1
|
Gera K, Cline C, Al-Mansour Z, Medvec A, Lee JH, Galochkina Z, Hsu J, Hiemenz J, Farhadfar N, Dean EA, Wingard JR, Brown R. A phase ib clinical trial of oral ciprofloxacin and etoposide in subjects with resistant acute myeloid leukemia. Leuk Lymphoma 2024; 65:1502-1510. [PMID: 38841781 DOI: 10.1080/10428194.2024.2361111] [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: 01/22/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024]
Abstract
A phase 1b study was conducted to evaluate the safety and feasibility of ciprofloxacin and etoposide combination treatment in subjects with relapsed and refractory acute myeloid leukemia. Eleven subjects were enrolled in the study. Utilizing the standard '3 + 3' design, escalating ciprofloxacin doses (750 mg, 1000 mg) twice daily on D1-D10 in combination with a fixed dose (200 mg) of etoposide on D2-D8 were administered. Maximum tolerated dose was determined to be 1000 mg of ciprofloxacin in combination with 200 mg of etoposide. Serious adverse events occurred in 54.5% (n = 6) subjects and 91% (n = 10) subjects reported ≥ grade 3 toxicities. Nine subjects completed treatment, one had a dose-limiting toxicity, and one withdrew. One subject achieved complete remission with a duration of 111 days and one subject achieved morphologic leukemia-free state after cycle 1. While the combination demonstrated safety and an acceptable toxicity profile, only modest hematologic and clinical benefits were observed.This trial was registered at www.clinicaltrials.gov as #NCT02773732.
Collapse
Affiliation(s)
- Kriti Gera
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Christina Cline
- Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Zeina Al-Mansour
- Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Andrew Medvec
- Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ji-Hyun Lee
- Department of Biostatistics, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Zhanna Galochkina
- Division of Quantitative Sciences, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Jack Hsu
- Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - John Hiemenz
- Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Nosha Farhadfar
- Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Erin A Dean
- Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - John R Wingard
- Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Randy Brown
- Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
2
|
Nickoloff JA. Targeting Replication Stress Response Pathways to Enhance Genotoxic Chemo- and Radiotherapy. Molecules 2022; 27:4736. [PMID: 35897913 PMCID: PMC9330692 DOI: 10.3390/molecules27154736] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/12/2022] Open
Abstract
Proliferating cells regularly experience replication stress caused by spontaneous DNA damage that results from endogenous reactive oxygen species (ROS), DNA sequences that can assume secondary and tertiary structures, and collisions between opposing transcription and replication machineries. Cancer cells face additional replication stress, including oncogenic stress that results from the dysregulation of fork progression and origin firing, and from DNA damage induced by radiotherapy and most cancer chemotherapeutic agents. Cells respond to such stress by activating a complex network of sensor, signaling and effector pathways that protect genome integrity. These responses include slowing or stopping active replication forks, protecting stalled replication forks from collapse, preventing late origin replication firing, stimulating DNA repair pathways that promote the repair and restart of stalled or collapsed replication forks, and activating dormant origins to rescue adjacent stressed forks. Currently, most cancer patients are treated with genotoxic chemotherapeutics and/or ionizing radiation, and cancer cells can gain resistance to the resulting replication stress by activating pro-survival replication stress pathways. Thus, there has been substantial effort to develop small molecule inhibitors of key replication stress proteins to enhance tumor cell killing by these agents. Replication stress targets include ATR, the master kinase that regulates both normal replication and replication stress responses; the downstream signaling kinase Chk1; nucleases that process stressed replication forks (MUS81, EEPD1, Metnase); the homologous recombination catalyst RAD51; and other factors including ATM, DNA-PKcs, and PARP1. This review provides an overview of replication stress response pathways and discusses recent pre-clinical studies and clinical trials aimed at improving cancer therapy by targeting replication stress response factors.
Collapse
Affiliation(s)
- Jac A Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
3
|
Lié O, Renault S, Augé-Gouillou C. SETMAR, a case of primate co-opted genes: towards new perspectives. Mob DNA 2022; 13:9. [PMID: 35395947 PMCID: PMC8994322 DOI: 10.1186/s13100-022-00267-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 03/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We carry out a review of the history and biological activities of one domesticated gene in higher primates, SETMAR, by discussing current controversies. Our purpose is to open a new outlook that will serve as a framework for future work about SETMAR, possibly in the field of cognition development. MAIN BODY What is newly important about SETMAR can be summarized as follows: (1) the whole protein sequence is under strong purifying pressure; (2) its role is to strengthen existing biological functions rather than to provide new ones; (3) it displays a tissue-specific pattern of expression, at least for the alternative-splicing it undergoes. Studies reported here demonstrate that SETMAR protein(s) may be involved in essential networks regulating replication, transcription and translation. Moreover, during embryogenesis, SETMAR appears to contribute to brain development. SHORT CONCLUSION Our review underlines for the first time that SETMAR directly interacts with genes involved in brain functions related to vocalization and vocal learning. These findings pave the way for future works regarding SETMAR and the development of cognitive abilities in higher primates.
Collapse
Affiliation(s)
- Oriane Lié
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,iBrain, Team Neurogenomics and Neuronal physiopathology, Faculty of Medicine, 10 Bd Tonnellé, Cedex 1, 37032, Tours, France
| | - Sylvaine Renault
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,iBrain, Team Neurogenomics and Neuronal physiopathology, Faculty of Medicine, 10 Bd Tonnellé, Cedex 1, 37032, Tours, France
| | - Corinne Augé-Gouillou
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France. .,iBrain, Team Neurogenomics and Neuronal physiopathology, Faculty of Medicine, 10 Bd Tonnellé, Cedex 1, 37032, Tours, France.
| |
Collapse
|
4
|
Nickoloff JA, Sharma N, Taylor L, Allen SJ, Hromas R. Nucleases and Co-Factors in DNA Replication Stress Responses. DNA 2022; 2:68-85. [PMID: 36203968 PMCID: PMC9534323 DOI: 10.3390/dna2010006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
DNA replication stress is a constant threat that cells must manage to proliferate and maintain genome integrity. DNA replication stress responses, a subset of the broader DNA damage response (DDR), operate when the DNA replication machinery (replisome) is blocked or replication forks collapse during S phase. There are many sources of replication stress, such as DNA lesions caused by endogenous and exogenous agents including commonly used cancer therapeutics, and difficult-to-replicate DNA sequences comprising fragile sites, G-quadraplex DNA, hairpins at trinucleotide repeats, and telomeres. Replication stress is also a consequence of conflicts between opposing transcription and replication, and oncogenic stress which dysregulates replication origin firing and fork progression. Cells initially respond to replication stress by protecting blocked replisomes, but if the offending problem (e.g., DNA damage) is not bypassed or resolved in a timely manner, forks may be cleaved by nucleases, inducing a DNA double-strand break (DSB) and providing a means to accurately restart stalled forks via homologous recombination. However, DSBs pose their own risks to genome stability if left unrepaired or misrepaired. Here we focus on replication stress response systems, comprising DDR signaling, fork protection, and fork processing by nucleases that promote fork repair and restart. Replication stress nucleases include MUS81, EEPD1, Metnase, CtIP, MRE11, EXO1, DNA2-BLM, SLX1-SLX4, XPF-ERCC1-SLX4, Artemis, XPG, and FEN1. Replication stress factors are important in cancer etiology as suppressors of genome instability associated with oncogenic mutations, and as potential cancer therapy targets to enhance the efficacy of chemo- and radiotherapeutics.
