can assist in the assignment of modification sites. Moreover, the unique isotopic pattern of platinum flags cross-linking products and modification sites by mass spectrometry.
Collapse
Affiliation(s)
- Huilin Li
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Yao Zhao
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Hazel I. A. Phillips
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Yulin Qi
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Tzu-Yung Lin
- School of Engineering, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Peter J. Sadler
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Peter B. O’Connor
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| |
Collapse
|
30
|
Le Breton C, Hennion M, Arimondo PB, Hyrien O. Replication-fork stalling and processing at a single psoralen interstrand crosslink in Xenopus egg extracts. PLoS One 2011; 6:e18554. [PMID: 21525992 PMCID: PMC3078125 DOI: 10.1371/journal.pone.0018554] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 03/04/2011] [Indexed: 01/17/2023] Open
Abstract
Interstrand crosslink (ICL)-inducing agents block the separation of the two DNA strands. They prevent transcription and replication and are used in clinics for the treatment of cancer and skin diseases. Here, we have introduced a single psoralen ICL at a specific site in plasmid DNA using a triplex-forming-oligonucleotide (TFO)-psoralen conjugate and studied its repair in Xenopus egg extracts that support nuclear assembly and replication of plasmid DNA. Replication forks arriving from either side stalled at the psoralen ICL. In contrast to previous observations with other ICL-inducing agents, the leading strands advanced up to the lesion without any prior pausing. Subsequently, incisions were introduced on one parental strand on both sides of the ICL. These incisions could be detected whether one or both forks reached the ICL. Using small molecule inhibitors, we found that the ATR-Chk1 pathway, but not the ATM-Chk2 pathway, stimulated both the incision step and the subsequent processing of the broken replication intermediates. Our results highlight both similarities and differences in fork stalling and repair induced by psoralen and by other ICL-forming agents.
Collapse
Affiliation(s)
- Cyrille Le Breton
- Institut de Biologie de l'Ecole Normale Supérieure, CNRS UMR 8197-Inserm U1024, Paris, France
| | - Magali Hennion
- Institut de Biologie de l'Ecole Normale Supérieure, CNRS UMR 8197-Inserm U1024, Paris, France
- Museum National d'Histoire Naturelle UMR 7196 CNRS-INSERM U565, Paris, France
| | - Paola B. Arimondo
- Museum National d'Histoire Naturelle UMR 7196 CNRS-INSERM U565, Paris, France
| | - Olivier Hyrien
- Institut de Biologie de l'Ecole Normale Supérieure, CNRS UMR 8197-Inserm U1024, Paris, France
- * E-mail:
| |
Collapse
|
31
|
Helbig L, Damrot J, Hülsenbeck J, Köberle B, Brozovic A, Osmak M, Fiket Z, Kaina B, Fritz G. Late activation of stress-activated protein kinases/c-Jun N-terminal kinases triggered by cisplatin-induced DNA damage in repair-defective cells. J Biol Chem 2011; 286:12991-3001. [PMID: 21324906 DOI: 10.1074/jbc.m110.190645] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Although stress-activated protein kinases/c-Jun N-terminal kinases (SAPK/JNK) are rapidly activated by genotoxins, the role of DNA damage in this response is not well defined. Here we show that the SEK1/MKK4-mediated dual phosphorylation of SAPK/JNK (Thr-183/Tyr-185) correlates with the level of cisplatin-DNA adducts at late times (16-24 h) after drug treatment in both human and mouse cells. Transfection of platinated plasmid DNA also caused SAPK/JNK activation. A defect in transcription-coupled nucleotide excision repair resting on a mutation in Cockayne syndrome group B protein promoted the late SAPK/JNK activation following cisplatin exposure. Signaling to SAPK/JNK was accompanied by activation of Ataxia telangiectasia mutated- and Rad3-related kinase, replication protein A, and checkpoint kinases as well as by the formation of DNA double strand breaks (DSBs). Ionizing radiation-induced DSBs did not provoke SAPK/JNK activation, and inhibition of transcription also failed to provoke this response. Late activation of SAPK/JNK stimulated by cisplatin-induced DNA lesions was reduced in the absence of specific DNA repair proteins, such as xeroderma pigmentosum protein C, pointing to an essential function of individual repair factors in DNA damage signaling to SAPK/JNK. Collectively, the data indicate that late SAPK/JNK activation is triggered by non-repaired cisplatin adducts in transcribed genes and involves replication-associated events, DSBs, tyrosine kinases, Rho GTPases, and specific repair factors.
