1
|
Petroni M, La Monica V, Fabretti F, Augusto M, Battaglini D, Polonara F, Di Giulio S, Giannini G. The Multiple Faces of the MRN Complex: Roles in Medulloblastoma and Beyond. Cancers (Basel) 2023; 15:3599. [PMID: 37509263 PMCID: PMC10377613 DOI: 10.3390/cancers15143599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/09/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Hypomorphic mutations in MRN complex genes are frequently found in cancer, supporting their role as oncosuppressors. However, unlike canonical oncosuppressors, MRN proteins are often overexpressed in tumor tissues, where they actively work to counteract DSBs induced by both oncogene-dependent RS and radio-chemotherapy. Moreover, at the same time, MRN genes are also essential genes, since the constitutive KO of each component leads to embryonic lethality. Therefore, even though it is paradoxical, MRN genes may work as oncosuppressive, oncopromoting, and essential genes. In this review, we discussed how alterations in the MRN complex impact the physiopathology of cancer, in light of our recent discoveries on the gene-dosage-dependent effect of NBS1 in Medulloblastoma. These updates aim to understand whether MRN complex can be realistically used as a prognostic/predictive marker and/or as a therapeutic target for the treatment of cancer patients in the future.
Collapse
Affiliation(s)
- Marialaura Petroni
- Department of Molecular Medicine, University La Sapienza, 00161 Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, 00161 Rome, Italy
| | - Veronica La Monica
- Department of Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - Francesca Fabretti
- Department of Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - Mariaconcetta Augusto
- Department of Molecular Medicine, University La Sapienza, 00161 Rome, Italy
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Damiana Battaglini
- Department of Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - Francesca Polonara
- Department of Molecular Medicine, University La Sapienza, 00161 Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, 00161 Rome, Italy
| | - Stefano Di Giulio
- Department of Molecular Medicine, University La Sapienza, 00161 Rome, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, University La Sapienza, 00161 Rome, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, 00161 Rome, Italy
| |
Collapse
|
2
|
Targeting DNA Damage Response and Repair to Enhance Therapeutic Index in Cisplatin-Based Cancer Treatment. Int J Mol Sci 2021; 22:ijms22158199. [PMID: 34360968 PMCID: PMC8347825 DOI: 10.3390/ijms22158199] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Platinum-based chemotherapies, such as cisplatin, play a large role in cancer treatment. The development of resistance and treatment toxicity creates substantial barriers to disease control, yet. To enhance the therapeutic index of cisplatin-based chemotherapy, it is imperative to circumvent resistance and toxicity while optimizing tumor sensitization. One of the primary mechanisms by which cancer cells develop resistance to cisplatin is through upregulation of DNA repair pathways. In this review, we discuss the DNA damage response in the context of cisplatin-induced DNA damage. We describe the proteins involved in the pathways and their roles in resistance development. Common biomarkers for cisplatin resistance and their utilization to improve patient risk stratification and treatment personalization are addressed. Finally, we discuss some of the current treatments and future strategies to circumvent the development of cisplatin resistance.
Collapse
|
3
|
Alblihy A, Alabdullah ML, Ali R, Algethami M, Toss MS, Mongan NP, Rakha EA, Madhusudan S. Clinicopathological and Functional Evaluation Reveal NBS1 as a Predictor of Platinum Resistance in Epithelial Ovarian Cancers. Biomedicines 2021; 9:biomedicines9010056. [PMID: 33435622 PMCID: PMC7826685 DOI: 10.3390/biomedicines9010056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 01/05/2023] Open
Abstract
Platinum resistance seriously impacts on the survival outcomes of patients with ovarian cancers. Platinum-induced DNA damage is processed through DNA repair. NBS1 is a key DNA repair protein. Here, we evaluated the role of NBS1 in ovarian cancers. NBS1 expression was investigated in clinical cohorts (protein level (n = 331) and at the transcriptomic level (n = 1259)). Pre-clinically, sub-cellular localization of NBS1 at baseline and following cisplatin therapy was tested in platinum resistant (A2780cis, PEO4) and sensitive (A2780, PEO1) ovarian cancer cells. NBS1 was depleted and cisplatin sensitivity was investigated in A2780cis and PEO4 cells. Nuclear NBS1 overexpression was associated with platinum resistance (p = 0.0001). In univariate and multivariate analysis, nuclear NBS1 overexpression was associated with progression free survival (PFS) (p-values = 0.003 and 0.017, respectively) and overall survival (OS) (p-values = 0.035 and 0.009, respectively). NBS1 mRNA overexpression was linked with poor PFS (p = 0.011). Pre-clinically, following cisplatin treatment, we observed nuclear localization of NBS1 in A2780cis and PEO4 compared to A2780 and PEO1 cells. NBS1 depletion increased cisplatin cytotoxicity, which was associated with accumulation of double strand breaks (DSBs), S-phase cell cycle arrest, and increased apoptosis. NBS1 is a predictor of platinum sensitivity and could aid stratification of ovarian cancer therapy.
Collapse
Affiliation(s)
- Adel Alblihy
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham NG5 1PB, UK; (A.A.); (M.L.A.); (R.A.); (M.A.)
- Medical Center, King Fahad Security College (KFSC), Riyadh 11461, Saudi Arabia
| | - Muslim L. Alabdullah
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham NG5 1PB, UK; (A.A.); (M.L.A.); (R.A.); (M.A.)