Collapse
Affiliation(s)
- Jac A. Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Ft. Collins, CO 80523, USA
| | - Neelam Sharma
- Department of Environmental and Radiological Health Sciences, Colorado State University, Ft. Collins, CO 80523, USA
| | - Lynn Taylor
- Department of Environmental and Radiological Health Sciences, Colorado State University, Ft. Collins, CO 80523, USA
| | - Sage J. Allen
- Department of Environmental and Radiological Health Sciences, Colorado State University, Ft. Collins, CO 80523, USA
| | - Robert Hromas
- Division of Hematology and Medical Oncology, Department of Medicine and the Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX 78229, USA
| |
Collapse
|
5
|
Nickoloff JA, Sharma N, Taylor L, Allen SJ, Lee SH, Hromas R. Metnase and EEPD1: DNA Repair Functions and Potential Targets in Cancer Therapy. Front Oncol 2022; 12:808757. [PMID: 35155245 PMCID: PMC8831698 DOI: 10.3389/fonc.2022.808757] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/12/2022] [Indexed: 12/30/2022] Open
Abstract
Cells respond to DNA damage by activating signaling and DNA repair systems, described as the DNA damage response (DDR). Clarifying DDR pathways and their dysregulation in cancer are important for understanding cancer etiology, how cancer cells exploit the DDR to survive endogenous and treatment-related stress, and to identify DDR targets as therapeutic targets. Cancer is often treated with genotoxic chemicals and/or ionizing radiation. These agents are cytotoxic because they induce DNA double-strand breaks (DSBs) directly, or indirectly by inducing replication stress which causes replication fork collapse to DSBs. EEPD1 and Metnase are structure-specific nucleases, and Metnase is also a protein methyl transferase that methylates histone H3 and itself. EEPD1 and Metnase promote repair of frank, two-ended DSBs, and both promote the timely and accurate restart of replication forks that have collapsed to single-ended DSBs. In addition to its roles in HR, Metnase also promotes DSB repair by classical non-homologous recombination, and chromosome decatenation mediated by TopoIIα. Although mutations in Metnase and EEPD1 are not common in cancer, both proteins are frequently overexpressed, which may help tumor cells manage oncogenic stress or confer resistance to therapeutics. Here we focus on Metnase and EEPD1 DNA repair pathways, and discuss opportunities for targeting these pathways to enhance cancer therapy.
Collapse
Affiliation(s)
- Jac A Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States
| | - Neelam Sharma
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States
| | - Lynn Taylor
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States
| | - Sage J Allen
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States
| | - Suk-Hee Lee
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Robert Hromas
- Division of Hematology and Medical Oncology, Department of Medicine and the Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, United States
| |
Collapse
|
6
|
Tellier M. Structure, Activity, and Function of SETMAR Protein Lysine Methyltransferase. Life (Basel) 2021; 11:life11121342. [PMID: 34947873 PMCID: PMC8704517 DOI: 10.3390/life11121342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 12/21/2022] Open
Abstract
SETMAR is a protein lysine methyltransferase that is involved in several DNA processes, including DNA repair via the non-homologous end joining (NHEJ) pathway, regulation of gene expression, illegitimate DNA integration, and DNA decatenation. However, SETMAR is an atypical protein lysine methyltransferase since in anthropoid primates, the SET domain is fused to an inactive DNA transposase. The presence of the DNA transposase domain confers to SETMAR a DNA binding activity towards the remnants of its transposable element, which has resulted in the emergence of a gene regulatory function. Both the SET and the DNA transposase domains are involved in the different cellular roles of SETMAR, indicating the presence of novel and specific functions in anthropoid primates. In addition, SETMAR is dysregulated in different types of cancer, indicating a potential pathological role. While some light has been shed on SETMAR functions, more research and new tools are needed to better understand the cellular activities of SETMAR and to investigate the therapeutic potential of SETMAR.
Collapse
Affiliation(s)
- Michael Tellier
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| |
Collapse
|
7
|
Antoine-Lorquin A, Arensburger P, Arnaoty A, Asgari S, Batailler M, Beauclair L, Belleannée C, Buisine N, Coustham V, Guyetant S, Helou L, Lecomte T, Pitard B, Stévant I, Bigot Y. Two repeated motifs enriched within some enhancers and origins of replication are bound by SETMAR isoforms in human colon cells. Genomics 2021; 113:1589-1604. [PMID: 33812898 DOI: 10.1016/j.ygeno.2021.03.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 11/15/2022]
Abstract
Setmar is a gene specific to simian genomes. The function(s) of its isoforms are poorly understood and their existence in healthy tissues remains to be validated. Here we profiled SETMAR expression and its genome-wide binding landscape in colon tissue. We found isoforms V3 and V6 in healthy and tumour colon tissues as well as incell lines. In two colorectal cell lines SETMAR binds to several thousand Hsmar1 and MADE1 terminal ends, transposons mostly located in non-genic regions of active chromatin including in enhancers. It also binds to a 12-bp motifs similar to an inner motif in Hsmar1 and MADE1 terminal ends. This motif is interspersed throughout the genome and is enriched in GC-rich regions as well as in CpG islands that contain constitutive replication origins. It is also found in enhancers other than those associated with Hsmar1 and MADE1. The role of SETMAR in the expression of genes, DNA replication and in DNA repair are discussed.
Collapse
Affiliation(s)
| | - Peter Arensburger
- Biological Sciences Department, California State Polytechnic University, Pomona, CA 91768, - United States
| | - Ahmed Arnaoty
- EA GICC, 7501, CHRU de Tours, 37044 TOURS, Cedex 09, France
| | - Sassan Asgari
- School of Biological Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Martine Batailler
- PRC, UMR INRA 0085, CNRS 7247, Centre INRA Val de Loire, 37380 Nouzilly, France
| | - Linda Beauclair
- PRC, UMR INRA 0085, CNRS 7247, Centre INRA Val de Loire, 37380 Nouzilly, France
| | | | - Nicolas Buisine
- UMR CNRS 7221, Muséum National d'Histoire Naturelle, 75005 Paris, France
| | | | - Serge Guyetant
- Tumorothèque du CHRU de Tours, 37044 Tours, Cedex, France
| | - Laura Helou
- PRC, UMR INRA 0085, CNRS 7247, Centre INRA Val de Loire, 37380 Nouzilly, France
| | | | - Bruno Pitard
- Université de Nantes, CNRS ERL6001, Inserm 1232, CRCINA, F-44000 Nantes, France
| | - Isabelle Stévant
- Institut de Génomique Fonctionnelle de Lyon, Univ Lyon, CNRS UMR 5242, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon, 1, 46 allée d'Italie, 69364 Lyon, France
| | - Yves Bigot
- PRC, UMR INRA 0085, CNRS 7247, Centre INRA Val de Loire, 37380 Nouzilly, France.
| |
Collapse
|
8
|
Sharma N, Speed MC, Allen CP, Maranon DG, Williamson E, Singh S, Hromas R, Nickoloff JA. Distinct roles of structure-specific endonucleases EEPD1 and Metnase in replication stress responses. NAR Cancer 2020; 2:zcaa008. [PMID: 32743552 PMCID: PMC7380491 DOI: 10.1093/narcan/zcaa008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 05/20/2020] [Accepted: 05/31/2020] [Indexed: 12/16/2022] Open
Abstract
Accurate DNA replication and segregation are critical for maintaining genome integrity and suppressing cancer. Metnase and EEPD1 are DNA damage response (DDR) proteins frequently dysregulated in cancer and implicated in cancer etiology and tumor response to genotoxic chemo- and radiotherapy. Here, we examine the DDR in human cell lines with CRISPR/Cas9 knockout of Metnase or EEPD1. The knockout cell lines exhibit slightly slower growth rates, significant hypersensitivity to replication stress, increased genome instability and distinct alterations in DDR signaling. Metnase and EEPD1 are structure-specific nucleases. EEPD1 is recruited to and cleaves stalled forks to initiate fork restart by homologous recombination. Here, we demonstrate that Metnase is also recruited to stalled forks where it appears to dimethylate histone H3 lysine 36 (H3K36me2), raising the possibility that H3K36me2 promotes DDR factor recruitment or limits nucleosome eviction to protect forks from nucleolytic attack. We show that stalled forks are cleaved normally in the absence of Metnase, an important and novel result because a prior study indicated that Metnase nuclease is important for timely fork restart. A double knockout was as sensitive to etoposide as either single knockout, suggesting a degree of epistasis between Metnase and EEPD1. We propose that EEPD1 initiates fork restart by cleaving stalled forks, and that Metnase may promote fork restart by processing homologous recombination intermediates and/or inducing H3K36me2 to recruit DDR factors. By accelerating fork restart, Metnase and EEPD1 reduce the chance that stalled replication forks will adopt toxic or genome-destabilizing structures, preventing genome instability and cancer. Metnase and EEPD1 are overexpressed in some cancers and thus may also promote resistance to genotoxic therapeutics.