Collapse
Affiliation(s)
- Lars Helbig
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Johansson K, Järvliden J, Gogvadze V, Morgenstern R. Multiple roles of microsomal glutathione transferase 1 in cellular protection: a mechanistic study. Free Radic Biol Med 2010; 49:1638-45. [PMID: 20727966 DOI: 10.1016/j.freeradbiomed.2010.08.013] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 08/10/2010] [Accepted: 08/11/2010] [Indexed: 01/05/2023]
Abstract
The aim of this study was to investigate the involvement of membrane-bound microsomal glutathione transferase 1 (MGST1) in cellular resistance against oxidative stress as well as its mechanism of protection. MGST1 is ubiquitously expressed and predominantly located in the endoplasmic reticulum and outer mitochondrial membrane. Utilizing MCF7 cells overexpressing MGST1 we show significant protection against agents that are known to induce lipid peroxidation (e.g., cumene hydroperoxide and tert-butylhydroperoxide) and an end-product of lipid peroxidation (e.g., 4-hydroxy-2-nonenal). Furthermore, our results demonstrate that MGST1 protection can be enhanced by vitamin E when toxicity depends on oxidative stress, but not when direct alkylation is the dominant mechanism. Mitochondria in MGST1-overexpressing cells were shown to be protected from oxidative insult as measured by calcium loading capacity and respiration. MGST1 induces cellular resistance against cisplatin. Here we used vitamin E to elucidate whether oxidative stress caused by cisplatin is significant for cell toxicity. The results indicate that oxidative stress and induction of lipid peroxidation are not the most prominent toxic mechanism of cisplatin in our cell system. We thus conclude that MGST1 protects cells (and mitochondria) by both conjugation and glutathione peroxidase functions. A new protective mechanism against cisplatin is also indicated.
Collapse
Affiliation(s)
- Katarina Johansson
- Division of Biochemical Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | |
Collapse
|
33
|
Lenglet G, David-Cordonnier MH. DNA-Destabilizing Agents as an Alternative Approach for Targeting DNA: Mechanisms of Action and Cellular Consequences. J Nucleic Acids 2010; 2010. [PMID: 20725618 PMCID: PMC2915751 DOI: 10.4061/2010/290935] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 05/27/2010] [Accepted: 06/03/2010] [Indexed: 01/06/2023] Open
Abstract
DNA targeting drugs represent a large proportion of the actual anticancer drug pharmacopeia, both in terms of drug brands and prescription volumes. Small DNA-interacting molecules share the ability of certain proteins to change the DNA helix's overall organization and geometrical orientation via tilt, roll, twist, slip, and flip effects. In this ocean of DNA-interacting compounds, most stabilize both DNA strands and very few display helix-destabilizing properties. These types of DNA-destabilizing effect are observed with certain mono- or bis-intercalators and DNA alkylating agents (some of which have been or are being developed as cancer drugs). The formation of locally destabilized DNA portions could interfere with protein/DNA recognition and potentially affect several crucial cellular processes, such as DNA repair, replication, and transcription. The present paper describes the molecular basis of DNA destabilization, the cellular impact on protein recognition, and DNA repair processes and the latter's relationships with antitumour efficacy.