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham NG5 1PB, UK; (M.S.T.); (E.A.R.)
| | - Reem Ali
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham NG5 1PB, UK; (A.A.); (M.L.A.); (R.A.); (M.A.)
| | - Mashael Algethami
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham NG5 1PB, UK; (A.A.); (M.L.A.); (R.A.); (M.A.)
| | - Michael S. Toss
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham NG5 1PB, UK; (M.S.T.); (E.A.R.)
| | - Nigel P. Mongan
- School Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham Biodiscovery Institute, Nottingham NG7 2RD, UK;
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Emad A. Rakha
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham NG5 1PB, UK; (M.S.T.); (E.A.R.)
| | - Srinivasan Madhusudan
- Translational Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham NG5 1PB, UK; (A.A.); (M.L.A.); (R.A.); (M.A.)
- Department of Oncology, Nottingham University Hospitals, Nottingham NG5 1PB, UK
- Correspondence:
| |
Collapse
|
4
|
McPherson MT, Holub AS, Husbands AY, Petreaca RC. Mutation Spectra of the MRN (MRE11, RAD50, NBS1/NBN) Break Sensor in Cancer Cells. Cancers (Basel) 2020; 12:cancers12123794. [PMID: 33339169 PMCID: PMC7765586 DOI: 10.3390/cancers12123794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/04/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary A DNA double strand break cuts a chromosome in two and is one of the most dangerous forms of DNA damage. Improper repair can lead to various chromosomal re-arrangements that have been detected in almost all cancer cells. A complex of three proteins (MRE11, RAD50, NBS1 or NBN) detects chromosome breaks and orchestrates repair processes. Mutations in these “break sensor” genes have been described in a multitude of cancers. Here, we provide a comprehensive analysis of reported mutations from data deposited on the Catalogue of Somatic Mutations in Cancer (COSMIC) archive. We also undertake an evolutionary analysis of these genes with the aim to understand whether these mutations preferentially accumulate in conserved residues. Interestingly, we find that mutations are overrepresented in evolutionarily conserved residues of RAD50 and NBS1/NBN but not MRE11. Abstract The MRN complex (MRE11, RAD50, NBS1/NBN) is a DNA double strand break sensor in eukaryotes. The complex directly participates in, or coordinates, several activities at the break such as DNA resection, activation of the DNA damage checkpoint, chromatin remodeling and recruitment of the repair machinery. Mutations in components of the MRN complex have been described in cancer cells for several decades. Using the Catalogue of Somatic Mutations in Cancer (COSMIC) database, we characterized all the reported MRN mutations. This analysis revealed several hotspot frameshift mutations in all three genes that introduce premature stop codons and truncate large regions of the C-termini. We also found through evolutionary analyses that COSMIC mutations are enriched in conserved residues of NBS1/NBN and RAD50 but not in MRE11. Given that all three genes are important to carcinogenesis, we propose these differential enrichment patterns may reflect a more severe pleiotropic role for MRE11.
Collapse
|
5
|
Shen B, Huang D, Ramsey AJ, Ig-Izevbekhai K, Zhang K, Lajud SA, O'Malley BW, Li D. PD-L1 and MRN synergy in platinum-based chemoresistance of head and neck squamous cell carcinoma. Br J Cancer 2020; 122:640-647. [PMID: 31853007 PMCID: PMC7054324 DOI: 10.1038/s41416-019-0697-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/19/2019] [Accepted: 12/05/2019] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND We have been investigating the molecular mechanisms of cisplatin-induced chemoresistance in head and neck squamous cell carcinoma (HNSCC). Based on our previous findings, the present study investigates how the Mre11, Rad50, and NBS1 (MRN) DNA repair complex interacts at the molecular level with the programmed cell death ligand 1 (PD-L1) in cisplatin-induced chemoresistance. METHODS Human HNSCC cell lines were used to determine the role played by PD-L1 in cisplatin resistance. Initial experiments investigated PD-L1 expression levels in cells exposed to cisplatin and whether PD-L1 interacts directly with the MRN complex. Finally, in vitro studies and in vivo experiments on BALB/c nu/nu mice were performed to determine whether interference of PD-L1 or NBS1 synthesis modulated cisplatin resistance. RESULTS Exposure to cisplatin resulted in PD-L1 being upregulated in the chemoresistant but not the chemosensitive cell line. Subsequent co-immunoprecipitation studies demonstrated that PD-L1 associates with NBS1. In addition, we found that the knockdown of either PD-L1 or NBS1 re-sensitised the chemoresistant cell line to cisplatin. Finally, but perhaps most importantly, synergy was observed when both PD-L1 and NBS1 were knocked down making the formerly chemoresistant strain highly cisplatin sensitive. CONCLUSIONS PD-L1 plays a pivotal role in cisplatin resistance in chemoresistant human HNSCC cell lines.
