601
|
|
602
|
Xu B, Ding J, Chen KX, Miao ZH, Huang H, Liu H, Luo XM. Advances in Cancer Chemotherapeutic Drug Research in China. RECENT ADVANCES IN CANCER RESEARCH AND THERAPY 2012. [PMCID: PMC7158183 DOI: 10.1016/b978-0-12-397833-2.00012-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Bin Xu
- Corresponding author: Bin Xu, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zi Road, Shanghai 201203, People’s Republic of China. Tel: O21-54920515 (o), 13501793936 (mobile), Fax: 021-54920568, e-mail:
| | | | | | | | | | | | | |
Collapse
|
603
|
Homologue pairing in flies and mammals: gene regulation when two are involved. GENETICS RESEARCH INTERNATIONAL 2011; 2012:430587. [PMID: 22567388 PMCID: PMC3335585 DOI: 10.1155/2012/430587] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 09/17/2011] [Accepted: 09/26/2011] [Indexed: 01/03/2023]
Abstract
Chromosome pairing is usually discussed in the context of meiosis. Association of homologues in germ cells enables chromosome segregation and is necessary for fertility. A few organisms, such as flies, also pair their entire genomes in somatic cells. Most others, including mammals, display little homologue pairing outside of the germline. Experimental evidence from both flies and mammals suggests that communication between homologues contributes to normal genome regulation. This paper will contrast the role of pairing in transmitting information between homologues in flies and mammals. In mammals, somatic homologue pairing is tightly regulated, occurring at specific loci and in a developmentally regulated fashion. Inappropriate pairing, or loss of normal pairing, is associated with gene misregulation in some disease states. While homologue pairing in flies is capable of influencing gene expression, the significance of this for normal expression remains unknown. The sex chromosomes pose a particularly interesting situation, as females are able to pair X chromosomes, but males cannot. The contribution of homologue pairing to the biology of the X chromosome will also be discussed.
Collapse
|
604
|
Cytotoxicity and cell death mechanisms induced by the polyamine-vectorized anti-cancer drug F14512 targeting topoisomerase II. Biochem Pharmacol 2011; 82:1843-52. [DOI: 10.1016/j.bcp.2011.08.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 08/30/2011] [Accepted: 08/31/2011] [Indexed: 11/24/2022]
|
605
|
Sapetto-Rebow B, McLoughlin SC, O'Shea LC, O'Leary O, Willer JR, Alvarez Y, Collery R, O'Sullivan J, Van Eeden F, Hensey C, Kennedy BN. Maternal topoisomerase II alpha, not topoisomerase II beta, enables embryonic development of zebrafish top2a-/- mutants. BMC DEVELOPMENTAL BIOLOGY 2011; 11:71. [PMID: 22111588 PMCID: PMC3287258 DOI: 10.1186/1471-213x-11-71] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 11/23/2011] [Indexed: 11/10/2022]
Abstract
BACKGROUND Genetic alterations in human topoisomerase II alpha (TOP2A) are linked to cancer susceptibility. TOP2A decatenates chromosomes and thus is necessary for multiple aspects of cell division including DNA replication, chromosome condensation and segregation. Topoisomerase II alpha is also required for embryonic development in mammals, as mouse Top2a knockouts result in embryonic lethality as early as the 4-8 cell stage. The purpose of this study was to determine whether the extended developmental capability of zebrafish top2a mutants arises from maternal expression of top2a or compensation from its top2b paralogue. RESULTS Here, we describe bloody minded (blm), a novel mutant of zebrafish top2a. In contrast to mouse Top2a nulls, zebrafish top2a mutants survive to larval stages (4-5 day post fertilization). Developmental analyses demonstrate abundant expression of maternal top2a but not top2b. Inhibition or poisoning of maternal topoisomerase II delays embryonic development by extending the cell cycle M-phase. Zygotic top2a and top2b are co-expressed in the zebrafish CNS, but endogenous or ectopic top2b RNA appear unable to prevent the blm phenotype. CONCLUSIONS We conclude that maternal top2a enables zebrafish development before the mid-zygotic transition (MZT) and that zebrafish top2a and top2b are not functionally redundant during development after activation of the zygotic genome.
Collapse
Affiliation(s)
- Beata Sapetto-Rebow
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
606
|
Ponder J, Yoo BH, Abraham AD, Li Q, Ashley AK, Amerin CL, Zhou Q, Reid BG, Reigan P, Hromas R, Nickoloff JA, LaBarbera DV. Neoamphimedine circumvents metnase-enhanced DNA topoisomerase IIα activity through ATP-competitive inhibition. Mar Drugs 2011; 9:2397-2408. [PMID: 22163192 PMCID: PMC3229241 DOI: 10.3390/md9112397] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 11/08/2011] [Accepted: 11/09/2011] [Indexed: 12/13/2022] Open
Abstract
Type IIα DNA topoisomerase (TopoIIα) is among the most important clinical drug targets for the treatment of cancer. Recently, the DNA repair protein Metnase was shown to enhance TopoIIα activity and increase resistance to TopoIIα poisons. Using in vitro DNA decatenation assays we show that neoamphimedine potently inhibits TopoIIα-dependent DNA decatenation in the presence of Metnase. Cell proliferation assays demonstrate that neoamphimedine can inhibit Metnase-enhanced cell growth with an IC50 of 0.5 μM. Additionally, we find that the apparent Km of TopoIIα for ATP increases linearly with higher concentrations of neoamphimedine, indicating ATP-competitive inhibition, which is substantiated by molecular modeling. These findings support the continued development of neoamphimedine as an anticancer agent, particularly in solid tumors that over-express Metnase.