Collapse
Affiliation(s)
- Gaëlle Lenglet
- INSERM U-837, Jean-Pierre Aubert Research Center (JPARC), Team 4 Molecular and Cellular Targeting for Cancer Treatment, Institute for Research on Cancer of Lille (IRCL), Lille F-59045, France
| | | |
Collapse
|
34
|
Legerski RJ. Repair of DNA interstrand cross-links during S phase of the mammalian cell cycle. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:540-51. [PMID: 20658646 PMCID: PMC2911997 DOI: 10.1002/em.20566] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
DNA interstrand cross-linking (ICL) agents are widely used in anticancer chemotherapy regimens, yet our understanding of the DNA repair mechanisms by which these lesions are removed from the genome remains incomplete. This is at least in part due to the enormously complicated nature and variety of the biochemical pathways that operate on these complex lesions. In this review, we have focused specifically on the S-phase pathway of ICL repair in mammalian cells, which appears to be the major mechanism by which these lesions are removed in cycling cells. The various stages and components of this pathway are discussed, and a putative molecular model is presented. In addition, we propose an explanation as to how this pathway can lead to the observed high levels of sister chromatid exchanges known to be induced by ICLs.
Collapse
Affiliation(s)
- Randy J Legerski
- Department of Genetics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA.
| |
Collapse
|
35
|
Muniandy PA, Liu J, Majumdar A, Liu ST, Seidman MM. DNA interstrand crosslink repair in mammalian cells: step by step. Crit Rev Biochem Mol Biol 2010; 45:23-49. [PMID: 20039786 PMCID: PMC2824768 DOI: 10.3109/10409230903501819] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Interstrand DNA crosslinks (ICLs) are formed by natural products of metabolism and by chemotherapeutic reagents. Work in E. coli identified a two cycle repair scheme involving incisions on one strand on either side of the ICL (unhooking) producing a gapped intermediate with the incised oligonucleotide attached to the intact strand. The gap is filled by recombinational repair or lesion bypass synthesis. The remaining monoadduct is then removed by nucleotide excision repair (NER). Despite considerable effort, our understanding of each step in mammalian cells is still quite limited. In part this reflects the variety of crosslinking compounds, each with distinct structural features, used by different investigators. Also, multiple repair pathways are involved, variably operative during the cell cycle. G(1) phase repair requires functions from NER, although the mechanism of recognition has not been determined. Repair can be initiated by encounters with the transcriptional apparatus, or a replication fork. In the case of the latter, the reconstruction of a replication fork, stalled or broken by collision with an ICL, adds to the complexity of the repair process. The enzymology of unhooking, the identity of the lesion bypass polymerases required to fill the first repair gap, and the functions involved in the second repair cycle are all subjects of active inquiry. Here we will review current understanding of each step in ICL repair in mammalian cells.
Collapse
Affiliation(s)
- Parameswary A Muniandy
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
36
|
|
37
|
Zheng Y, Sanche L. Gold nanoparticles enhance DNA damage induced by anti-cancer drugs and radiation. Radiat Res 2009; 172:114-9. [PMID: 19580513 DOI: 10.1667/rr1689.1] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The chemotherapeutic agent cisplatin was chemically linked to pGEM-3Zf(-) plasmid DNA to produce a cisplatin-DNA complex, Gold nanoparticles, which bind electrostatically to pure DNA, could also be added to this complex. Dry films of pure plasmid DNA and DNA-cisplatin, DNA-gold nanoparticles and DNA-cisplatin-gold nanoparticles complexes were bombarded by 60 keV electrons. The yields of single- and double-strand breaks were measured as a function of exposure by electrophoresis. From a comparison of such yields from the different type of films, we found that the binding of only one gold nanoparticle to a plasmid-cisplatin complex containing 3197 base pairs increases by a factor of 3 the efficiency of the chemotherapeutic agent cisplatin to produce double-strand breaks in irradiated DNA. Furthermore, adding two cisplatin molecules and one gold nanoparticle to DNA enhances radiation-induced DSBs by a factor of 7.5. A number of phenomena could contribute to this huge enhancement, including the higher density of low-energy electrons and reactive species around the gold nanoparticles and the weakening of bonds adjacent to cisplatin in the DNA backbone. The addition of gold nanoparticles to cisplatin and other platinum agents may therefore provide interesting avenues of research to improve the treatment of cancer by concomitant chemoradiation.