Collapse
Affiliation(s)
- Neelam Sharma
- Department of Environmental and Radiological Health Sciences, Colorado State University, 1618 Campus Delivery, Fort Collins, CO 80523-1618, USA
| | - Michael C Speed
- Department of Environmental and Radiological Health Sciences, Colorado State University, 1618 Campus Delivery, Fort Collins, CO 80523-1618, USA
| | - Christopher P Allen
- Department of Microbiology, Immunology, and Pathology, Colorado State University, 1601Campus Delivery, Fort Collins, CO 80523-1601, USA
| | - David G Maranon
- Department of Environmental and Radiological Health Sciences, Colorado State University, 1618 Campus Delivery, Fort Collins, CO 80523-1618, USA
| | - Elizabeth Williamson
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas HealthScience Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Sudha Singh
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas HealthScience Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Robert Hromas
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas HealthScience Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Jac A Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, 1618 Campus Delivery, Fort Collins, CO 80523-1618, USA
| |
Collapse
|
9
|
Shmakova A, Germini D, Vassetzky Y. HIV-1, HAART and cancer: A complex relationship. Int J Cancer 2020; 146:2666-2679. [PMID: 31603989 DOI: 10.1002/ijc.32730] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/30/2019] [Accepted: 10/02/2019] [Indexed: 12/14/2022]
Abstract
HIV infected people are at higher risk of developing cancer, although it is globally diminished in the era of highly active antiretroviral treatment (HAART). Recently, antioncogenic properties of some HAART drugs were discovered. We discuss the role of HAART in the prevention and improvement of treatment outcomes of cancers in HIV-infected people. We describe different trends in HAART-cancer relationships: cancer-predisposing as well as cancer-preventing. We cover the roles of particular drug regimens in cancer prevention. We also describe the causes of cancer treatment with HAART drugs in HIV-negative people, including ongoing clinical studies that may directly point to a possible independent anti-oncogenic activity of HAART drugs. We conclude that despite potent antioncogenic activities of every class of HAART drugs reported in preclinical models, the evidence to date indicates that their independent clinical impact in HIV-infected people is limited. Improved cancer prevention strategies besides HAART are needed to reduce HIV-cancer-related mortality.
Collapse
Affiliation(s)
- Anna Shmakova
- UMR 8126, CNRS, Univ. Paris-Sud, Institut Gustave Roussy, Université Paris Saclay, Édouard-Vaillant, Villejuif, France
- LIA 1066 LFR2O French-Russian Joint Cancer Research Laboratory, Édouard-Vaillant, Villejuif, France
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Diego Germini
- UMR 8126, CNRS, Univ. Paris-Sud, Institut Gustave Roussy, Université Paris Saclay, Édouard-Vaillant, Villejuif, France
- LIA 1066 LFR2O French-Russian Joint Cancer Research Laboratory, Édouard-Vaillant, Villejuif, France
| | - Yegor Vassetzky
- UMR 8126, CNRS, Univ. Paris-Sud, Institut Gustave Roussy, Université Paris Saclay, Édouard-Vaillant, Villejuif, France
- LIA 1066 LFR2O French-Russian Joint Cancer Research Laboratory, Édouard-Vaillant, Villejuif, France
- Koltzov Institute of Developmental Biology, Moscow, Russia
| |
Collapse
|
10
|
Mușat MG, Nițulescu GM, Surleac M, Tsatsakis A, Spandidos DA, Margină D. HIV‑1 integrase inhibitors targeting various DDE transposases: Retroviral integration versus RAG‑mediated recombination (Review). Mol Med Rep 2019; 20:4749-4762. [PMID: 31702817 PMCID: PMC6854553 DOI: 10.3892/mmr.2019.10777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 10/25/2019] [Indexed: 12/18/2022] Open
Abstract
Transposases are ubiquitous mobile genetic elements responsible for genome development, driving rearrangements, such as insertions, deletions and translocations. Across species evolution, some transposases are tamed by their host and are made part of complex cellular systems. The proliferation of retroviruses is also dependent on transposase related enzymes termed integrases. Recombination‑activating gene protein (RAG)1 and metnase are just two examples of transposase domestication and together with retroviral integrases (INs), they belong to the DDE polynucleotidyl transferases superfamily. They share mechanistic and structural features linked to the RNase H‑like fold, harboring a DDE(D) metal dependent catalytic motif. Recent antiretroviral compounds target the catalytic domain of integrase, but they also have the potential of inhibiting other related enzymes. In this review, we report the activity of different classes of integrase inhibitors on various DDE transposases. Computational simulations are useful to predict the extent of off‑target activity and have been employed to study the interactions between RAG1 recombinase and compounds from three different pharmacologic classes. We demonstrate that strand‑transfer inhibitors display a higher affinity towards the RAG1 RNase H domain, as suggested by experimental data compared to allosteric inhibitors. While interference with RAG1 and 2 recombination is associated with a negative impact on immune function, the inhibition of metnase or HTLV‑1 integrase opens the way for the development of novel therapies for refractory cancers.
Collapse
Affiliation(s)
- Mihaela Georgiana Mușat
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - George Mihai Nițulescu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Marius Surleac
- National Institute for Infectious Diseases ‘Matei Bals’, 021105 Bucharest, Romania
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Denisa Margină
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania
| |
Collapse
|
11
|
Aldaghi SA, Jalal R. Concentration-Dependent Dual Effects of Ciprofloxacin on SB-590885-Resistant BRAF V600E A375 Melanoma Cells. Chem Res Toxicol 2019; 32:645-658. [PMID: 30829029 DOI: 10.1021/acs.chemrestox.8b00335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BRAF inhibitors (BRAFi) have been applied to treat melanoma harboring V600E mutations. Several studies showed that BRAFi-resistant melanomas are dependent on mitochondrial biogenesis. Therefore, the present study aimed to investigate the influence of ciprofloxacin (CIP), a mitochondria-targeting antibiotic, on SB-590885-resistant BRAFV600E A375 melanoma (A375/SB) cells. The cytotoxicity activity of CIP and SB-590885, a potent and specific BRAFi, on A375 and A375/SB cells was evaluated by MTT, colony formation, migration, and spheroid formation assays. Moreover, SB-590885-induced cell death in A375 cells was analyzed. SB-590885 showed time- and concentration-dependent cytotoxic effects on A375 cells. Twenty-five μg/mL CIP decreased the cell viability of A375 and A375/SB cells in a time-dependent manner. This concentration of CIP markedly decreased clonogenicity in both cells and caused a reduction in the growth of A375/SB spheroids. The cytotoxicity of 5 μg/mL CIP on A375/SB cells was less than that of A375 cells. The colony formation and migration ability of A375/SB cells was increased in the presence of 5 μg/mL CIP. Ten μM SB-590885 induced a massive vacuolization in A375 cells. Cell death assays suggested a simultaneous activation of autophagy, paraptosis, apoptosis, and necrosis. For the first time, this study reveals that CIP at the maximum concentration in serum (5 μg/mL) can enhance the colony formation and migration abilities in BRAFi-resistant melanoma cells, while it has cytotoxic activity against these cells at a higher concentration than serum level. This study suggests that CIP may promote aggressive growth properties in BRAFi-resistant melanomas, at a concentration present in serum.
Collapse
Affiliation(s)
- Seyyede Araste Aldaghi
- Department of Chemistry, Faculty of Science , Ferdowsi University of Mashhad , Mashhad , Iran
| | - Razieh Jalal
- Department of Chemistry, Faculty of Science , Ferdowsi University of Mashhad , Mashhad , Iran.,Department of Research Cell and Molecular Biology, Institute of Biotechnology , Ferdowsi University of Mashhad , Mashhad , Iran
| |
Collapse
|
12
|
Schmidl S, Iancu CV, Choe JY, Oreb M. Ligand Screening Systems for Human Glucose Transporters as Tools in Drug Discovery. Front Chem 2018; 6:183. [PMID: 29888221 PMCID: PMC5980966 DOI: 10.3389/fchem.2018.00183] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/07/2018] [Indexed: 12/22/2022] Open
Abstract
Hexoses are the major source of energy and carbon skeletons for biosynthetic processes in all kingdoms of life. Their cellular uptake is mediated by specialized transporters, including glucose transporters (GLUT, SLC2 gene family). Malfunction or altered expression pattern of GLUTs in humans is associated with several widespread diseases including cancer, diabetes and severe metabolic disorders. Their high relevance in the medical area makes these transporters valuable drug targets and potential biomarkers. Nevertheless, the lack of a suitable high-throughput screening system has impeded the determination of compounds that would enable specific manipulation of GLUTs so far. Availability of structural data on several GLUTs enabled in silico ligand screening, though limited by the fact that only two major conformations of the transporters can be tested. Recently, convenient high-throughput microbial and cell-free screening systems have been developed. These remarkable achievements set the foundation for further and detailed elucidation of the molecular mechanisms of glucose transport and will also lead to great progress in the discovery of GLUT effectors as therapeutic agents. In this mini-review, we focus on recent efforts to identify potential GLUT-targeting drugs, based on a combination of structural biology and different assay systems.