Collapse
Affiliation(s)
- Jessica Ponder
- Department of Pharmaceutical Sciences, The University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO 80045, USA; E-Mails: (J.P.); (B.H.Y.); (A.D.A.); (Q.L.); (Q.Z.); (B.G.R.); (P.R.)
| | - Byong Hoon Yoo
- Department of Pharmaceutical Sciences, The University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO 80045, USA; E-Mails: (J.P.); (B.H.Y.); (A.D.A.); (Q.L.); (Q.Z.); (B.G.R.); (P.R.)
| | - Adedoyin D. Abraham
- Department of Pharmaceutical Sciences, The University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO 80045, USA; E-Mails: (J.P.); (B.H.Y.); (A.D.A.); (Q.L.); (Q.Z.); (B.G.R.); (P.R.)
| | - Qun Li
- Department of Pharmaceutical Sciences, The University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO 80045, USA; E-Mails: (J.P.); (B.H.Y.); (A.D.A.); (Q.L.); (Q.Z.); (B.G.R.); (P.R.)
| | - Amanda K. Ashley
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; E-Mails: (A.K.A.); (C.L.A.); (J.A.N.)
| | - Courtney L. Amerin
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; E-Mails: (A.K.A.); (C.L.A.); (J.A.N.)
| | - Qiong Zhou
- Department of Pharmaceutical Sciences, The University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO 80045, USA; E-Mails: (J.P.); (B.H.Y.); (A.D.A.); (Q.L.); (Q.Z.); (B.G.R.); (P.R.)
| | - Brian G. Reid
- Department of Pharmaceutical Sciences, The University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO 80045, USA; E-Mails: (J.P.); (B.H.Y.); (A.D.A.); (Q.L.); (Q.Z.); (B.G.R.); (P.R.)
| | - Philip Reigan
- Department of Pharmaceutical Sciences, The University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO 80045, USA; E-Mails: (J.P.); (B.H.Y.); (A.D.A.); (Q.L.); (Q.Z.); (B.G.R.); (P.R.)
| | - Robert Hromas
- Department of Medicine, University of Florida and Shands Health Care System, Gainesville, FL 32610, USA; E-Mail:
| | - Jac A. Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; E-Mails: (A.K.A.); (C.L.A.); (J.A.N.)
| | - Daniel V. LaBarbera
- Department of Pharmaceutical Sciences, The University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO 80045, USA; E-Mails: (J.P.); (B.H.Y.); (A.D.A.); (Q.L.); (Q.Z.); (B.G.R.); (P.R.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-303-724-4116; Fax: +1-303-724-7266
| |
Collapse
|
607
|
Design, synthesis, and biological evaluation of a novel series of bisintercalating DNA-binding piperazine-linked bisanthrapyrazole compounds as anticancer agents. Bioorg Med Chem 2011; 19:7023-32. [PMID: 22041173 DOI: 10.1016/j.bmc.2011.10.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 09/23/2011] [Accepted: 10/05/2011] [Indexed: 12/13/2022]
Abstract
A series of bisintercalating DNA binding bisanthrapyrazole compounds containing piperazine linkers were designed by molecular modeling and docking techniques. Because the anthrapyrazoles are not quinones they are unable to be reductively activated like doxorubicin and other anthracyclines and thus they should not be cardiotoxic. The concentration dependent increase in DNA melting temperature was used to determine the strength of DNA binding and the bisintercalation potential of the compounds. Compounds with more than a three-carbon linker that could span four DNA base pairs achieved bisintercalation. All of the bisanthrapyrazoles inhibited human erythroleukemic K562 cell growth in the low to submicromolar concentration range. They also strongly inhibited the decatenation activity of topoisomerase IIα and the relaxation activity of topoisomerase I. However, as measured by their ability to induce double strand breaks in plasmid DNA, the bisanthrapyrazole compounds did not act as topoisomerase IIα poisons. In conclusion, a novel group of bisanthrapyrazole compounds were designed, synthesized, and biologically evaluated as potential anticancer agents.
Collapse
|
608
|
García-Rubio ML, Aguilera A. Topological constraints impair RNA polymerase II transcription and causes instability of plasmid-borne convergent genes. Nucleic Acids Res 2011; 40:1050-64. [PMID: 21998294 PMCID: PMC3273821 DOI: 10.1093/nar/gkr840] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Despite the theoretical bases for the association of topoisomerases and supercoiling changes with transcription and replication, our knowledge of the impact of topological constraints on transcription and replication is incomplete. Although mutation of topoisomerases affects expression and stability of the rDNA region it is not clear whether the same is the case for RNAPII transcription and genome integrity in other regions. We developed new assays in which two convergent RNAPII-driven genes are transcribed simultaneously. Plasmid-based systems were constructed with and without a transcription terminator between the two convergent transcription units, so that the impact of transcription interference could also be evaluated. Using these assays we show that Topos I and II play roles in RNAPII transcription in vivo and reduce the stability of RNAPII-transcribed genes in Saccharomyces cerevisiae. Supercoiling accumulation in convergent transcription units impairs RNAPII transcription in top1Δ strains, but Topo II is also required for efficient transcription independent of Topo I and of detectable supercoiling accumulation. Our work shows that topological constraints negatively affect RNAPII transcription and genetic integrity, and provides an assay to study gene regulation by transcription interference.