Collapse
Affiliation(s)
- Bin Shen
- Department of Otolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
- Department of Otolaryngology-Head and Neck Surgery, Shanghai General Hospital, Shanghai, China
| | - Dongyan Huang
- Department of Otolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
- Department of Otolaryngology-Head and Neck Surgery, PLA General Hospital, Beijing, China
| | - Andrew J Ramsey
- Department of Otolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Kevin Ig-Izevbekhai
- Department of Otolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Kevin Zhang
- Department of Otolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Shayanne A Lajud
- Department of Otolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
- Department of Otolaryngology-Head and Neck Surgery, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Bert W O'Malley
- Department of Otolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Daqing Li
- Department of Otolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
6
|
Kim YJ, Kim TW, Park SR, Kim HT, Ryu SY, Jung JY. Expression of the Mre11-Rad50-Nbs1 complex in cisplatin nephrotoxicity. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 40:12-17. [PMID: 26056972 DOI: 10.1016/j.etap.2015.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/22/2015] [Accepted: 04/28/2015] [Indexed: 06/04/2023]
Abstract
The aim of this study was to explore whether the Mre11, Rad50, and Nbs1 (MRN) complex is associated with DNA repair mechanisms in cisplatin-induced acute renal failure. Rats were randomly allocated into three groups: control, sacrificed 5 days (5D), and 10 days (10D) after 5mg/kg of cisplatin injection. The 5D group showed disrupted renal function together with enhanced MRN complex- and DNA repair-related protein expression. Meanwhile, in the 10D group, recovery from cisplatin-induced damage was accompanied by the reduced MRN expression, although the expression was still distinctive in proximal tubular cells and higher than the control group. Moreover, pretreatment with mirin, an MRN complex inhibitor, decreased cell viability and inhibited proliferating cell nuclear antigen expression in cisplatin-treated human embryonic kidney 293 cells. Taken together, cisplatin treatment could trigger the MRN complex expression in the kidney and inhibition of the complex might aggravate damage recovery processes.
Collapse
Affiliation(s)
- Young-Jung Kim
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon 305-764, Republic of Korea
| | - Tae-Won Kim
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon 305-764, Republic of Korea
| | - So-Ra Park
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon 305-764, Republic of Korea
| | - Hyun-Tae Kim
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon 305-764, Republic of Korea
| | - Si-Yun Ryu
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon 305-764, Republic of Korea
| | - Ju-Young Jung
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon 305-764, Republic of Korea.
| |
Collapse
|
7
|
Doherty R, Madhusudan S. DNA Repair Endonucleases: Physiological Roles and Potential as Drug Targets. ACTA ACUST UNITED AC 2015; 20:829-41. [PMID: 25877151 DOI: 10.1177/1087057115581581] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/22/2015] [Indexed: 12/15/2022]
Abstract
Genomic DNA is constantly exposed to endogenous and exogenous damaging agents. To overcome these damaging effects and maintain genomic stability, cells have robust coping mechanisms in place, including repair of the damaged DNA. There are a number of DNA repair pathways available to cells dependent on the type of damage induced. The removal of damaged DNA is essential to allow successful repair. Removal of DNA strands is achieved by nucleases. Exonucleases are those that progressively cut from DNA ends, and endonucleases make single incisions within strands of DNA. This review focuses on the group of endonucleases involved in DNA repair pathways, their mechanistic functions, roles in cancer development, and how targeting these enzymes is proving to be an exciting new strategy for personalized therapy in cancer.
Collapse
Affiliation(s)
- Rachel Doherty
- Laboratory of Molecular Oncology, Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals, Nottingham, UK
| | - Srinivasan Madhusudan
- Laboratory of Molecular Oncology, Academic Unit of Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals, Nottingham, UK
| |
Collapse
|
8
|
Lajud SA, Nagda DA, Yamashita T, Zheng J, Tanaka N, Abuzeid WM, Civantos A, Bezpalko O, O'Malley BW, Li D. Dual disruption of DNA repair and telomere maintenance for the treatment of head and neck cancer. Clin Cancer Res 2014; 20:6465-78. [PMID: 25324139 DOI: 10.1158/1078-0432.ccr-14-0176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Poly(ADP-ribose) polymerases (PARP) and the Mre11, Rad50, and Nbs1 (MRN) complex are key regulators of DNA repair, and have been recently shown to independently regulate telomere length. Sensitivity of cancers to PARPi is largely dependent on the BRCAness of the cells. Unfortunately, the vast majority of cancers are BRCA-proficient. In this study, therefore, we investigated whether a targeted molecular "hit" on the MRN complex, which is upstream of BRCA, can effectively sensitize BRCA-proficient head and neck squamous cell carcinoma (HNSCC) to PARP inhibitor (PARPi). EXPERIMENTAL DESIGN Human HNSCC cell lines and a mouse model with HNSCC xenografts were used in this study. In vitro and in vivo studies were conducted to evaluate the effects and underlying mechanisms of dual molecular disruption of PARP and the MRN complex, using a pharmacologic inhibitor and a dominant-negative Nbs1 expression vector, respectively. RESULTS Our findings demonstrate that downregulation of the MRN complex disrupts homologous recombination, and, when combined with PARPi, leads to accumulation of lethal DNA double-strand breaks. Moreover, we show that PARPi and MRN complex disruption induces significantly shortening telomere length. Together, our results demonstrate that dual disruption of these pathways causes significant cell death in BRCA-proficient tumor cells both in vitro and in vivo. CONCLUSION Our study, for the first time, elucidates a novel mechanism for MRN complex and PARP inhibition beyond DNA repair, demonstrating the feasibility of a dual disruption approach that extends the utility of PARPi to the treatment of BRCA-proficient cancers.