Collapse
Affiliation(s)
- Sina Schmidl
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Cristina V Iancu
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Jun-Yong Choe
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Mislav Oreb
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
13
|
Dussaussois-Montagne A, Jaillet J, Babin L, Verrelle P, Karayan-Tapon L, Renault S, Rousselot-Denis C, Zemmoura I, Augé-Gouillou C. SETMAR isoforms in glioblastoma: A matter of protein stability. Oncotarget 2017; 8:9835-9848. [PMID: 28038463 PMCID: PMC5354774 DOI: 10.18632/oncotarget.14218] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 12/05/2016] [Indexed: 01/10/2023] Open
Abstract
Glioblastomas (GBMs) are the most frequent and the most aggressive brain tumors, known for their chemo- and radio-resistance, making them often incurable. We also know that SETMAR is a protein involved in chromatin dynamics and genome plasticity, of which overexpression confers chemo- and radio-resistance to some tumors. The relationships between SETMAR and GBM have never been explored. To fill this gap, we define the SETMAR status of 44 resected tumors and of GBM derived cells, at both the mRNA and the protein levels. We identify a new, small SETMAR protein (so called SETMAR-1200), enriched in GBMs and GBM stem cells as compared to the regular enzyme (SETMAR-2100). We show that SETMAR-1200 is able to increase DNA repair by non-homologous end-joining, albeit with a lower efficiency than the regular SETMAR protein. Interestingly, the regular/small ratio of SETMAR in GBM cells changes depending on cell type, providing evidence that SETMAR expression is regulated by alternative splicing. We also demonstrate that SETMAR expression can be regulated by the use of an alternative ATG. In conclusion, various SETMAR proteins can be synthesized in human GBM that may each have specific biophysical and/or biochemical properties and characteristics. Among them, the small SETMAR may play a role in GBMs biogenesis. On this basis, we would like to consider SETMAR-1200 as a new potential therapeutic target to investigate, in addition to the regular SETMAR protein already considered by others.
Collapse
Affiliation(s)
| | - Jérôme Jaillet
- EA 6306 IGC, University François Rabelais, 37200 Tours, France
| | - Laetitia Babin
- EA 6306 IGC, University François Rabelais, 37200 Tours, France
- UMR CNRS 7292 GICC, University François Rabelais, 37000 Tours, France
| | - Pierre Verrelle
- EA 7283 CREaT, Université d′Auvergne, BP 10448, 63000 Clermont-Ferrand, France
- Institut Curie, Dpt d'Oncologie Radiothérapique, 75005 Paris, France
- Centre Jean Perrin, Service Radiothérapie, Laboratoire de Radio-Oncologie Expérimentale, 63000 Clermont-Ferrand, France
| | - Lucie Karayan-Tapon
- INSERM U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, F-86021 Poitiers, France
- University of Poitiers, F-86022 Poitiers, France
- CHU of Poitiers, Laboratoire de Cancérologie Biologique, F-86021 Poitiers, France
| | | | | | - Ilyess Zemmoura
- INSERM U930 Imagerie & Cerveau, University François Rabelais, 37000 Tours, France
- CHRU of Tours, Service de Neurochirurgie, 37000 Tours, France
| | | |
Collapse
|
14
|
Theanine from tea and its semi-synthetic derivative TBrC suppress human cervical cancer growth and migration by inhibiting EGFR/Met-Akt/NF-κB signaling. Eur J Pharmacol 2016; 791:297-307. [DOI: 10.1016/j.ejphar.2016.09.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 09/04/2016] [Accepted: 09/05/2016] [Indexed: 01/02/2023]
|
15
|
Zhang G, Li Z, Wan X, Zhang Y, Zhu R, Liu Z, Ji D, Zhang H, Wu F, Tian H, Liu K, Wu B. Repression of Human Hepatocellular Carcinoma Growth by Regulating Met/EGFR/VEGFR-Akt/NF-κB Pathways with Theanine and Its Derivative, (R)-2-(6,8-Dibromo-2-oxo-2H-chromene-3-carboxamido)-5-(ethylamino)-5-oxopentanoic Ethyl Ester (DTBrC). JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:7002-7013. [PMID: 27569455 DOI: 10.1021/acs.jafc.6b02509] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
To explore the potential of theanine against cancer, we have studied the anticancer activities of theanine from tea and its semisynthesized derivative, (R)-2-(6,8-dibromo-2-oxo-2H-chromene-3-carboxamido)-5-(ethylamino)-5-oxopentanoic ethyl ester (DTBrC), in in vitro, ex vivo, and in vivo models of human hepatocellular carcinoma (HHC). Theanine and DTBrC displayed inhibitory effects on the growth and migration of HHC cells in vitro, ex vivo, and in vivo. Theanine and DTBrC significantly enhanced the repression of HHC cell growth in combination with anticancer drug pirarubicin. Theanine and DTBrC completely suppressed HGF- and EGF+HGF-induced migration with a reduction of p53 tumor suppressor level and enhanced the p53 protein expression in HHC cells. The Akt and NF-κB knockdown greatly reduced cancer cell migration with a decrease in CD44 expression. DTBrC and theanine significantly repressed the protein expressions in the Met/EGFR/VEGFR-Akt/NF-κB pathways, which might be the mechanism for their biologic effects.
Collapse
Affiliation(s)
- Guoying Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University , Yantai, 264005, People's Republic of China
| | - Zheng Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University , Yantai, 264005, People's Republic of China
| | - Xiaochun Wan
- Key Laboratory of Tea Biochemistry & Biotechnology, Ministry of Agriculture, Anhui Agricultural University , Hefei, Anhui Province 230036, People's Republic of China
| | - Ying Zhang
- Shandong Yingdong Yinghao Biotechnology Inc. , No. 101 Hangtianlu, Gaoxinqu, Yantai, Shandong Province 264670, People's Republic of China
| | - Rongqin Zhu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University , Yantai, 264005, People's Republic of China
| | - Zhenzhen Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University , Yantai, 264005, People's Republic of China
| | - Dexin Ji
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University , Yantai, 264005, People's Republic of China
| | - Huarong Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University , Yantai, 264005, People's Republic of China
| | - Fei Wu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University , Yantai, 264005, People's Republic of China
| | - Huihui Tian
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University , Yantai, 264005, People's Republic of China
| | - Kun Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University , Yantai, 264005, People's Republic of China
| | - Benhao Wu
- Shandong Yingdong Yinghao Biotechnology Inc. , No. 101 Hangtianlu, Gaoxinqu, Yantai, Shandong Province 264670, People's Republic of China
| |
Collapse
|
16
|
York D, Withers SS, Watson KD, Seo KW, Rebhun RB. Enrofloxacin enhances the effects of chemotherapy in canine osteosarcoma cells with mutant and wild-type p53. Vet Comp Oncol 2016; 15:1087-1100. [PMID: 27333821 DOI: 10.1111/vco.12250] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/19/2016] [Accepted: 05/26/2016] [Indexed: 12/15/2022]
Abstract
Adjuvant chemotherapy improves survival time in dogs receiving adequate local control for appendicular osteosarcoma, but most dogs ultimately succumb to metastatic disease. The fluoroquinolone antibiotic enrofloxacin has been shown to inhibit survival and proliferation of canine osteosarcoma cells in vitro. Others have reported that fluoroquinolones may modulate cellular responses to DNA damaging agents and that these effects may be differentially mediated by p53 activity. We therefore determined p53 status and activity in three canine osteosarcoma cell lines and examined the effects of enrofloxacin when used alone or in combination with doxorubicin or carboplatin chemotherapy. Moresco and Abrams canine osteosarcoma cell lines contained mutations in p53, while no mutations were identified in the D17 cells or in a normal canine osteoblast cell line. The addition of enrofloxacin to either doxorubicin or carboplatin resulted in further reductions in osteosarcoma cell viability; this effect was apparent regardless of p53 mutational status or downstream activity.