Collapse
Affiliation(s)
- María L García-Rubio
- Centro Andaluz de Biología Molecular y Medicina Regenerativa CABIMER, Universidad de Sevilla-CSIC, Av. Américo Vespucio s/n 41092 Sevilla, and Departamento de Genética, Universidad de Sevilla, 41012 Sevilla, Spain
| | | |
Collapse
|
609
|
Casu L, Cottiglia F, Leonti M, De Logu A, Agus E, Tse-Dinh YC, Lombardo V, Sissi C. Ungeremine effectively targets mammalian as well as bacterial type I and type II topoisomerases. Bioorg Med Chem Lett 2011; 21:7041-4. [PMID: 22014547 DOI: 10.1016/j.bmcl.2011.09.097] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 09/22/2011] [Accepted: 09/23/2011] [Indexed: 11/16/2022]
Abstract
From the methanol extract of the bulbs of Pancratium illyricum L., three phenanthridine type alkaloids, ungeremine (1), (-)-lycorine (2) and (+)-vittatine (3) were isolated. For the evaluation of their anticancer and antibacterial potential, compounds 1-3 were tested against human (I, IIα) and bacterial (IA, IV) topoisomerases. Our data demonstrated that ungeremine impairs the activity of both, human and bacterial topoisomerases. Remarkably, ungeremine was found to largely increments the DNA cleavage promoted by bacterial topoisomerase IA, a new target in antimicrobial chemotherapy.
Collapse
Affiliation(s)
- Laura Casu
- Dipartimento Farmaco Chimico Tecnologico, University of Cagliari, Cagliari, Italy
| | | | | | | | | | | | | | | |
Collapse
|
610
|
Li C, Sun SY, Khuri FR, Li R. Pleiotropic functions of EAPII/TTRAP/TDP2: cancer development, chemoresistance and beyond. Cell Cycle 2011; 10:3274-83. [PMID: 21926483 DOI: 10.4161/cc.10.19.17763] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
EAPII (also called TTRAP, TDP2), a protein identified a decade ago, has recently been shown to function as an oncogenic factor. This protein was also proven to be the first 5'- tyrosyl-DNA phosphodiesterase. EAPII has been demonstrated to have promiscuous protein associations, broad responsiveness to various extracellular signals, and pleiotropic functions in the development of human diseases including cancer and neurodegenerative disease. Emerging data suggest that EAPII is a multi-functional protein: EAPII repairs enzyme (topoisomerase)-mediated DNA damage by removing phosphotyrosine from DNA adducts; EAPII is involved in multiple signal transduction pathways such as TNF-TNFR, TGFβ and MAPK, and EAPII is responsive to immune defense, inflammatory response, virus infection and DNA toxins (chemo or radiation therapy). This review focuses on the current understanding of EAPII biology and its potential relations to many aspects of cancer development, including chromosome instability, tumorigenesis, tumor metastasis and chemoresistance, suggesting it as a potential target for intervention in cancer and other human diseases.
Collapse
Affiliation(s)
- Chunyang Li
- Winship Cancer Institute, Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, GA, USA
| | | | | | | |
Collapse
|
611
|
Morgan MA, Onono FO, Spielmann HP, Subramanian T, Scherr M, Venturini L, Dallmann I, Ganser A, Reuter CWM. Modulation of anthracycline-induced cytotoxicity by targeting the prenylated proteome in myeloid leukemia cells. J Mol Med (Berl) 2011; 90:149-61. [PMID: 21915711 DOI: 10.1007/s00109-011-0814-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 08/14/2011] [Accepted: 08/25/2011] [Indexed: 12/13/2022]
Abstract
Deregulation of Ras/ERK signaling in myeloid leukemias makes this pathway an interesting target for drug development. Myeloid leukemia cell lines were screened for idarubicin-induced apoptosis, cell-cycle progression, cell-cycle-dependent MAP kinase kinase (MEK-1/2) activation, and Top2 expression. Cell-cycle-dependent activation of MEK/ERK signaling was blocked using farnesyltransferase inhibitor (FTI) BMS-214,662 and dual prenyltransferase inhibitor (DPI) L-778,123 to disrupt Ras signaling. Idarubicin caused a G2/M cell-cycle arrest characterized by elevated diphosphorylated MEK-1/2 and Top2α expression levels. The FTI/DPIs elicited distinct effects on Ras signaling, protein prenylation, cell cycling and apoptosis. Combining these FTI/DPIs with idarubicin synergistically inhibited proliferation of leukemia cell lines, but the L-778,123+idarubicin combination exhibited synergistic growth inhibition over a greater range of drug concentrations. Interestingly, combined FTI/DPI treatment synergistically inhibited cell proliferation, induced apoptosis and nearly completely blocked protein prenylation. Inhibition of K-Ras expression by RNA interference or blockade of its post-translational prenylation led to increased BMS-214,662-induced apoptosis. Our results suggest that nearly complete inhibition of protein prenylation using an FTI + DPI combination is the most effective method to induce apoptosis and to block anthracycline-induced activation of ERK signaling.