Collapse
Affiliation(s)
- Shayanne A Lajud
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Danish A Nagda
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Taku Yamashita
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania. Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Jun Zheng
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Nobuaki Tanaka
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania. Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Waleed M Abuzeid
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania. Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California
| | - Alyssa Civantos
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Orysia Bezpalko
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Bert W O'Malley
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Daqing Li
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
9
|
Jekimovs C, Bolderson E, Suraweera A, Adams M, O’Byrne KJ, Richard DJ. Chemotherapeutic compounds targeting the DNA double-strand break repair pathways: the good, the bad, and the promising. Front Oncol 2014; 4:86. [PMID: 24795863 PMCID: PMC4001069 DOI: 10.3389/fonc.2014.00086] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 04/08/2014] [Indexed: 01/09/2023] Open
Abstract
The repair of DNA double-strand breaks (DSBs) is a critical cellular mechanism that exists to ensure genomic stability. DNA DSBs are the most deleterious type of insult to a cell's genetic material and can lead to genomic instability, apoptosis, or senescence. Incorrectly repaired DNA DSBs have the potential to produce chromosomal translocations and genomic instability, potentially leading to cancer. The prevalence of DNA DSBs in cancer due to unregulated growth and errors in repair opens up a potential therapeutic window in the treatment of cancers. The cellular response to DNA DSBs is comprised of two pathways to ensure DNA breaks are repaired: homologous recombination and non-homologous end joining. Identifying chemotherapeutic compounds targeting proteins involved in these DNA repair pathways has shown promise as a cancer therapy for patients, either as a monotherapy or in combination with genotoxic drugs. From the beginning, there have been a number of chemotherapeutic compounds that have yielded successful responses in the clinic, a number that have failed (CGK-733 and iniparib), and a number of promising targets for future studies identified. This review looks in detail at how the cell responds to these DNA DSBs and investigates the chemotherapeutic avenues that have been and are currently being explored to target this repair process.
Collapse
Affiliation(s)
- Christian Jekimovs
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Emma Bolderson
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Amila Suraweera
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Mark Adams
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kenneth J. O’Byrne
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Derek J. Richard
- Cancer and Ageing Research Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
10
|
Carvalho JFS, Kanaar R. Targeting homologous recombination-mediated DNA repair in cancer. Expert Opin Ther Targets 2014; 18:427-58. [PMID: 24491188 DOI: 10.1517/14728222.2014.882900] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION DNA is the target of many traditional non-specific chemotherapeutic drugs. New drugs or therapeutic approaches with a more rational and targeted component are mandatory to improve the success of cancer therapy. The homologous recombination (HR) pathway is an attractive target for the development of inhibitors because cancer cells rely heavily on HR for repair of DNA double-strand breaks resulting from chemotherapeutic treatments. Additionally, the discovery that poly(ADP)ribose polymerase-1 inhibitors selectively kill cells with genetic defects in HR has spurned an even greater interest in inhibitors of HR. AREAS COVERED HR drives the repair of broken DNA via numerous protein-mediated sequential DNA manipulations. Due to extensive number of steps and proteins involved, the HR pathway provides a rich pool of potential drug targets. This review discusses the latest developments concerning the strategies being explored to inhibit HR. Particular attention is given to the identification of small molecule inhibitors of key HR proteins, including the BRCA proteins and RAD51. EXPERT OPINION Current HR inhibitors are providing the basis for pharmaceutical development of more potent and specific inhibitors to be applied in mono- or combinatorial therapy regimes, while novel targets will be uncovered by experiments aimed to gain a deeper mechanistic understanding of HR and its subpathways.
Collapse
Affiliation(s)
- João F S Carvalho
- Erasmus MC Cancer Institute, Department of Genetics, Department of Radiation Oncology, Cancer Genomics Netherlands , PO Box 2040, 3000 CA Rotterdam , The Netherlands
| | | |
Collapse
|
11
|
Rajamoorthi A, Shrivastava S, Steele R, Nerurkar P, Gonzalez JG, Crawford S, Varvares M, Ray RB. Bitter melon reduces head and neck squamous cell carcinoma growth by targeting c-Met signaling. PLoS One 2013; 8:e78006. [PMID: 24147107 PMCID: PMC3798549 DOI: 10.1371/journal.pone.0078006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 09/16/2013] [Indexed: 01/15/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains difficult to treat, and despite of advances in treatment, the overall survival rate has only modestly improved over the past several years. Thus, there is an urgent need for additional therapeutic modalities. We hypothesized that treatment of HNSCC cells with a dietary product such as bitter melon extract (BME) modulates multiple signaling pathways and regresses HNSCC tumor growth in a preclinical model. We observed a reduced cell proliferation in HNSCC cell lines. The mechanistic studies reveal that treatment of BME in HNSCC cells inhibited c-Met signaling pathway. We also observed that BME treatment in HNSCC reduced phosphoStat3, c-myc and Mcl-1 expression, downstream signaling molecules of c-Met. Furthermore, BME treatment in HNSCC cells modulated the expression of key cell cycle progression molecules leading to halted cell growth. Finally, BME feeding in mice bearing HNSCC xenograft tumor resulted in an inhibition of tumor growth and c-Met expression. Together, our results suggested that BME treatment in HNSCC cells modulates multiple signaling pathways and may have therapeutic potential for treating HNSCC.