Collapse
Affiliation(s)
- D York
- The Comparative Oncology Laboratory and Center for Companion Animal Health, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA
| | - S S Withers
- The Comparative Oncology Laboratory and Center for Companion Animal Health, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA
| | - K D Watson
- UC Davis School of Veterinary Medicine, William R. Pritchard Veterinary Medical Teaching Hospital, University of California-Davis, Davis, CA, USA
| | - K W Seo
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Chungnam National University, Daejon, Korea
| | - R B Rebhun
- The Comparative Oncology Laboratory and Center for Companion Animal Health, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, USA
| |
Collapse
|
17
|
George Thompson AM, Ursu O, Babkin P, Iancu CV, Whang A, Oprea TI, Choe JY. Discovery of a specific inhibitor of human GLUT5 by virtual screening and in vitro transport evaluation. Sci Rep 2016; 6:24240. [PMID: 27074918 PMCID: PMC4831007 DOI: 10.1038/srep24240] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/23/2016] [Indexed: 02/07/2023] Open
Abstract
GLUT5, a fructose-transporting member of the facilitative glucose transporter (GLUT, SLC2) family, is a therapeutic target for diabetes and cancer but has no potent inhibitors. We virtually screened a library of 6 million chemicals onto a GLUT5 model and identified N-[4-(methylsulfonyl)-2-nitrophenyl]-1,3-benzodioxol-5-amine (MSNBA) as an inhibitor of GLUT5 fructose transport in proteoliposomes. MSNBA inhibition was specific to GLUT5; this inhibitor did not affect the fructose transport of human GLUT2 or the glucose transport of human GLUT1-4 or bacterial GlcPSe. In MCF7 cells, a human breast cancer cell line, MSNBA competitively inhibited GLUT5 fructose uptake with a KI of 3.2 ± 0.4 μM. Ligand docking, mutagenesis and functional studies indicate that MSNBA binds near the active site and inhibitor discrimination involves H387 of GLUT5. Thus, MSNBA is a selective and potent inhibitor of fructose transport via GLUT5, and the first chemical probe for this transporter. Our data indicate that active site differences in GLUT members could be exploited to further enhance ligand specificity.
Collapse
Affiliation(s)
- Alayna M George Thompson
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Oleg Ursu
- Department of Internal Medicine, Translational Informatics Division, MSC09 5025, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Petr Babkin
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Cristina V Iancu
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Alex Whang
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Tudor I Oprea
- Department of Internal Medicine, Translational Informatics Division, MSC09 5025, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Jun-yong Choe
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| |
Collapse
|
18
|
Kim HS, Kim SK, Hromas R, Lee SH. The SET Domain Is Essential for Metnase Functions in Replication Restart and the 5' End of SS-Overhang Cleavage. PLoS One 2015; 10:e0139418. [PMID: 26437079 PMCID: PMC4593633 DOI: 10.1371/journal.pone.0139418] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/14/2015] [Indexed: 11/19/2022] Open
Abstract
Metnase (also known as SETMAR) is a chimeric SET-transposase protein that plays essential role(s) in non-homologous end joining (NHEJ) repair and replication fork restart. Although the SET domain possesses histone H3 lysine 36 dimethylation (H3K36me2) activity associated with an improved association of early repair components for NHEJ, its role in replication restart is less clear. Here we show that the SET domain is necessary for the recovery from DNA damage at the replication forks following hydroxyurea (HU) treatment. Cells overexpressing the SET deletion mutant caused a delay in fork restart after HU release. Our In vitro study revealed that the SET domain but not the H3K36me2 activity is required for the 5’ end of ss-overhang cleavage with fork and non-fork DNA without affecting the Metnase-DNA interaction. Together, our results suggest that the Metnase SET domain has a positive role in restart of replication fork and the 5’ end of ss-overhang cleavage, providing a new insight into the functional interaction of the SET and the transposase domains.
Collapse
Affiliation(s)
- Hyun-Suk Kim
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Sung-Kyung Kim
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Robert Hromas
- Department of Medicine, University of Florida and Shands Health Care System, Gainesville, Florida, United States of America
| | - Suk-Hee Lee
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Indiana University Simon Cancer Center, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
19
|
Zhang G, Ye X, Ji D, Zhang H, Sun F, Shang C, Zhang Y, Wu E, Wang F, Wu F, Tian H, Liu X, Chen L, Liu K, Wang Y, Liu H, Zhang W, Guan Y, Wang Q, Zhao X, Wan X. Inhibition of lung tumor growth by targeting EGFR/VEGFR-Akt/NF-κB pathways with novel theanine derivatives. Oncotarget 2015; 5:8528-43. [PMID: 25138052 PMCID: PMC4226702 DOI: 10.18632/oncotarget.2336] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The molecularly targeted agents, including anti-VEGF or anti-EGFR monoclonal antibody and some inhibitors of EGFR tyrosine kinase, are effective in the treatment of non-small-cell lung cancer (NSCLC) to a certain extent, but the benefit for a proportion of patients is still limited. Hence, it is necessary and urgent to develop more selective and effective molecular targeted agents against lung cancer. Here, we have synthesized novel theanine derivatives, methyl coumarin-3-carboxylyl L-theanine (TMC), ethyl coumarin-3-carboxylyl L-theanine (TEC), ethyl 6-fluorocoumarin- 3-carboxylyl L-theanine (TFC), and ethyl 6-nitrocoumarin-3-carboxylyl L-theanine (TNC), which are fluorescent small molecules, based on their parental compound theanine and studied their anticancer activities in vitro, ex vivo and in vivo models of human and mouse cancers. Our results show that the four theanine derivatives significantly inhibit the lung cancer cell migration and the growth of lung cancer and leukemia cell lines. TFC and TNC display enhanced effects with anticancer drugs cytarabine vincristine, andmethotrexate on inhibition of lung cancer cell growth and no toxicity to the normal human embryonic lung fibroblast and peripheral blood lymphocytes. TFC and TNC exhibit strong suppression of the highly metastatic Lewis lung cancer (LLC) and A549 tumor growth in tumor-bearing mice without toxicity to mice. TFC and TNC can effectively suppress the growth of lung cancer cells in vitro, ex vivo and in vivo by targeting EGFR/VEGFR-Akt/NF-κB pathways. Our study has suggested that TFC and TNC may have the therapeutic and/or adjuvant therapeutic applications in the treatment of lung cancers and other cancer.