Collapse
Affiliation(s)
- Michael A Morgan
- Department of Hematology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625, Hannover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
612
|
Srikantan S, Abdelmohsen K, Lee EK, Tominaga K, Subaran SS, Kuwano Y, Kulshrestha R, Panchakshari R, Kim HH, Yang X, Martindale JL, Marasa BS, Kim MM, Wersto RP, Indig FE, Chowdhury D, Gorospe M. Translational control of TOP2A influences doxorubicin efficacy. Mol Cell Biol 2011; 31:3790-801. [PMID: 21768308 PMCID: PMC3165726 DOI: 10.1128/mcb.05639-11] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 07/05/2011] [Indexed: 12/15/2022] Open
Abstract
The cellular abundance of topoisomerase IIα (TOP2A) critically maintains DNA topology after replication and determines the efficacy of TOP2 inhibitors in chemotherapy. Here, we report that the RNA-binding protein HuR, commonly overexpressed in cancers, binds to the TOP2A 3'-untranslated region (3'UTR) and increases TOP2A translation. Reducing HuR levels triggered the recruitment of TOP2A transcripts to RNA-induced silencing complex (RISC) components and to cytoplasmic processing bodies. Using a novel MS2-tagged RNA precipitation method, we identified microRNA miR-548c-3p as a mediator of these effects and further uncovered that the interaction of miR-548c-3p with the TOP2A 3'UTR repressed TOP2A translation by antagonizing the action of HuR. Lowering TOP2A by silencing HuR or by overexpressing miR-548c-3p selectively decreased DNA damage after treatment with the chemotherapeutic agent doxorubicin. In sum, HuR enhances TOP2A translation by competing with miR-548c-3p; their combined actions control TOP2A expression levels and determine the effectiveness of doxorubicin.
Collapse
MESH Headings
- 3' Untranslated Regions
- Antibiotics, Antineoplastic/pharmacology
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/genetics
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- DNA/metabolism
- DNA Damage/drug effects
- DNA Topoisomerases, Type II/biosynthesis
- DNA Topoisomerases, Type II/genetics
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- Doxorubicin/pharmacology
- ELAV Proteins
- ELAV-Like Protein 1
- Gene Expression Regulation
- HeLa Cells
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Poly-ADP-Ribose Binding Proteins
- Protein Biosynthesis
- RNA Interference
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering
- RNA-Binding Proteins/antagonists & inhibitors
- RNA-Binding Proteins/biosynthesis
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- RNA-Induced Silencing Complex/metabolism
Collapse
Affiliation(s)
- Subramanya Srikantan
- Laboratory of Molecular Biology and Immunology, NIA-IRP, NIH, Baltimore, Maryland 21224
| | - Kotb Abdelmohsen
- Laboratory of Molecular Biology and Immunology, NIA-IRP, NIH, Baltimore, Maryland 21224
| | - Eun Kyung Lee
- Laboratory of Molecular Biology and Immunology, NIA-IRP, NIH, Baltimore, Maryland 21224
| | - Kumiko Tominaga
- Laboratory of Molecular Biology and Immunology, NIA-IRP, NIH, Baltimore, Maryland 21224
| | - Sarah S. Subaran
- Research Resources Branch, NIA-IRP, NIH, Baltimore, Maryland 21224
| | - Yuki Kuwano
- Laboratory of Molecular Biology and Immunology, NIA-IRP, NIH, Baltimore, Maryland 21224
| | - Ritu Kulshrestha
- Dana-Farber Cancer Institute, Department of Radiation Oncology, Harvard Medical School, Boston, Massachusetts 02118
| | - Rohit Panchakshari
- Dana-Farber Cancer Institute, Department of Radiation Oncology, Harvard Medical School, Boston, Massachusetts 02118
| | - Hyeon Ho Kim
- Laboratory of Molecular Biology and Immunology, NIA-IRP, NIH, Baltimore, Maryland 21224
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Xiaoling Yang
- Laboratory of Molecular Biology and Immunology, NIA-IRP, NIH, Baltimore, Maryland 21224
| | | | - Bernard S. Marasa
- Laboratory of Molecular Biology and Immunology, NIA-IRP, NIH, Baltimore, Maryland 21224
| | - Mihee M. Kim
- Laboratory of Molecular Biology and Immunology, NIA-IRP, NIH, Baltimore, Maryland 21224
| | - Robert P. Wersto
- Research Resources Branch, NIA-IRP, NIH, Baltimore, Maryland 21224
| | - Fred E. Indig
- Research Resources Branch, NIA-IRP, NIH, Baltimore, Maryland 21224
| | - Dipanjan Chowdhury
- Dana-Farber Cancer Institute, Department of Radiation Oncology, Harvard Medical School, Boston, Massachusetts 02118
| | - Myriam Gorospe
- Laboratory of Molecular Biology and Immunology, NIA-IRP, NIH, Baltimore, Maryland 21224
| |
Collapse
|
613
|
Qi X, Hou S, Lepp A, Li R, Basir Z, Lou Z, Chen G. Phosphorylation and stabilization of topoisomerase IIα protein by p38γ mitogen-activated protein kinase sensitize breast cancer cells to its poisons. J Biol Chem 2011; 286:35883-35890. [PMID: 21878638 DOI: 10.1074/jbc.m111.229260] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Cancer drugs suppress tumor cell growth by inhibiting specific cellular targets. However, most drugs also activate several cellular nonspecific stress pathways, and the implications of these off-target effects are mostly unknown. Here, we report that p38γ, but not p38α, MAPK is specifically activated by treatment of breast cancer cells with topoisomerase II (Topo II) drugs, whereas paclitaxel (Taxol) does not have this effect. The activated p38γ in turn phosphorylates and stabilizes Topo IIα protein, and this enhances the growth inhibition by Topo II drugs. Moreover, p38γ activity was shown to be necessary and sufficient for Topo IIα expression, the drug-p38γ-Topo IIα axis is only detected in intrinsically sensitive but not resistant cells, and p38γ is co-overexpressed with Topo IIα protein in primary breast cancers. These results reveal a new paradigm in which p38γ actively regulates the drug-Topo IIα signal transduction, and this may be exploited to increase the therapeutic activity of Topo II drugs.