Collapse
Affiliation(s)
- Yi Zheng
- Groupe en sciences des radiations, Faculté de médecine, Université de Sherbrooke, Sherbrooke, Québec, Canada J1H 5N4
| | | |
Collapse
|
38
|
Abdul AB, Abdelwahab SI, Bin Jalinas J, Al-Zubairi AS, Taha MME. Combination of zerumbone and cisplatin to treat cervical intraepithelial neoplasia in female BALB/c mice. Int J Gynecol Cancer 2009; 19:1004-10. [PMID: 19820360 DOI: 10.1111/igc.0b013e3181a83b51] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Recent in vitro and in vivo studies have demonstrated that zerumbone (ZER) possesses anticancer properties. The main objective of this study was to examine the effectiveness of the combination of ZER and cisplatin (CIS) to treat cervical intraepithelial neoplasia (CIN) in vivo. Microculture tetrazolium assay and immunohistochemistry of proliferating cellular nuclear antigen were used to study the antitumor effect of ZER. Prenatally exposed female BALB/c mice were used as a model. The progenies with CIN were injected peritoneally with isotonic sodium chloride solution (positive control), CIS, ZER, and a combination of both compounds. All treated and untreated mice were humanely killed, and serum and cervix were obtained for interleukin 6 analysis and histopathologic studies using hematoxylin and eosin staining, respectively. Zerumbone has revealed an antitumor effect on human cervical cancer cells and downregulates immunoexpression of proliferating cellular nuclear antigen (P < 0.05). In vivo study indicates that ZER at 16 mg/kg and CIS at 10 mg/kg have a regressing effect on CIN. The combination of ZER and CIS also showed similar effectiveness in regressing CIN. Our results indicate that the combination of ZER and CIS has modulated the serum level of interleukin 6 when compared with that in mice treated with isotonic sodium chloride solution (P < 0.05). The effectiveness of combining ZER and CIS could be further explored as a new therapeutic intervention of early precancerous stages of carcinogenesis before the invasive stage begins.
Collapse
Affiliation(s)
- Ahmad Bustamam Abdul
- UPM-MAKNA Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | | | | | | | | |
Collapse
|
39
|
Olive PL, Banáth JP. Kinetics of H2AX phosphorylation after exposure to cisplatin. CYTOMETRY PART B-CLINICAL CYTOMETRY 2009; 76:79-90. [PMID: 18727058 DOI: 10.1002/cyto.b.20450] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Cisplatin is a widely used cancer chemotherapeutic drug that causes DNA crosslinking and stimulates H2AX phosphorylation. Our goal was to assess the potential of gammaH2AX to help predict tumor response to cisplatin treatment. METHODS The kinetics of cisplatin-induced DNA interstrand crosslinks was measured using the alkaline comet assay and compared with gammaH2AX formation and clonogenic cell survival in several DNA repair proficient or deficient human and rodent cell lines. RESULTS The comet assay was effective in ranking cell lines according to their relative sensitivity to cisplatin based on reduced crosslink formation measured 6 h after drug exposure or by the failure of irs3 and UV41 cell lines to subsequently remove crosslinks. In comparison, the initial rate of phosphorylation of H2AX measured over the first 6 h after cisplatin treatment was unrelated to drug sensitivity or crosslinking proficiency. However, for proliferating cell cultures, the fraction of cells that retained gammaH2AX foci 24 h after cisplatin treatment was correlated with the fraction of cells that lost clonogenic potential (slope = 1.1, r(2) = 0.85). CONCLUSIONS H2AX phosphorylation occurs in response to replication fork damage caused by cisplatin induced DNA lesions, probably interstrand crosslinks. Although early kinetics of gammaH2AX formation was uninformative, retention of gammaH2AX foci 24 h after treatment was shown to be a useful indicator of cell response to killing by cisplatin. However, for gammaH2AX to serve as an indicator of cell viability after cisplatin treatment, cells must have the opportunity to transit S phase during the recovery period.