Collapse
Affiliation(s)
- Guoying Zhang
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, Qing Quan Lu, Yantai, Shandong Province, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, P R China
| | - Xinshan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, P R China
| | - Dexin Ji
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, Qing Quan Lu, Yantai, Shandong Province, China; These authors contributed equally to this work
| | - Huarong Zhang
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, Qing Quan Lu, Yantai, Shandong Province, China; These authors contributed equally to this work
| | - Fujia Sun
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, Qing Quan Lu, Yantai, Shandong Province, China; These authors contributed equally to this work
| | - Chunqing Shang
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, Qing Quan Lu, Yantai, Shandong Province, China; These authors contributed equally to this work
| | - Ying Zhang
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, Qing Quan Lu, Yantai, Shandong Province, China; Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA; These authors contributed equally to this work
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Fengfei Wang
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Fei Wu
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, Qing Quan Lu, Yantai, Shandong Province, China
| | - Huihui Tian
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, Qing Quan Lu, Yantai, Shandong Province, China
| | - Xin Liu
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, Qing Quan Lu, Yantai, Shandong Province, China
| | - Linlin Chen
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, Qing Quan Lu, Yantai, Shandong Province, China
| | - Kun Liu
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, Qing Quan Lu, Yantai, Shandong Province, China
| | - Yishan Wang
- The Center of Non-Traumatic Treatment and Diagnosis of Tumor, Binzhou Medical College affiliated The PLA 107 Hospital, Southern Zhichu Road, Yantai, Shandong Province, China
| | - Hanchen Liu
- The Center of Non-Traumatic Treatment and Diagnosis of Tumor, Binzhou Medical College affiliated The PLA 107 Hospital, Southern Zhichu Road, Yantai, Shandong Province, China
| | - Wenhua Zhang
- The Center of Non-Traumatic Treatment and Diagnosis of Tumor, Binzhou Medical College affiliated The PLA 107 Hospital, Southern Zhichu Road, Yantai, Shandong Province, China
| | - Yukun Guan
- Laboratory of Molecular Pharmacology, School of Pharmacy, Yantai University, Qing Quan Lu, Yantai, Shandong Province, China
| | - Qinwen Wang
- Department of Radiotherapy,307 Hospital of PLA,Academy of Military Medical Science,Beijing,P. R. China
| | - Xiaohang Zhao
- National Laboratory of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, 17 Panjiayuan, Chaoyangqu, Beijing, P. R. China
| | - Xiaochun Wan
- Key Laboratory of Tea Biochemistry & Biotechnology, Ministry of Agriculture, Anhui Agricultural University, Hefei, Anhui Province, China
| |
Collapse
|
20
|
Wei J, Moran T, Zou Z, Qian X, Wang L, Camps C, Hu W, Chaib I, Sanchez B, Xu L, Karachaliou N, Sanchez-Ronco M, Liu B, Rosell R. Customized chemotherapy in metastatic non-small cell lung cancer (NSCLC). Transl Lung Cancer Res 2015; 2:180-8. [PMID: 25806231 DOI: 10.3978/j.issn.2218-6751.2013.02.03] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 02/19/2013] [Indexed: 01/22/2023]
Abstract
Metastatic non-small cell lung cancer (NSCLC) unfortunately remains a lethal disease, despite recent genetic characterization of subclasses of NSCLC, mainly adenocarcinoma, which has led to the development of targeted therapies that improve progression-free survival (PFS). Ultimately, however, patients fatally relapse. In this review we will focus on the search to improve survival for NSCLC patients deemed to be pan-negative for the common driver alterations susceptible to targeted therapy, above all those with EGFR mutations or ALK, ROS or RET translocations. Other uncommon driver mutations such as HER2 and BRAF mutations should be tested in order to rule out targeted treatment before assigning patients to chemotherapy. Chemotherapy yields short lived response with median survival still less than one year. Customized chemotherapy represents one way to attempt to prolong survival, although to date no prospective randomized customized studies have reported sufficient evidence to support this. In one attempt to demonstrate the role of tailoring chemotherapy, the Spanish Lung Cancer Group (SLCG) phase II customized chemotherapy trial (NCT00883480) showed that RAP80, a component of the BRCA1-A complex, influenced outcome in patients with low BRCA1 expression treated with cisplatin/gemcitabine, and in patients with intermediate/high BRCA1 levels receiving cisplatin/docetaxel or docetaxel alone. We are currently performing a prospective, randomized phase III trial comparing non-customized cisplatin/docetaxel with customized therapy in metastatic NSCLC patients (NCT00617656/GECP-BREC) and a parallel phase II study (ChiCTR-TRC-12001860) is being carried out in China (BREC-China) under the auspices of the SLCG.
Collapse
Affiliation(s)
- Jia Wei
- Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Teresa Moran
- Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Ctra Canyet s/n, 08916 Badalona, Spain
| | - Zhengyun Zou
- Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Xiaoping Qian
- Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Lifeng Wang
- Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Carlos Camps
- Hospital General Universitario de Valencia, Av Tres Cruces, s/n, 46014 Valencia, Spain
| | - Wenjing Hu
- Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Imane Chaib
- Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Ctra Canyet s/n, 08916 Badalona, Spain
| | - Belén Sanchez
- Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Ctra Canyet s/n, 08916 Badalona, Spain
| | - Lixia Xu
- Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Niki Karachaliou
- Breakthrough Cancer Research Unit, Pangaea Biotech S.L, Quiron Dexeus University Hospital, Sabino Arana 5-19, 08028 Barcelona
| | - María Sanchez-Ronco
- Universidad Alcalá de Henares, Pza. San Diego s/n, 28801 Alcalá de Henares, Madrid, Spain
| | - Baorui Liu
- Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Rafael Rosell
- Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Ctra Canyet s/n, 08916 Badalona, Spain ; ; Breakthrough Cancer Research Unit, Pangaea Biotech S.L, Quiron Dexeus University Hospital, Sabino Arana 5-19, 08028 Barcelona
| |
Collapse
|
21
|
Apostolou P, Toloudi M, Kourtidou E, Mimikakou G, Vlachou I, Chatziioannou M, Kipourou V, Papasotiriou I. Potential role for the Metnase transposase fusion gene in colon cancer through the regulation of key genes. PLoS One 2014; 9:e109741. [PMID: 25333365 PMCID: PMC4198141 DOI: 10.1371/journal.pone.0109741] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 09/08/2014] [Indexed: 01/06/2023] Open
Abstract
The Metnase fusion gene consists of a SET histone methyltransferase domain and a transposase domain from Mariner transposase. This transposable element is involved in chromosome decatenation, enhances DNA repair, promotes foreign DNA integration, and assists topoisomerase II function. This study investigates the role of Metnase in colon cancer homeostasis and maintenance of the stemness phenotype in colon cancer stem cells (CSCs). Silencing of Metnase was performed in human cancer cell lines before and after treatment with cisplatin, and in colon CSCs. Subsequent changes in the expression of genes involved in repair mechanisms, DNA synthesis, topoisomerase II function, and metastasis as well stemness transcription factors were studied with RT-qPCR experiments. Cellular viability and apoptosis were evaluated by flow cytometry. The results suggest that Metnase influences the expression of many genes involved in the above processes. Furthermore, Metnase levels appear to impact upon expression of NANOG, OCT3/4, and SOX2. Suppression of Metnase also led to an increase in apoptosis. Therefore, Metnase may possess an important role in DNA repair, topoisomerase II function, and the maintenance of stemness during colon cancer development.
Collapse
Affiliation(s)
| | - Maria Toloudi
- Research Genetic Cancer Centre Ltd (R.G.C.C. Ltd), Filotas, Florina, Greece
| | - Eleni Kourtidou
- Research Genetic Cancer Centre Ltd (R.G.C.C. Ltd), Filotas, Florina, Greece
| | - Georgia Mimikakou
- Research Genetic Cancer Centre Ltd (R.G.C.C. Ltd), Filotas, Florina, Greece
| | - Ioanna Vlachou
- Research Genetic Cancer Centre Ltd (R.G.C.C. Ltd), Filotas, Florina, Greece
| | | | - Vasiliki Kipourou
- Research Genetic Cancer Centre Ltd (R.G.C.C. Ltd), Filotas, Florina, Greece
| | - Ioannis Papasotiriou
- Research Genetic Cancer Centre Ltd (R.G.C.C. Ltd), Filotas, Florina, Greece
- * E-mail:
| |
Collapse
|
22
|
Kiang JG, Garrison BR, Smith JT, Fukumoto R. Ciprofloxacin as a potential radio-sensitizer to tumor cells and a radio-protectant for normal cells: differential effects on γ-H2AX formation, p53 phosphorylation, Bcl-2 production, and cell death. Mol Cell Biochem 2014; 393:133-43. [PMID: 24802382 PMCID: PMC4122264 DOI: 10.1007/s11010-014-2053-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 04/03/2014] [Indexed: 02/07/2023]
Abstract
Ionizing radiation increases cell mortality in a dose-dependent manner. Increases in DNA double strand breaks, γ-H2AX, p53 phophorylation, and protein levels of p53 and Bax also occur. We investigated the ability of ciprofloxacin (CIP), a widely prescribed antibiotic, to inhibit DNA damage induced by ionizing radiation. Human tumor TK6, NH32 (p53 (-/-) of TK6) cells, and human normal peripheral blood mononuclear cells (PBMCs) were exposed to 2-8 Gy (60)Co-γ-photon radiation. γ-H2AX (an indicator of DNA strand breaks), phosphorylated p53 (responsible for cell-cycle arrest), Bcl-2 (an apoptotic protein, and cell death were measured. Ionizing irradiation increased γ-H2AX amounts in TK6 cells (p53(+/+)) within 1 h in a radiation dose-dependent manner. CIP pretreatment and posttreatment effectively inhibited the increase in γ-H2AX. CIP pretreatment reduced Bcl-2 production but promoted p53 phosphorylation, caspase-3 activation and cell death. In NH32 cells, CIP failed to significantly inhibit the radiation-induced γ-H2AX increase, suggesting that CIP inhibition involves in p53-dependent mechanisms. In normal healthy human PBMCs, CIP failed to block the radiation-induced γ-H2AX increase but effectively increased Bcl-2 production, but blocked the phospho-p53 increase and subsequent cell death. CIP increased Gadd45α, and enhanced p21 protein 24 h postirradiation. Results suggest that CIP exerts its effect in TK6 cells by promoting p53 phosphorylation and inhibiting Bcl-2 production and in PBMCs by inhibiting p53 phosphorylation and increasing Bcl-2 production. Our data are the first to support the view that CIP may be effective to protect normal tissue cells from radiation injury, while enhancing cancer cell death in radiation therapy.