Collapse
Affiliation(s)
- Xiaomei Qi
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Songwang Hou
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Adrienne Lepp
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Rongshan Li
- Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Zainab Basir
- Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic and Foundation, Rochester, Minnesota 55905
| | - Guan Chen
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226; Department of Research Services, Zablocki Veterans Affairs Medical Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226.
| |
Collapse
|
614
|
Chelouah S, Monod-Wissler C, Bailly C, Barret JM, Guilbaud N, Vispé S, Käs E. An integrated Drosophila model system reveals unique properties for F14512, a novel polyamine-containing anticancer drug that targets topoisomerase II. PLoS One 2011; 6:e23597. [PMID: 21853156 PMCID: PMC3154508 DOI: 10.1371/journal.pone.0023597] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 07/21/2011] [Indexed: 11/19/2022] Open
Abstract
F14512 is a novel anti-tumor molecule based on an epipodophyllotoxin core coupled to a cancer-cell vectoring spermine moiety. This polyamine linkage is assumed to ensure the preferential uptake of F14512 by cancer cells, strong interaction with DNA and potent inhibition of topoisomerase II (Topo II). The antitumor activity of F14512 in human tumor models is significantly higher than that of other epipodophyllotoxins in spite of a lower induction of DNA breakage. Hence, the demonstrated superiority of F14512 over other Topo II poisons might not result solely from its preferential uptake by cancer cells, but could also be due to unique effects on Topo II interactions with DNA. To further dissect the mechanism of action of F14512, we used Drosophila melanogaster mutants whose genetic background leads to an easily scored phenotype that is sensitive to changes in Topo II activity and/or localization. F14512 has antiproliferative properties in Drosophila cells and stabilizes ternary Topo II/DNA cleavable complexes at unique sites located in moderately repeated sequences, suggesting that the drug specifically targets a select and limited subset of genomic sequences. Feeding F14512 to developing mutant Drosophila larvae led to the recovery of flies expressing a striking phenotype, "Eye wide shut," where one eye is replaced by a first thoracic segment. Other recovered F14512-induced gain- and loss-of-function phenotypes similarly correspond to precise genetic dysfunctions. These complex in vivo results obtained in a whole developing organism can be reconciled with known genetic anomalies and constitute a remarkable instance of specific alterations of gene expression by ingestion of a drug. "Drosophila-based anticancer pharmacology" hence reveals unique properties for F14512, demonstrating the usefulness of an assay system that provides a low-cost, rapid and effective complement to mammalian models and permits the elucidation of fundamental mechanisms of action of candidate drugs of therapeutic interest in humans.
Collapse
Affiliation(s)
- Sonia Chelouah
- Université de Toulouse, UPS, Université Paul Sabatier, Laboratoire de Biologie Moléculaire Eucaryote; Toulouse; France
- CNRS, Centre National de la Recherche Scientifique, UMR5099, Laboratoire de Biologie Moléculaire Eucaryote, Toulouse, France
| | - Caroline Monod-Wissler
- Université de Toulouse, UPS, Université Paul Sabatier, Laboratoire de Biologie Moléculaire Eucaryote; Toulouse; France
- CNRS, Centre National de la Recherche Scientifique, UMR5099, Laboratoire de Biologie Moléculaire Eucaryote, Toulouse, France
| | - Christian Bailly
- Centre de Recherche en Oncologie Expérimentale, Institut de Recherche Pierre Fabre, Toulouse, France
| | - Jean-Marc Barret
- Centre de Recherche en Oncologie Expérimentale, Institut de Recherche Pierre Fabre, Toulouse, France
| | - Nicolas Guilbaud
- Centre de Recherche en Oncologie Expérimentale, Institut de Recherche Pierre Fabre, Toulouse, France
| | - Stéphane Vispé
- Centre de Recherche en Oncologie Expérimentale, Institut de Recherche Pierre Fabre, Toulouse, France
- * E-mail: (EK); (SV)
| | - Emmanuel Käs
- Université de Toulouse, UPS, Université Paul Sabatier, Laboratoire de Biologie Moléculaire Eucaryote; Toulouse; France
- CNRS, Centre National de la Recherche Scientifique, UMR5099, Laboratoire de Biologie Moléculaire Eucaryote, Toulouse, France
- * E-mail: (EK); (SV)
| |
Collapse
|
615
|
Ferlin MG, Gia O, Dalla Via L. Synthesis and Antiproliferative Activity of Some Ellipticine-Like 11H-Pyrido[a]carbazole Derivatives. ChemMedChem 2011; 6:1872-83. [DOI: 10.1002/cmdc.201100233] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 07/11/2011] [Indexed: 11/05/2022]
|
616
|
Chou SM, Huang TH, Chen HC, Li TK. Calcium-induced cleavage of DNA topoisomerase I involves the cytoplasmic-nuclear shuttling of calpain 2. Cell Mol Life Sci 2011; 68:2769-84. [PMID: 21086148 PMCID: PMC11114693 DOI: 10.1007/s00018-010-0591-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 10/29/2010] [Accepted: 11/02/2010] [Indexed: 12/18/2022]
Abstract
Important to the function of calpains is temporal and spatial regulation of their proteolytic activity. Here, we demonstrate that cytoplasm-resident calpain 2 cleaves human nuclear topoisomerase I (hTOP1) via Ca(2+)-activated proteolysis and nucleoplasmic shuttling of proteases. This proteolysis of hTOP1 was induced by either ionomycin-caused Ca(2+) influx or addition of Ca(2+) in cellular extracts. Ca(2+) failed to induce hTOP1 proteolysis in calpain 2-knockdown cells. Moreover, calpain 2 cleaved hTOP1 in vitro. Furthermore, calpain 2 entered the nucleus upon Ca(2+) influx, and calpastatin interfered with this process. Calpain 2 cleavage sites were mapped at K(158) and K(183) of hTOP1. Calpain 2-truncated hTOP1 exhibited greater relaxation activity but remained able to interact with nucleolin and to form cleavable complexes. Interestingly, calpain 2 appears to be involved in ionomycin-induced protection from camptothecin-induced cytotoxicity. Thus, our data suggest that nucleocytoplasmic shuttling may serve as a novel type of regulation for calpain 2-mediated nuclear proteolysis.