Collapse
Affiliation(s)
- Peggy L Olive
- Medical Biophysics Department, British Columbia Cancer Research Centre, Vancouver, BC, Canada.
| | | |
Collapse
|
40
|
Reedijk J. Platinum Anticancer Coordination Compounds: Study of DNA Binding Inspires New Drug Design. Eur J Inorg Chem 2009. [DOI: 10.1002/ejic.200900054] [Citation(s) in RCA: 293] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Jan Reedijk
- Leiden Institute of Chemistry, Leiden University, P. O. Box 9502, 2300 RA, Leiden, The Netherlands
| |
Collapse
|
41
|
Gao EJ, Yin HX, Zhu MC, Sun YG, Gu XF, Wu Q, Ren LX. Study on the interaction of a palladium complex with DNA. J STRUCT CHEM+ 2009. [DOI: 10.1007/s10947-008-0177-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
42
|
Kozelka J. Molecular origin of the sequence-dependent kinetics of reactions between cisplatin derivatives and DNA. Inorganica Chim Acta 2009. [DOI: 10.1016/j.ica.2008.04.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
43
|
Kurosawa A, Koyama H, Takayama S, Miki K, Ayusawa D, Fujii M, Iiizumi S, Adachi N. The requirement of Artemis in double-strand break repair depends on the type of DNA damage. DNA Cell Biol 2008; 27:55-61. [PMID: 17941805 DOI: 10.1089/dna.2007.0649] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Artemis is a recently identified factor involved in V(D)J recombination and nonhomologous end joining (NHEJ) of DNA double-strand break (DSB) repair. Here, we performed targeted disruption of the Artemis gene (ARTEMIS) in the human pre-B cell line Nalm-6. Unexpectedly, we found that cells lacking Artemis exhibit increased sensitivity to low doses, but not high doses, of ionizing radiation. We also show that ARTEMIS-deficient cells are hypersensitive to the topoisomerase II inhibitor etoposide, but to a much lesser extent than cells lacking DNA ligase IV, a critical component of NHEJ. Unlike DNA ligase IV-deficient cells, ARTEMIS-deficient cells were not hypersensitive to ICRF-193, a topoisomerase II inhibitor that does not stabilize topoisomerase II-DNA cleavable complexes. Collectively, our results suggest that Artemis only partially participates in the NHEJ pathway to repair DSBs in human somatic cells.
Collapse
Affiliation(s)
- Aya Kurosawa
- Graduate School of Integrated Science, Yokohama City University, Yokohama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
44
|
The influence of temperature on antiproliferative effects, cellular uptake and DNA platination of the clinically employed Pt(II)-drugs. J Inorg Biochem 2007; 102:629-35. [PMID: 18037490 DOI: 10.1016/j.jinorgbio.2007.10.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Revised: 08/22/2007] [Accepted: 10/12/2007] [Indexed: 11/22/2022]
Abstract
Cellular uptake of a drug is one of the most important factors influencing its pharmacodynamics and pharmacokinetics. Our laboratory has previously studied platinum uptake following cisplatin, carboplatin and oxaliplatin treatment at sub-lethal doses of selected tumour cell lines. Here we report on the influence of temperature on dose-dependent antiproliferative effects, cellular uptake and DNA platination of these platinum-based drugs tested on MCF-7 human mammary carcinoma cell line. Inductively coupled plasma-mass spectrometry (ICP-MS) technique has been chosen to perform Pt determinations on cells treated with drug concentrations similar with those usually found in vivo in human plasma. The high sensitivity and analytical rapidity of this technique made possible to carry out a very large amount of Pt determinations (about 300) necessary for this study. Hyperthermia (43 degrees C) proved a synergistic effect with cisplatin on cell growth inhibition, while only an additive effect was demonstrated for carboplatin and oxaliplatin. This behaviour might be explained by the higher DNA platination ratio between data at 43 and 37 degrees C of cisplatin with respect to those of carboplatin and oxaliplatin.