Collapse
Affiliation(s)
- Juliann G Kiang
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Bethesda, MD, 20889-5603, USA,
| | | | | | | |
Collapse
|
23
|
Williamson EA, Wu Y, Singh S, Byrne M, Wray J, Lee SH, Nickoloff JA, Hromas R. The DNA repair component Metnase regulates Chk1 stability. Cell Div 2014; 9:1. [PMID: 25024738 PMCID: PMC4094783 DOI: 10.1186/1747-1028-9-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 06/09/2014] [Indexed: 12/31/2022] Open
Abstract
Chk1 both arrests replication forks and enhances repair of DNA damage by phosphorylation of downstream effectors. Metnase (also termed SETMAR) is a SET histone methylase and transposase nuclease protein that promotes both DNA double strand break (DSB) repair and re-start of stalled replication forks. We previously found that Chk1 phosphorylation of Metnase on S495 enhanced its DNA DSB repair activity but decreased its ability to re-start stalled replication forks. Here we show that phosphorylated Metnase feeds back to increase the half-life of Chk1. Chk1 half-life is regulated by DDB1 targeting it to Cul4A for ubiquitination and destruction. Metnase decreases Chk1 interaction with DDB1, and decreases Chk1 ubiquitination. These data define a novel pathway for Chk1 regulation, whereby a target of Chk1, Metnase, feeds back to amplify Chk1 stability, and therefore enhance replication fork arrest.
Collapse
Affiliation(s)
- Elizabeth A Williamson
- Department of Medicine, University of Florida College of Medicine, 1600 Archer Rd SW, Gainesville, FL 32610, USA
| | - Yuehan Wu
- Department of Medicine, University of Florida College of Medicine, 1600 Archer Rd SW, Gainesville, FL 32610, USA
| | - Sudha Singh
- Department of Medicine, University of Florida College of Medicine, 1600 Archer Rd SW, Gainesville, FL 32610, USA
| | - Michael Byrne
- Department of Medicine, University of Florida College of Medicine, 1600 Archer Rd SW, Gainesville, FL 32610, USA
| | - Justin Wray
- Department of Medicine, University of Florida College of Medicine, 1600 Archer Rd SW, Gainesville, FL 32610, USA
| | - Suk-Hee Lee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jac A Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Robert Hromas
- Department of Medicine, University of Florida College of Medicine, 1600 Archer Rd SW, Gainesville, FL 32610, USA
| |
Collapse
|
24
|
Wolkowicz U, Morris ER, Robson M, Trubitsyna M, Richardson JM. Structural basis of Mos1 transposase inhibition by the anti-retroviral drug Raltegravir. ACS Chem Biol 2014; 9:743-51. [PMID: 24397848 PMCID: PMC3977574 DOI: 10.1021/cb400791u] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 01/07/2014] [Indexed: 11/28/2022]
Abstract
DNA transposases catalyze the movement of transposons around genomes by a cut-and-paste mechanism related to retroviral integration. Transposases and retroviral integrases share a common RNaseH-like domain with a catalytic DDE/D triad that coordinates the divalent cations required for DNA cleavage and integration. The anti-retroviral drugs Raltegravir and Elvitegravir inhibit integrases by displacing viral DNA ends from the catalytic metal ions. We demonstrate that Raltegravir, but not Elvitegravir, binds to Mos1 transposase in the presence of Mg(2+) or Mn(2+), without the requirement for transposon DNA, and inhibits transposon cleavage and DNA integration in biochemical assays. Crystal structures at 1.7 Å resolution show Raltegravir, in common with integrases, coordinating two Mg(2+) or Mn(2+) ions in the Mos1 active site. However, in the absence of transposon ends, the drug adopts an unusual, compact binding mode distinct from that observed in the active site of the prototype foamy virus integrase.
Collapse
Affiliation(s)
- Urszula
M. Wolkowicz
- School of Biological Sciences, University
of Edinburgh, Mayfield
Road, Edinburgh EH9 3JR, United Kingdom
| | - Elizabeth R. Morris
- School of Biological Sciences, University
of Edinburgh, Mayfield
Road, Edinburgh EH9 3JR, United Kingdom
| | - Michael Robson
- School of Biological Sciences, University
of Edinburgh, Mayfield
Road, Edinburgh EH9 3JR, United Kingdom
| | - Maryia Trubitsyna
- School of Biological Sciences, University
of Edinburgh, Mayfield
Road, Edinburgh EH9 3JR, United Kingdom
| | - Julia M. Richardson
- School of Biological Sciences, University
of Edinburgh, Mayfield
Road, Edinburgh EH9 3JR, United Kingdom
| |
Collapse
|
25
|
Wu Q, Yang Z, Nie Y, Shi Y, Fan D. Multi-drug resistance in cancer chemotherapeutics: mechanisms and lab approaches. Cancer Lett 2014; 347:159-66. [PMID: 24657660 DOI: 10.1016/j.canlet.2014.03.013] [Citation(s) in RCA: 518] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 02/16/2014] [Accepted: 03/11/2014] [Indexed: 12/17/2022]
Abstract
Multi-drug resistance (MDR) has become the largest obstacle to the success of cancer chemotherapies. The mechanisms of MDR and the approaches to test MDR have been discovered, yet not fully understood. This review covers the in vivo and in vitro approaches for the detection of MDR in the laboratory and the mechanisms of MDR in cancers. This study also envisages the future developments toward the clinical and therapeutic applications of MDR in cancer treatment. Future therapeutics for cancer treatment will likely combine the existing therapies with drugs originated from MDR mechanisms such as anti-cancer stem cell drugs, anti-miRNA drugs or anti-epigenetic drugs. The challenges for the clinical detection of MDR will be to find new biomarkers and to determine new evaluation systems before the drug resistance emerges.
Collapse
Affiliation(s)
- Qiong Wu
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Zhiping Yang
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yongquan Shi
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
26
|
Jeyaratnam DC, Baduin BS, Hansen MC, Hansen M, Jørgensen JM, Aggerholm A, Ommen HB, Hokland P, Nyvold CG. Delineation of known and new transcript variants of the SETMAR (Metnase) gene and the expression profile in hematologic neoplasms. Exp Hematol 2014; 42:448-56.e4. [PMID: 24607956 DOI: 10.1016/j.exphem.2014.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 02/11/2014] [Accepted: 02/25/2014] [Indexed: 12/23/2022]
Abstract
SET domain and mariner transposase fusion gene (SETMAR), also known as Metnase, has previously been shown to suppress the formation of chromosomal translocation in mouse fibroblasts. Despite the fact that hematologic malignancies are often characterized by chromosomal rearrangements, no studies have hitherto investigated the expression pattern of the gene in these disorders. We hypothesized that a high expression of SETMAR protected the cells from chromosomal rearrangements; thus, we examined the mRNA expression of SETMAR transcript variants in hematologic patients. We identified six transcript variants (var1, var2, var5, varA, varB, varC), of which three had not been reported previously. Expression levels were quantified by transcript-specific quantitative polymerase chain reaction in 15 healthy individuals, 70 acute myeloid leukemia (AML) patients (translocation positive, n= 30 [AML(TPos)], translocation negative, n = 40 [AML(TNeg)]), seven patients with mantle cell lymphoma (t [11,14] positive), and 13 patients with chronic myeloid leukemia (t [9,22] positive). All variants were significantly overexpressed in both subgroups of AML compared with healthy individuals (var1 and var2: p < 0.00001 for both AML subgroups, varA and varB: p = 0.0002, var5: p = 0.0008, and varC: p = 0.0001 for AML(TNeg); varA: p = 0.0048, varB and var5: p = 0.0001, varC: p = 0.0017). When comparing the expression in AML(TNeg) and AML(TPos), we found a significantly increased expression of the full length SETMAR in AML(TNeg) (var1: p = 0.047), suggesting a protective effect of high SETMAR expression on formation of chromosomal translocations. In conclusion, we have found known and novel SETMAR splice variants to be significantly increased in AML. To our knowledge, this is the first study that describes an expression profile of SETMAR in subgroups of hematologic malignancies, which can be linked to the incidence of chromosomal rearrangements.