Collapse
Affiliation(s)
- Shang-Min Chou
- Department and Graduate Institute of Microbiology, College of Medicine, Taipei, Taiwan
| | - Ting-Hsiang Huang
- Department and Graduate Institute of Microbiology, College of Medicine, Taipei, Taiwan
| | - Hsiang-Chin Chen
- Department and Graduate Institute of Microbiology, College of Medicine, Taipei, Taiwan
| | - Tsai-Kun Li
- Department and Graduate Institute of Microbiology, College of Medicine, Taipei, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
617
|
Laulier C, Cheng A, Stark JM. The relative efficiency of homology-directed repair has distinct effects on proper anaphase chromosome separation. Nucleic Acids Res 2011; 39:5935-44. [PMID: 21459848 PMCID: PMC3152340 DOI: 10.1093/nar/gkr187] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 03/15/2011] [Accepted: 03/16/2011] [Indexed: 01/17/2023] Open
Abstract
Homology-directed repair (HDR) is essential to limit mutagenesis, chromosomal instability (CIN) and tumorigenesis. We have characterized the consequences of HDR deficiency on anaphase, using markers for incomplete chromosome separation: DAPI-bridges and Ultra-fine bridges (UFBs). We show that multiple HDR factors (Rad51, Brca2 and Brca1) are critical for complete chromosome separation during anaphase, while another chromosome break repair pathway, non-homologous end joining, does not affect chromosome segregation. We then examined the consequences of mild versus severe HDR disruption, using two different dominant-negative alleles of the strand exchange factor, Rad51. We show that mild HDR disruption is viable, but causes incomplete chromosome separation, as detected by DAPI-bridges and UFBs, while severe HDR disruption additionally results in multipolar anaphases and loss of clonogenic survival. We suggest that mild HDR disruption favors the proliferation of cells that are prone to CIN due to defective chromosome separation during anaphase, whereas, severe HDR deficiency leads to multipolar divisions that are prohibitive for cell proliferation.
Collapse
Affiliation(s)
| | | | - Jeremy M. Stark
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Rd., Duarte, CA 91010, USA
| |
Collapse
|
618
|
Choudhuri R, Degraff W, Gamson J, Mitchell JB, Cook JA. Guggulsterone-mediated enhancement of radiosensitivity in human tumor cell lines. Front Oncol 2011; 1:19. [PMID: 22649756 PMCID: PMC3355920 DOI: 10.3389/fonc.2011.00019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 07/05/2011] [Indexed: 01/03/2023] Open
Abstract
Purpose: To observe the effect of guggulsterone (GS) on the radiation response in human cancer cell lines. Materials and methods: The radiation response of cancer cells treated with GS was observed by cell survival studies, cell growth assay, NF-κB activity assay, western blotting of some key growth promoting receptors, the DNA repair protein γH2AX, and flow cytometry for DNA analyses. Results: GS inhibited radiation induced NF-κB activation and enhanced radiosensitivity in the pancreatic cell line, PC-Sw. It reduced both cell cycle movement and cell growth. GS reduced ERα protein in MCF7 cells and IGF1-Rβ protein in colon cancer cells and pancreatic cancer cells and inhibited DNA double strand break (DSB) repair following radiation. Conclusion: GS induced radiation sensitization may be due to several different mechanisms including the inhibition of NF-κB activation and reductions in IGF1-Rβ. In addition, GS induced γH2AX formation, primarily in the S-phase, indicates that DNA DSB's in the S-phase may be another reason for GS induced radiosensitivity. ERα down-regulation in response to GS suggests that it can be of potential use in the treatment of estrogen positive tumors that are resistant to tamoxifen.
Collapse
Affiliation(s)
- Rajani Choudhuri
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
619
|
Topoisomerase II binds nucleosome-free DNA and acts redundantly with topoisomerase I to enhance recruitment of RNA Pol II in budding yeast. Proc Natl Acad Sci U S A 2011; 108:12693-8. [PMID: 21771901 DOI: 10.1073/pnas.1106834108] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
DNA topoisomerases are believed to promote transcription by removing excessive DNA supercoils produced during elongation. However, it is unclear how topoisomerases in eukaryotes are recruited and function in the transcription pathway in the context of nucleosomes. To address this problem we present high-resolution genome-wide maps of one of the major eukaryotic topoisomerases, Topoisomerase II (Top2) and nucleosomes in the budding yeast, Saccharomyces cerevisiae. Our data indicate that at promoters Top2 binds primarily to DNA that is nucleosome-free. However, although nucleosome loss enables Top2 occupancy, the opposite is not the case and the loss of Top2 has little effect on nucleosome density. We also find that Top2 is involved in transcription. Not only is Top2 enriched at highly transcribed genes, but Top2 is required redundantly with Top1 for optimal recruitment of RNA polymerase II at their promoters. These findings and the examination of candidate-activated genes suggest that nucleosome loss induced by nucleosome remodeling factors during gene activation enables Top2 binding, which in turn acts redundantly with Top1 to enhance recruitment of RNA polymerase II.