Collapse
|
45
|
Matlawska-Wasowska K, Rainczuk K, Kalinowska-Lis U, Osiecka R, Ochocki J. Genotoxicity of novel trans-platinum(II) complex with diethyl (pyridin-4-ylmethyl)phosphate in human non-small cell lung cancer cells A549. Chem Biol Interact 2007; 168:135-42. [PMID: 17499650 DOI: 10.1016/j.cbi.2007.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Revised: 04/02/2007] [Accepted: 04/03/2007] [Indexed: 10/23/2022]
Abstract
The dynamic development of metal-containing anticancer drugs has started since the discovery of cis-diamminedichloroplatinum(II). For many years it was believed that trans platinum(II) compounds were non-active as antitumour agents because trans-diamminedichloroplatinum is biologically inactive although it binds to DNA and also forms monoadducts and cross-links. In the present work the ability of a novel platinum(II) compound trans-[PtCl(2)(4-pmOpe)(2)] to induce DNA damage in human non-small cell lung cancer cells A549 was examined using the alkaline comet assay. The obtained results revealed that the novel trans platinum(II) complex induced DNA strand breaks, which were effectively repaired during 2h of post-incubation, and cross-links which remained unrepaired under these test conditions. Apart from that, the modified comet assay with incubation with proteinase K was used to verify the ability of trans-[PtCl(2)(4-pmOpe)(2)] and cis-DDP to form DNA-protein cross-links. It has been proved that only trans-[PtCl(2)(4-pmOpe)(2)] complex exhibits the ability to induce DNA-protein cross-links. The results suggest a different mechanism of action of this compound in comparison to cis-DDP. It seems that trans geometry and the presence of two diethyl (pyridin-4-ylmethyl)phosphates as non-leaving ligands can determine dissimilar properties of the adducts formed on DNA and the different mechanism of action of trans-[PtCl(2)(4-pmOpe)(2)] and in consequence the efficacy in killing cancer cells.
Collapse
Affiliation(s)
- Ksenia Matlawska-Wasowska
- Department of Cytogenetics and Plant Molecular Biology, University of Lodz, Banacha 12/16, Lodz, Poland.
| | | | | | | | | |
Collapse
|
46
|
Zorbas-Seifried S, Hartinger CG, Meelich K, Galanski MS, Keppler BK, Zorbas H. DNA Interactions of pH-Sensitive, Antitumor Bis(aminoalcohol)dichloroplatinum(II) Complexes,. Biochemistry 2006; 45:14817-25. [PMID: 17144675 DOI: 10.1021/bi061063i] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
(SP-4-2)-Bis(2-aminoethanol)dichloroplatinum(II) (KP1356) and (SP-4-2)-bis[(R)-(-)-2-aminobutanol)]dichloroplatinum(II) (KP1433) are promising cytotoxic agents capable of changing their chemical structure depending on the pH value. On the basis of this, they are supposed to be active only in or preferentially in hypoxic tumors with low pH. In this study, we investigated the kinetics of changes of the DNA secondary structure, of the DNA modification degree, and of the formation of interstrand cross-links caused by these complexes in comparison to the parental compound cis-diamminedichloroplatinum(II) (cisplatin). All examinations were performed at physiological pH 7.4 and at pH 6.0 mimicking the acidified environment of many tumor tissues. In general, cisplatin displayed a higher reactivity accompanied by more pronounced DNA compaction, untwisting, and formation of interstrand cross-links at both pH values. Additionally, it was shown for the first time that cisplatin generates interstrand cross-links faster at pH 6.0 than at 7.4. However, the difference between pH 7.4 and 6.0 was much larger for KP1356 and KP1433 than for cisplatin, since they were essentially nonreactive and induced almost no secondary structures at pH 7.4, as contrasted to cisplatin. Our data suggest that formed adducts, i.e., intra- and/or interstrand cross-links, may be the sole cause of the cytotoxicity of KP1356 and KP1433 at pH 6.0. The results of this study may stimulate and contribute to further improvement of these novel, specific cytotoxic drugs that are anticipated to exert their full power in the tumor while being reasonably inactive in normal tissue.