Collapse
Affiliation(s)
- Ananthi Rajamoorthi
- Department of Pathology, Saint Louis University, St. Louis, Missouri, United States of America
| | - Shubham Shrivastava
- Department of Pathology, Saint Louis University, St. Louis, Missouri, United States of America
| | - Robert Steele
- Department of Pathology, Saint Louis University, St. Louis, Missouri, United States of America
| | - Pratibha Nerurkar
- Laboratory of Metabolic Disorders and Alternative Medicine, Department of Molecular Biosciences and Bioengineering, College of Tropical Agriculture and Human Resources, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Juan G. Gonzalez
- Department of Pathology, Saint Louis University, St. Louis, Missouri, United States of America
| | - Susan Crawford
- Department of Pathology, Saint Louis University, St. Louis, Missouri, United States of America
- Saint Louis University Cancer Center, Saint Louis University, St. Louis, Missouri, United States of America
| | - Mark Varvares
- Saint Louis University Cancer Center, Saint Louis University, St. Louis, Missouri, United States of America
| | - Ratna B. Ray
- Department of Pathology, Saint Louis University, St. Louis, Missouri, United States of America
- Saint Louis University Cancer Center, Saint Louis University, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
12
|
Xu JL, Hu LM, Huang MD, Zhao W, Yin YM, Hu ZB, Ma HX, Shen HB, Shu YQ. Genetic Variants of NBS1 Predict Clinical Outcome of Platinum-based Chemotherapy in Advanced Non-small Cell Lung Cancer in Chinese. Asian Pac J Cancer Prev 2012; 13:851-6. [DOI: 10.7314/apjcp.2012.13.3.851] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
13
|
Garner KM, Eastman A. Variations in Mre11/Rad50/Nbs1 status and DNA damage-induced S-phase arrest in the cell lines of the NCI60 panel. BMC Cancer 2011; 11:206:1-13. [PMID: 21619594 PMCID: PMC3128005 DOI: 10.1186/1471-2407-11-206] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 05/27/2011] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The Mre11/Rad50/Nbs1 (MRN) complex is a regulator of cell cycle checkpoints and DNA repair. Defects in MRN can lead to defective S-phase arrest when cells are damaged. Such defects may elicit sensitivity to selected drugs providing a chemical synthetic lethal interaction that could be used to target therapy to tumors with these defects. The goal of this study was to identify these defects in the NCI60 panel of cell lines and identify compounds that might elicit selective cytotoxicity. METHODS We screened the NCI60 panel in search of cell lines that express low levels of MRN proteins, or that fail to arrest in S-phase in response to the topisomerase I inhibitor SN38. The NCI COMPARE program was used to discover compounds that preferentially target cells with these phenotypes. RESULTS HCT116 cells were initially identified as defective in MRN and S phase arrest. Transfection with Mre11 also elevated Rad50 and Nbs1, and rescued the defective S-phase arrest. Cells of the NCI60 panel exhibited a large range of protein expression but a strong correlation existed between Mre11, Rad50 and Nbs1 consistent with complex formation determining protein stability. Mre11 mRNA correlated best with protein level suggesting it was the primary determinant of the overall level of the complex. Three other cell lines failed to arrest in response to SN38, two of which also had low MRN. However, other cell lines with low MRN still arrested suggesting low MRN does not predict an inability to arrest. Many compounds, including a family of benzothiazoles, correlated with the failure to arrest in S phase. The activity of benzothiazoles has been attributed to metabolic activation and DNA alkylation, but we note several cell lines in which sensitivity does not correlate with metabolism. We propose that the checkpoint defect imposes an additional mechanism of sensitivity on cells. CONCLUSIONS We have identified cells with possible defects in the MRN complex and S phase arrest, and a series of compounds that may preferentially target S phase-defective cells. We discuss limitations of the COMPARE program when attempting to identify compounds that selectively inhibit only a few cell lines.
Collapse
Affiliation(s)
- Kristen M Garner
- Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, NH 03756, USA
| | - Alan Eastman
- Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, NH 03756, USA
| |
Collapse
|
14
|
Araki K, Yamashita T, Reddy N, Wang H, Abuzeid WM, Khan K, O'Malley BW, Li D. Molecular disruption of NBS1 with targeted gene delivery enhances chemosensitisation in head and neck cancer. Br J Cancer 2010; 103:1822-30. [PMID: 21063405 PMCID: PMC3008607 DOI: 10.1038/sj.bjc.6605980] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 09/07/2010] [Accepted: 10/13/2010] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND a fibroblast growth factor 2 (FGF2)-targeted adenoviral system can alter viral tropism and allow for improved transduction and reduced systemic toxicity. This study is to investigate if the FGF2-targeted adenoviral mutant Nijmegen breakage syndrome 1 (FGF2-Ad-NBS1) gene transfer can enhance cisplatin chemosensitisation not only by targeting DNA repair, but also through the induction of antiangiogenesis, whereas at the same time reducing toxicities in treating head and neck squamous cell carcinoma (HNSCC). METHODS the human HNSCC cell line was treated in vitro and in a nude mouse xenograft model. We conducted verification of binding ability of mutant NBS1 and downregulation of MRN complex, evaluation of transduction efficiency and combined antitumour activities. The antiangiogenesis mechanism was also investigated. Finally, we estimated the distribution of adenoviral vector in the liver. RESULTS the mutant NBS1 protein retains the binding ability and effectively suppresses the expression level of the MRN in infected cells. Transduction efficiency in vitro and cisplatin chemosensitisation were upregulated. The FGF2-Ad-NBS1 also showed detargeting the viral vectors away from the liver. The downregulation of NF-κB expression was supposed to correlate with increased antiangiogenesis. CONCLUSIONS FGF2-targeted adenoviral system enhances the cisplatin chemosensitisation of mutant NBS1 and may avoid viral-associated liver toxicities.