Collapse
|
620
|
Timsit Y. Local sensing of global DNA topology: from crossover geometry to type II topoisomerase processivity. Nucleic Acids Res 2011; 39:8665-76. [PMID: 21764774 PMCID: PMC3203592 DOI: 10.1093/nar/gkr556] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Type II topoisomerases are ubiquitous enzymes that control the topology and higher order structures of DNA. Type IIA enzymes have the remarkable property to sense locally the global DNA topology. Although many theoretical models have been proposed, the molecular mechanism of chiral discrimination is still unclear. While experimental studies have established that topoisomerases IIA discriminate topology on the basis of crossover geometry, a recent single-molecule experiment has shown that the enzyme has a different processivity on supercoiled DNA of opposite sign. Understanding how cross-over geometry influences enzyme processivity is, therefore, the key to elucidate the mechanism of chiral discrimination. Analysing this question from the DNA side reveals first, that the different stability of chiral DNA cross-overs provides a way to locally sense the global DNA topology. Second, it shows that these enzymes have evolved to recognize the G- and T-segments stably assembled into a right-handed cross-over. Third, it demonstrates how binding right-handed cross-overs across their large angle imposes a different topological link between the topoIIA rings and the plectonemes of opposite sign thus directly affecting the enzyme freedom of motion and processivity. In bridging geometry and kinetic data, this study brings a simple solution for type IIA topoisomerase chiral discrimination.
Collapse
Affiliation(s)
- Youri Timsit
- Information Génomique et Structurale, CNRS - UPR2589, Institut de Microbiologie de la Méditerranée, Aix-Marseille University, Parc Scientifique de Luminy, Marseille, France
| |
Collapse
|
621
|
Baviskar AT, Madaan C, Preet R, Mohapatra P, Jain V, Agarwal A, Guchhait SK, Kundu CN, Banerjee UC, Bharatam PV. N-fused imidazoles as novel anticancer agents that inhibit catalytic activity of topoisomerase IIα and induce apoptosis in G1/S phase. J Med Chem 2011; 54:5013-30. [PMID: 21644529 DOI: 10.1021/jm200235u] [Citation(s) in RCA: 222] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
On the basis of structures of known topoisomerase II catalytic inhibitors and initial molecular docking studies, bicyclic N-fused aminoimidazoles were predicted as potential topoisomerase II inhibitors. They were synthesized by multicomponent reactions and evaluated against human topoisomerase IIα (hTopoIIα) in decatenation, relaxation, cleavage complex, and DNA intercalation in vitro assays. Among 31 compounds of eight different bicyclic scaffolds, it was found that imidazopyridine, imidazopyrazole, and imidazopyrazine with suitable substituents exhibited potent inhibition of catalytic activity of hTopoIIα while not showing DNA intercalation. Molecular docking studies and molecular dynamics (MD) simulation analysis, ATPase-kinetics and ATP-dependent plasmid relaxation assay revealed the catalytic mode of inhibition of the title compounds plausibly by blocking the ATP-binding site. N-Fused aminoimidazoles showed potent anticancer activities in kidney and breast cancer cell lines, low toxicity to normal cells, relatively higher potency compared to etoposide and 5-fluorouracil in kidney cancer cell lines, and potent inhibition in cell migration. These compounds were found to exert apoptotic effect in G1/S phase.
Collapse
Affiliation(s)
- Ashish T Baviskar
- National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS Nagar (Mohali), Punjab-160062, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
622
|
Yamamoto Y, Fujita M, Koma H, Yamamori M, Nakamura T, Okamura N, Yagami T. 15-Deoxy-Δ12,14-prostaglandin J2 enhanced the anti-tumor activity of camptothecin against renal cell carcinoma independently of topoisomerase-II and PPARγ pathways. Biochem Biophys Res Commun 2011; 410:563-7. [PMID: 21683069 DOI: 10.1016/j.bbrc.2011.06.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 06/02/2011] [Indexed: 10/18/2022]
Abstract
Renal cell carcinoma (RCC) is chemoresistant cancer. Although several clinical trials were conducted to explore effective medications, the chemoresistance of RCC has not yet been conquered. An endogenous ligand for peroxisome proliferator-activated receptor-γ (PPARγ), 15-deoxy-Δ(12,14)-prostaglandin J(2) (15d-PGJ(2)), induces apoptosis in RCC. Here, we examined synergistic effects of several carcinostatics on the anti-tumor activity of 15d-PGJ(2) in Caki-2 cell line by MTT assay. A topoisomerase-I inhibitor, camptothecin (CPT), exhibited synergistically toxicity with 15d-PGJ(2), but neither 5-fluorouracil nor cisplatin did. The combination of 15d-PGJ(2) and a topoisomerase-II inhibitor, doxorubicine, did not cause synergistic cell growth inhibition. The synergistic effect of topoisomerase-I and II inhibitors was not also detected. A PPARγ antagonist, GW9662, did not prevent Caki-2 from undergoing 15d-PGJ(2)-induced cytotoxicity. The treatment of CPT combined with 15d-PGJ(2) activated caspase-3 more than the separate treatment. These results suggest that 15d-PGJ(2) exhibited the anti-tumor activity synergistically with CPT independent of topoisomerase-II and PPARγ.