Collapse
|
47
|
Lehoczký P, McHugh PJ, Chovanec M. DNA interstrand cross-link repair in Saccharomyces cerevisiae. FEMS Microbiol Rev 2006; 31:109-33. [PMID: 17096663 DOI: 10.1111/j.1574-6976.2006.00046.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
DNA interstrand cross-links (ICL) present a formidable challenge to the cellular DNA repair apparatus. For Escherichia coli, a pathway which combines nucleotide excision repair (NER) and homologous recombination repair (HRR) to eliminate ICL has been characterized in detail, both genetically and biochemically. Mechanisms of ICL repair in eukaryotes have proved more difficult to define, primarily as a result of the fact that several pathways appear compete for ICL repair intermediates, and also because these competing activities are regulated in the cell cycle. The budding yeast Saccharomyces cerevisiae has proven a powerful tool for dissecting ICL repair. Important roles for NER, HRR and postreplication/translesion synthesis pathways have all been identified. Here we review, with reference to similarities and differences in higher eukaryotes, what has been discovered to date concerning ICL repair in this simple eukaryote.
Collapse
Affiliation(s)
- Peter Lehoczký
- Department of Molecular Genetics, Cancer Research Institute, Bratislava, Slovak Republic
| | | | | |
Collapse
|
48
|
Niedernhofer LJ, Odijk H, Budzowska M, van Drunen E, Maas A, Theil AF, de Wit J, Jaspers NGJ, Beverloo HB, Hoeijmakers JHJ, Kanaar R. The structure-specific endonuclease Ercc1-Xpf is required to resolve DNA interstrand cross-link-induced double-strand breaks. Mol Cell Biol 2004; 24:5776-87. [PMID: 15199134 PMCID: PMC480908 DOI: 10.1128/mcb.24.13.5776-5787.2004] [Citation(s) in RCA: 391] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2003] [Revised: 01/07/2004] [Accepted: 04/06/2004] [Indexed: 11/20/2022] Open
Abstract
Interstrand cross-links (ICLs) are an extremely toxic class of DNA damage incurred during normal metabolism or cancer chemotherapy. ICLs covalently tether both strands of duplex DNA, preventing the strand unwinding that is essential for polymerase access. The mechanism of ICL repair in mammalian cells is poorly understood. However, genetic data implicate the Ercc1-Xpf endonuclease and proteins required for homologous recombination-mediated double-strand break (DSB) repair. To examine the role of Ercc1-Xpf in ICL repair, we monitored the phosphorylation of histone variant H2AX (gamma-H2AX). The phosphoprotein accumulates at DSBs, forming foci that can be detected by immunostaining. Treatment of wild-type cells with mitomycin C (MMC) induced gamma-H2AX foci and increased the amount of DSBs detected by pulsed-field gel electrophoresis. Surprisingly, gamma-H2AX foci were also induced in Ercc1(-/-) cells by MMC treatment. Thus, DSBs occur after cross-link damage via an Ercc1-independent mechanism. Instead, ICL-induced DSB formation required cell cycle progression into S phase, suggesting that DSBs are an intermediate of ICL repair that form during DNA replication. In Ercc1(-/-) cells, MMC-induced gamma-H2AX foci persisted at least 48 h longer than in wild-type cells, demonstrating that Ercc1 is required for the resolution of cross-link-induced DSBs. MMC triggered sister chromatid exchanges in wild-type cells but chromatid fusions in Ercc1(-/-) and Xpf mutant cells, indicating that in their absence, repair of DSBs is prevented. Collectively, these data support a role for Ercc1-Xpf in processing ICL-induced DSBs so that these cytotoxic intermediates can be repaired by homologous recombination.