Collapse
Affiliation(s)
- K Araki
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, 415 Curie Boulevard, CRB Room 145, Philadelphia, PA 19104, USA
- Department of Otorhinolaryngology-Head & Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - T Yamashita
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, 415 Curie Boulevard, CRB Room 145, Philadelphia, PA 19104, USA
- Department of Otorhinolaryngology-Head & Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - N Reddy
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, 415 Curie Boulevard, CRB Room 145, Philadelphia, PA 19104, USA
| | - H Wang
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, 415 Curie Boulevard, CRB Room 145, Philadelphia, PA 19104, USA
| | - W M Abuzeid
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, 415 Curie Boulevard, CRB Room 145, Philadelphia, PA 19104, USA
| | - K Khan
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, 415 Curie Boulevard, CRB Room 145, Philadelphia, PA 19104, USA
| | - B W O'Malley
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, 415 Curie Boulevard, CRB Room 145, Philadelphia, PA 19104, USA
| | - D Li
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, 415 Curie Boulevard, CRB Room 145, Philadelphia, PA 19104, USA
| |
Collapse
|
15
|
Figures MR, Wobb J, Araki K, Liu T, Xu L, Zhu H, O'Malley BW, Li D. Head and neck squamous cell carcinoma targeted chemosensitization. Otolaryngol Head Neck Surg 2009; 141:177-83. [PMID: 19643248 DOI: 10.1016/j.otohns.2009.04.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Revised: 04/21/2009] [Accepted: 04/30/2009] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The current treatment for advanced head and neck squamous cell carcinoma continues to result in poor outcomes. The purpose of this study is to investigate the benefit of fibroblast growth factor 2-targeted adenovirus-mediated mutant-Rad50 (FGF2-Ad-Rad50) gene transfer in enhancing chemosensitization for head and neck squamous cell carcinoma and reducing chemotoxicity. STUDY DESIGN Randomized controlled laboratory study. SETTING University of Pennsylvania, Philadelphia, PA. SUBJECTS AND METHODS Human head and neck squamous cell carcinoma tumor cells and a mouse model with human head and neck squamous cell carcinoma were used for this study. There were five mice in each study group. FGF2-fab' molecule was conjugated with an adenoviral mutant-Rad50 construct. FGF2-targeted transgene expression efficiency was evaluated in vitro. Tumor cytotoxicity and growth inhibition were examined after combined FGF2-Ad-Rad50 with cisplatin treatment in vitro and in vivo. Anti-tumor mechanisms were investigated. RESULTS FGF2-targeted gene transfer approach significantly improved transgene expression in head and neck squamous cell carcinoma tumor cells over a nontargeted approach (207.51+/-33.62 vs 51.44+/-8.28, respectively). FGF2-Ad-Rad50 with cisplatin demonstrated a superior tumor suppression effect (264.5+/-124.1 mm3 vs 567.1+/-267.6 mm3), increased DNA double-strand breaks (1349+/-51.67 vs 774+/-28.56), and anti-angiogenesis (%ROI: 0.76%+/-0.38% vs 2.10%+/-1.66%) in tumor cells over nontargeted adenovirus. CONCLUSION Combination of FGF2-Ad-Rad50 with cisplatin significantly improves anti-tumor effect by targeting DNA repair systems and tumor angiogenesis. The great benefit of this strategy supports clinical trial for novel treatment of head and neck squamous cell carcinoma.
Collapse
Affiliation(s)
- Mindy R Figures
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Abuzeid WM, Jiang X, Shi G, Wang H, Paulson D, Araki K, Jungreis D, Carney J, O’Malley BW, Li D. Molecular disruption of RAD50 sensitizes human tumor cells to cisplatin-based chemotherapy. J Clin Invest 2009; 119:1974-85. [PMID: 19487811 PMCID: PMC2701852 DOI: 10.1172/jci33816] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Accepted: 03/19/2009] [Indexed: 11/17/2022] Open
Abstract
Platinum-based drugs that induce DNA damage are commonly used first-line chemotherapy agents for testicular, bladder, head and neck, lung, esophageal, stomach, and ovarian cancers. The inherent resistance of tumors to DNA damage often limits the therapeutic efficacy of these agents, such as cisplatin. An enhanced DNA repair and telomere maintenance response by the Mre11/Rad50/Nbs1 (MRN) complex is critical in driving this chemoresistance. We hypothesized therefore that the targeted impairment of native cellular MRN function could sensitize tumor cells to cisplatin. To test this, we designed what we believe to be a novel dominant-negative adenoviral vector containing a mutant RAD50 gene that significantly downregulated MRN expression and markedly disrupted MRN function in human squamous cell carcinoma cells. A combination of cisplatin and mutant RAD50 therapy produced significant tumor cytotoxicity in vitro, with a corresponding increase in DNA damage and telomere shortening. In cisplatin-resistant human squamous cell cancer xenografts in nude mice, this combination therapy caused dramatic tumor regression with increased apoptosis. Our findings suggest the use of targeted RAD50 disruption as what we believe to be a novel chemosensitizing approach for cancer therapy in the context of chemoresistance. This strategy is potentially applicable to several types of malignant tumors that demonstrate chemoresistance and may positively impact the treatment of these patients.