Collapse
Affiliation(s)
| | | | | | - Maria Hansen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Anni Aggerholm
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Hans Beier Ommen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Hokland
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
27
|
Kim HS, Chen Q, Kim SK, Nickoloff JA, Hromas R, Georgiadis MM, Lee SH. The DDN catalytic motif is required for Metnase functions in non-homologous end joining (NHEJ) repair and replication restart. J Biol Chem 2014; 289:10930-10938. [PMID: 24573677 PMCID: PMC4036204 DOI: 10.1074/jbc.m113.533216] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Metnase (or SETMAR) arose from a chimeric fusion of the Hsmar1 transposase downstream of a protein methylase in anthropoid primates. Although the Metnase transposase domain has been largely conserved, its catalytic motif (DDN) differs from the DDD motif of related transposases, which may be important for its role as a DNA repair factor and its enzymatic activities. Here, we show that substitution of DDN610 with either DDD610 or DDE610 significantly reduced in vivo functions of Metnase in NHEJ repair and accelerated restart of replication forks. We next tested whether the DDD or DDE mutants cleave single-strand extensions and flaps in partial duplex DNA and pseudo-Tyr structures that mimic stalled replication forks. Neither substrate is cleaved by the DDD or DDE mutant, under the conditions where wild-type Metnase effectively cleaves ssDNA overhangs. We then characterized the ssDNA-binding activity of the Metnase transposase domain and found that the catalytic domain binds ssDNA but not dsDNA, whereas dsDNA binding activity resides in the helix-turn-helix DNA binding domain. Substitution of Asn-610 with either Asp or Glu within the transposase domain significantly reduces ssDNA binding activity. Collectively, our results suggest that a single mutation DDN610 → DDD610, which restores the ancestral catalytic site, results in loss of function in Metnase.
Collapse
Affiliation(s)
- Hyun-Suk Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Qiujia Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Sung-Kyung Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Jac A Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado 80523
| | - Robert Hromas
- Department of Medicine, University of Florida and Shands Health Care System, Gainesville, Florida 32610
| | - Millie M Georgiadis
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Chemistry and Chemical Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana 46202
| | - Suk-Hee Lee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202.
| |
Collapse
|
28
|
Pflieger A, Waffo Teguo P, Papastamoulis Y, Chaignepain S, Subra F, Munir S, Delelis O, Lesbats P, Calmels C, Andreola ML, Merillon JM, Auge-Gouillou C, Parissi V. Natural stilbenoids isolated from grapevine exhibiting inhibitory effects against HIV-1 integrase and eukaryote MOS1 transposase in vitro activities. PLoS One 2013; 8:e81184. [PMID: 24312275 PMCID: PMC3842960 DOI: 10.1371/journal.pone.0081184] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 10/18/2013] [Indexed: 01/07/2023] Open
Abstract
Polynucleotidyl transferases are enzymes involved in several DNA mobility mechanisms in prokaryotes and eukaryotes. Some of them such as retroviral integrases are crucial for pathogenous processes and are therefore good candidates for therapeutic approaches. To identify new therapeutic compounds and new tools for investigating the common functional features of these proteins, we addressed the inhibition properties of natural stilbenoids deriving from resveratrol on two models: the HIV-1 integrase and the eukaryote MOS-1 transposase. Two resveratrol dimers, leachianol F and G, were isolated for the first time in Vitis along with fourteen known stilbenoids: E-resveratrol, E-piceid, E-pterostilbene, E-piceatannol, (+)-E-ε-viniferin, E-ε-viniferinglucoside, E-scirpusin A, quadragularin A, ampelopsin A, pallidol, E-miyabenol C, E-vitisin B, hopeaphenol, and isohopeaphenol and were purified from stalks of Vitis vinifera (Vitaceae), and moracin M from stem bark of Milliciaexelsa (Moraceae). These compounds were tested in in vitro and in vivo assays reproducing the activity of both enzymes. Several molecules presented significant inhibition on both systems. Some of the molecules were found to be active against both proteins while others were specific for one of the two models. Comparison of the differential effects of the molecules suggested that the compounds could target specific intermediate nucleocomplexes of the reactions. Additionally E-pterostilbene was found active on the early lentiviral replication steps in lentiviruses transduced cells. Consequently, in addition to representing new original lead compounds for further modelling of new active agents against HIV-1 integrase, these molecules could be good tools for identifying such reaction intermediates in DNA mobility processes.
Collapse
Affiliation(s)
- Aude Pflieger
- Université François Rabelais de Tours, EA 6306, UFR Sciences Pharmaceutiques, Parc Grandmont, Tours, France
| | - Pierre Waffo Teguo
- Groupe d'Etude des Substances Végétales à Activité Biologique, EA 3675 - UFR Pharmacie, Université Bordeaux Segalen, Institut des Sciences de la Vigne et du Vin (ISVV), Bordeaux, France
| | - Yorgos Papastamoulis
- Groupe d'Etude des Substances Végétales à Activité Biologique, EA 3675 - UFR Pharmacie, Université Bordeaux Segalen, Institut des Sciences de la Vigne et du Vin (ISVV), Bordeaux, France
| | - Stéphane Chaignepain
- Plateforme Protéome - Centre Génomique Fonctionnelle, UMR 5248 CBMN, Université Bordeaux Segalen, Bordeaux France
| | | | | | | | - Paul Lesbats
- Université François Rabelais de Tours, EA 6306, UFR Sciences Pharmaceutiques, Parc Grandmont, Tours, France
- Cancer Research UK, London Research Institute, Clare Hall Laboratories, Potters Bar, United Kingdom
| | - Christina Calmels
- Laboratoire MFP, UMR 5234-CNRS, Université Bordeaux Segalen, Bordeaux, France
| | - Marie-Line Andreola
- Laboratoire MFP, UMR 5234-CNRS, Université Bordeaux Segalen, Bordeaux, France
| | - Jean-Michel Merillon
- Groupe d'Etude des Substances Végétales à Activité Biologique, EA 3675 - UFR Pharmacie, Université Bordeaux Segalen, Institut des Sciences de la Vigne et du Vin (ISVV), Bordeaux, France
| | - Corinne Auge-Gouillou
- Université François Rabelais de Tours, EA 6306, UFR Sciences Pharmaceutiques, Parc Grandmont, Tours, France
| | - Vincent Parissi
- Laboratoire MFP, UMR 5234-CNRS, Université Bordeaux Segalen, Bordeaux, France
- * E-mail:
| |
Collapse
|
29
|
Métifiot M, Marchand C, Pommier Y. HIV integrase inhibitors: 20-year landmark and challenges. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2013; 67:75-105. [PMID: 23885999 DOI: 10.1016/b978-0-12-405880-4.00003-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Since the discovery of HIV as the cause for AIDS 30 years ago, major progress has been made, including the discovery of drugs that now control the disease. Here, we review the integrase (IN) inhibitors from the discovery of the first compounds 20 years ago to the approval of two highly effective IN strand transfer inhibitors (INSTIs), raltegravir (Isentress) and elvitegravir (Stribild), and the promising clinical activity of dolutegravir. After summarizing the molecular mechanism of action of the INSTIs as interfacial inhibitors, we discuss the remaining challenges. Those include: overcoming resistance to clinical INSTIs, long-term safety of INSTIs, cost of therapy, place of the INSTIs in prophylactic treatments, and the development of new classes of inhibitors (the LEDGINs) targeting IN outside its catalytic site. We also discuss the role of chromatin and host DNA repair factor for the completion of integration.
Collapse
Affiliation(s)
- Mathieu Métifiot
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | |
Collapse
|