Collapse
Affiliation(s)
- Laura J Niedernhofer
- Department of Cell Biology and Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ghezzi A, Aceto M, Cassino C, Gabano E, Osella D. Uptake of antitumor platinum(II)-complexes by cancer cells, assayed by inductively coupled plasma mass spectrometry (ICP-MS). J Inorg Biochem 2004; 98:73-8. [PMID: 14659635 DOI: 10.1016/j.jinorgbio.2003.08.014] [Citation(s) in RCA: 196] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
A systematic study on intracellular Pt uptake and Pt accumulation ratio in breast cancer MCF-7 cell line has been performed on a number of Pt(II)-complexes, namely cisplatin, carboplatin and oxaliplatin, clinically employed as antitumor drugs, trans- and cis-[Pt(Cl)2(pyridine)2] and cis-[Pt(Cl)2(pyridine)(5-SO3H-isoquinoline)] complexes, previously investigated also as potential telomerase inhibitors. In particular, long incubation times have been chosen in order to understand the fate of the complexes in the cells. For this purpose, sub-acute drug concentrations must be employed and, therefore, a very sensitive method of analysis like as inductively coupled plasma mass spectrometry (ICP-MS) superior to the widely employed atomic absorption spectroscopy (AAS) has been adopted. Any relationships among uptake/accumulation and several parameters such as drug structure, lipophilicity, drug concentration and incubation time have been sought and analyzed: the bulk of data point for a passive diffusion mechanism through the cell membrane.
Collapse
Affiliation(s)
- AnnaRita Ghezzi
- Dipartimento di Scienze e Tecnologie Avanzate, Università del Piemonte Orientale A. Avogadro, Corso Borsalino 54, I-15100 Alessandria, Italy
| | | | | | | | | |
Collapse
|
50
|
Fourrier L, Brooks P, Malinge JM. Binding discrimination of MutS to a set of lesions and compound lesions (base damage and mismatch) reveals its potential role as a cisplatin-damaged DNA sensing protein. J Biol Chem 2003; 278:21267-75. [PMID: 12654906 DOI: 10.1074/jbc.m301390200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The DNA mismatch repair (MMR) system plays a critical role in sensitizing both prokaryotic and eukaryotic cells to the clinically potent anticancer drug cisplatin. It is thought to mediate cytotoxicity through recognition of cisplatin DNA lesions. This drug generates a range of lesions that may also give rise to compound lesions resulting from the misincorporation of a base during translesion synthesis. Using gel mobility shift competition assays and surface plasmon resonance, we have analyzed the interaction of Escherichia coli MutS protein with site-specifically modified DNA oligonucleotides containing each of the four cisplatin cross-links or a set of compound lesions. The major 1,2-d(GpG) cisplatin intrastrand cross-link was recognized with only a 1.5-fold specificity, whereas a 47-fold specificity was found with a natural G/T containing DNA substrate. The rate of association, kon, for binding to the 1,2-d(GpG) adduct was 3.1 x 104 m-1 s-1 and the specificity of binding was essentially dependent on koff. DNA duplexes containing a single 1,2-d(ApG), 1,3-d(GpCpG) adduct, and an interstrand cross-link of cisplatin were not preferentially recognized. Among 12 DNA substrates, each containing a different cisplatin compound lesion derived from replicative misincorporation of one base opposite either of the 1,2-intrastrand adducts, 10 were specifically recognized including those that are more likely formed in vivo based on cisplatin mutation spectra. Moreover, among these lesions, two compound lesions formed when an adenine was misincorporated opposite a 1,2-d(GpG) adduct were not substrates for the MutY-dependent mismatch repair pathway. The ability of MutS to sense differentially various platinated DNA substrates suggests that cisplatin compound lesions formed during misincorporation of a base opposite either adducted base of both 1,2-intrastrand cross-links are more plausible critical lesions for MMR-mediated cisplatin cytotoxicity.
Collapse
Affiliation(s)
- Laurence Fourrier
- Centre de Biophysique Moléculaire, CNRS, Rue Charles Sadron, 45071 Orléans Cedex 02, France
| | | | | |
Collapse
|