Collapse
Affiliation(s)
- Waleed M. Abuzeid
- Department of Otorhinolaryngology — Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Otolaryngology — Head and Neck Surgery, and
Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Xiaoling Jiang
- Department of Otorhinolaryngology — Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Otolaryngology — Head and Neck Surgery, and
Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Guoli Shi
- Department of Otorhinolaryngology — Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Otolaryngology — Head and Neck Surgery, and
Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Hui Wang
- Department of Otorhinolaryngology — Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Otolaryngology — Head and Neck Surgery, and
Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - David Paulson
- Department of Otorhinolaryngology — Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Otolaryngology — Head and Neck Surgery, and
Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Koji Araki
- Department of Otorhinolaryngology — Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Otolaryngology — Head and Neck Surgery, and
Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - David Jungreis
- Department of Otorhinolaryngology — Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Otolaryngology — Head and Neck Surgery, and
Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - James Carney
- Department of Otorhinolaryngology — Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Otolaryngology — Head and Neck Surgery, and
Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Bert W. O’Malley
- Department of Otorhinolaryngology — Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Otolaryngology — Head and Neck Surgery, and
Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Daqing Li
- Department of Otorhinolaryngology — Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Otolaryngology — Head and Neck Surgery, and
Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
17
|
Saito K, Khan K, Yu SZ, Ronson S, Rhee J, Li G, Van Echo D, Suntharalingam M, O'Malley BW, Li D. The predictive and therapeutic value of thymidine phosphorylase and dihydropyrimidine dehydrogenase in capecitabine (Xeloda)-based chemotherapy for head and neck cancer. Laryngoscope 2009; 119:82-8. [DOI: 10.1002/lary.20003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
18
|
Araki K, Ahmad SM, Li G, Bray DA, Saito K, Wang D, Wirtz U, Sreedharan S, O'Malley BW, Li D. Retinoblastoma RB94 enhances radiation treatment of head and neck squamous cell carcinoma. Clin Cancer Res 2008; 14:3514-9. [PMID: 18519784 DOI: 10.1158/1078-0432.ccr-07-4538] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To assess whether adenovirus-mediated retinoblastoma 94 (Ad-RB94) transgene expression enhances efficacy of radiation therapy (XRT) of human head and neck squamous cell carcinoma (HNSCC). EXPERIMENTAL DESIGN The HNSCC cell lines (JHU006 and JHU012) were treated in vitro and in a nude mouse xenograft model with Ad-RB94, Ad-DL312 control vector, or untreated as mock control. Cell viability and tumor growth were evaluated and combined RB94/XRT antitumor activity was analyzed by measuring DNA double-strand breaks, apoptosis-associated early DNA fragmentation, and levels of RB-regulated cell cycle progression E2F1 transcription factor. RESULTS Ad-RB94/XRT resulted in significant HNSCC cell growth inhibition compared with XRT alone or Ad-RB94 alone in vitro and caused significant tumor regression compared with XRT alone and Ad-DL312/XRT in JHU006 and with XRT alone, Ad-DL312/XRT and Ad-RB94 alone in JHU012 in vivo. Neutral comet analysis revealed that DNA damage was significantly elevated in cells treated with Ad-RB94 alone and Ad-RB94/XRT. Tumors treated with Ad-RB94 alone showed a striking increase in early apoptosis DNA fragmentation, and DNA fragmentation was further enhanced with XRT. In addition, levels of E2F1 were up-regulated by Ad-RB94/XRT combination, whereas Ad-RB94 alone did not affect E2F1 levels and XRT alone led to down-regulation of E2F1. CONCLUSIONS A potent antitumor effect has been observed after Ad-RB94/XRT combination treatment in HNSCC xenograft tumors. Enhanced tumor regression correlated with increased apoptosis. Ad-RB94 treatment enhances the efficacy of XRT through tumor cell sensitization by arresting the cells at the radiation-sensitive G(2)-M cell cycle and via E2F1 up-regulation.
Collapse
Affiliation(s)
- Koji Araki
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Rink L, Slupianek A, Stoklosa T, Nieborowska-Skorska M, Urbanska K, Seferynska I, Reiss K, Skorski T. Enhanced phosphorylation of Nbs1, a member of DNA repair/checkpoint complex Mre11-RAD50-Nbs1, can be targeted to increase the efficacy of imatinib mesylate against BCR/ABL-positive leukemia cells. Blood 2007; 110:651-60. [PMID: 17431132 PMCID: PMC1924483 DOI: 10.1182/blood-2006-08-042630] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2006] [Accepted: 01/27/2007] [Indexed: 02/06/2023] Open
Abstract
Nbs1, a member of the Mre11-RAD50-Nbs1 complex, is phosphorylated by ATM, the product of the ataxia-telangiectasia mutated gene and a member of the phosphatidylinositol 3-kinase-related family of serine-threonine kinases, in response to DNA double-strand breaks (DSBs) to regulate DNA damage checkpoints. Here we show that BCR/ABL stimulated Nbs1 expression by induction of c-Myc-dependent transactivation and protection from caspase-dependent degradation. BCR/ABL-related fusion tyrosine kinases (FTKs) such as TEL/JAK2, TEL/PDGFbetaR, TEL/ABL, TEL/TRKC, BCR/FGFR1, and NPM/ALK as well as interleukin 3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF), and stem cell factor (SCF) also stimulated Nbs1 expression. Enhanced ATM kinase-dependent phosphorylation of Nbs1 on serine 343 (S343) in response to genotoxic treatment was detected in leukemia cells expressing BCR/ABL and other FTKs in comparison to normal counterparts stimulated with IL-3, GM-CSF, and SCF. Expression of Nbs1-S343A mutant disrupted the intra-S-phase checkpoint, decreased homologous recombinational repair (HRR) activity, down-regulated XIAP expression, and sensitized BCR/ABL-positive cells to cytotoxic drugs. Interestingly, inhibition of Nbs1 phosphorylation by S343A mutant enhanced the antileukemia effect of the combination of imatinib and genotoxic agent.
Collapse
Affiliation(s)
- Lori Rink
- Department of Microbiology and Immunology, Temple University, Philadelphia, PA 19140, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Choudhury SA, Kauler P, Devic S, Chow TYK. Silencing of endo-exonuclease expression sensitizes mouse B16F10 melanoma cells to DNA damaging agents. Invest New Drugs 2007; 25:399-410. [PMID: 17492398 DOI: 10.1007/s10637-007-9056-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Accepted: 04/20/2007] [Indexed: 11/25/2022]
Abstract
We previously identified an endo-exonuclease that is highly expressed in cancer cells and plays an important role in DSB repair mechanisms. A small molecular compound pentamidine, which specifically inhibited nuclease activity of the isolated endo-exonuclease from yeast as well as from mammalian cells, was capable of sensitizing tumor cells to DNA damaging agents. In this study, we investigated the effect of precisely silencing the endo-exonuclease expression by small interfering RNA (siRNA) upon treatment with a variety of DNA damaging agents in mouse B16F10 melanoma cells. A maximum of 3.6 to approximately 4-fold reduction in endo-exonuclease mRNA expression was achieved, over a period of 48-72 h of post transfection with a concomitant reduction in protein expression (approximately 4-5 fold), resulting in a substantial reduction (approximately 45-50%) of the corresponding nuclease activity. Suppressed endo-exonuclease expression conferred significant decrease in cell survival, ranging from approximately 30 to approximately 50% cell killing, in presence of DNA damaging drugs methyl methane sulfonate (MMS), cisplatin, 5-fluoro uracil (5-FU) and gamma-irradiation but not at varying dosages of ultra violet (UV) radiation. The data strongly support a role for the endo-exonuclease in repairing DNA damages, induced by MMS, cisplatin, 5-FU and gamma irradiation but not by UV radiation. The results presented in this study suggest that the endo-exonuclease siRNA could be useful as a therapeutic tool in targeting the endo-exonuclease in cancer therapy.
Collapse
Affiliation(s)
- Sibgat A Choudhury
- Department of Oncology, Faculty of Medicine, McGill University, 845 Sherbrooke St., Montreal, QC, H3A 2T5, Canada
| | | | | | | |
Collapse
|
21
|
Zhang P, Zhang Z, Zhou X, Qiu W, Chen F, Chen W. Identification of genes associated with cisplatin resistance in human oral squamous cell carcinoma cell line. BMC Cancer 2006; 6:224. [PMID: 16978399 PMCID: PMC1586025 DOI: 10.1186/1471-2407-6-224] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2006] [Accepted: 09/15/2006] [Indexed: 11/19/2022] Open
Abstract
Background Cisplatin is widely used for chemotherapy of head and neck squamous cell carcinoma. However, details of the molecular mechanism responsible for cisplatin resistance are still unclear. The aim of this study was to identify the expression of genes related to cisplatin resistance in oral squamous cell carcinoma cells. Methods A cisplatin-resistant cell line, Tca/cisplatin, was established from a cisplatin-sensitive cell line, Tca8113, which was derived from moderately-differentiated tongue squamous cell carcinoma. Global gene expression in this resistant cell line and its sensitive parent cell line was analyzed using Affymetrix HG-U95Av2 microarrays. Candidate genes involved in DNA repair, the MAP pathway and cell cycle regulation were chosen to validate the microarray analysis results. Cell cycle distribution and apoptosis following cisplatin exposure were also investigated. Results Cisplatin resistance in Tca/cisplatin cells was stable for two years in cisplatin-free culture medium. The IC50 for cisplatin in Tca/cisplatin was 6.5-fold higher than that in Tca8113. Microarray analysis identified 38 genes that were up-regulated and 25 that were down-regulated in this cell line. Some were novel candidates, while others are involved in well-characterized mechanisms that could be relevant to cisplatin resistance, such as RECQL for DNA repair and MAP2K6 in the MAP pathway; all the genes were further validated by Real-time PCR. The cell cycle-regulated genes CCND1 and CCND3 were involved in cisplatin resistance; 24-hour exposure to 10 μM cisplatin induced a marked S phase block in Tca/cisplatin cells but not in Tca8113 cells. Conclusion The Tca8113 cell line and its stable drug-resistant variant Tca/cisplatin provided a useful model for identifying candidate genes responsible for the mechanism of cisplatin resistance in oral squamous cell carcinoma. Our data provide a useful basis for screening candidate targets for early diagnosis and further intervention in cisplatin resistance.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Oral & Maxillofacial Surgery, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Zhiyuan Zhang
- Department of Oral & Maxillofacial Surgery, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Xiaojian Zhou
- Department of Oral & Maxillofacial Surgery, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Weiliu Qiu
- Department of Oral & Maxillofacial Surgery, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Fangan Chen
- Department of Oral and Maxillofacial Surgery, School of Medicine, New York University, New York 10016, USA
| | - Wantao Chen
- Department of Oral & Maxillofacial Surgery, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| |
Collapse
|