Collapse
Affiliation(s)
- Yasuhiro Yamamoto
- Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 2-1, Kami-ohno 7-Chome, Himeji, Hyogo 670-8524, Japan
| | | | | | | | | | | | | |
Collapse
|
623
|
Homologous recombination repair is essential for repair of vosaroxin-induced DNA double-strand breaks. Oncotarget 2011; 1:606-19. [PMID: 21317456 DOI: 10.18632/oncotarget.101106] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Vosaroxin (formerly voreloxin) is a first-in-class anticancer quinolone derivative that intercalates DNA and inhibits topoisomerase II, inducing site-selective double-strand breaks (DSB), G2 arrest and apoptosis. Objective responses and complete remissions were observed in phase 2 studies of vosaroxin in patients with solid and hematologic malignancies, and responses were seen in patients whose cancers were resistant to anthracyclines. The quinolone-based scaffold differentiates vosaroxin from the anthracyclines and anthracenediones, broadly used DNA intercalating topoisomerase II poisons. Here we report that vosaroxin induces a cell cycle specific pattern of DNA damage and repair that is distinct from the anthracycline, doxorubicin. Both drugs stall replication and preferentially induce DNA damage in replicating cells, with damage in G2 / M > S >> G1. However, detectable replication fork collapse, as evidenced by DNA fragmentation and long tract recombination during S phase, is induced only by doxorubicin. Furthermore, vosaroxin induces less overall DNA fragmentation. Homologous recombination repair (HRR) is critical for recovery from DNA damage induced by both agents, identifying the potential to clinically exploit synthetic lethality.
Collapse
|
624
|
Wu X, Yalowich JC, Hasinoff BB. Cadmium is a catalytic inhibitor of DNA topoisomerase II. J Inorg Biochem 2011; 105:833-8. [PMID: 21497582 PMCID: PMC3091975 DOI: 10.1016/j.jinorgbio.2011.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 02/16/2011] [Accepted: 02/17/2011] [Indexed: 12/11/2022]
Abstract
Cadmium (Cd(2+)) is a highly toxic and carcinogenic metal that is an environmental and occupational hazard. DNA topoisomerase II is an essential nuclear enzyme and its inhibition can result in the formation of genotoxic and recombinogenic DNA double strand breaks. In this study we showed that cadmium chloride strongly inhibited the DNA decatenation activity of human topoisomerase IIα in the low micromolar concentration range and that its inhibitory effects were reduced by glutathione. Because the activity of topoisomerase II is strongly inhibited by thiol-reactive compounds this result suggested that cadmium may be binding to critical topoisomerase II cysteine thiols. Cadmium, however, did not stabilize DNA-topoisomerase II covalent complexes, as measured by the lack of formation of DNA double strand breaks. Hence, it is not likely to be a topoisomerase II poison. Consistent with the idea that cadmium cytotoxicity may be modulated by glutathione levels, buthionine sulfoximine pretreatment to decrease glutathione levels resulted in a greatly increased cadmium-induced cytotoxicity in K562 cells. The results of this study suggest that cadmium may exert some of its cell growth inhibitory, and possibly its toxicity and carcinogenicity, by inhibiting topoisomerase IIα through reaction with critical cysteine thiols.
Collapse
Affiliation(s)
| | | | - Brian B. Hasinoff
- Corresponding author. Faculty of Pharmacy, Apotex Centre, University of Manitoba, 750 McDermot Ave., Winnipeg, Manitoba, R3E 0T5, Canada. Tel: 204-474-8325. Fax: 204-474-7617. (Brian Hasinoff)
| |
Collapse
|
625
|
Kawatani M, Takayama H, Muroi M, Kimura S, Maekawa T, Osada H. Identification of a Small-Molecule Inhibitor of DNA Topoisomerase II by Proteomic Profiling. ACTA ACUST UNITED AC 2011; 18:743-51. [DOI: 10.1016/j.chembiol.2011.03.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 03/29/2011] [Accepted: 03/30/2011] [Indexed: 01/03/2023]
|
626
|
FANCD2 but not FANCA promotes cellular resistance to type II topoisomerase poisons. Cancer Lett 2011; 305:86-93. [PMID: 21414716 DOI: 10.1016/j.canlet.2011.02.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 02/17/2011] [Accepted: 02/21/2011] [Indexed: 11/22/2022]
Abstract
Genetic or epigenetic inactivation of the pathway formed by the Fanconi Anemia (FA) and BRCA proteins occurs in several cancer types, including lung and breast cancer, rendering the affected tumors potentially hypersensitive to DNA crosslinking agents. However, the cytotoxicity of other commonly used cancer therapeutics in cells with FA/BRCA pathway defects remains to be defined. Building on earlier data that implicated BRCA1 and BRCA2 in the repair of DNA damage caused by the topoisomerase II poison etoposide, we studied the role of FANCD2 in mediating resistance to several topoisomerase II poisons. We establish that the loss of FANCD2 increases cell death in response to etoposide. FANCD2 promotes homologous recombination repair (HRR) and prevents DNA double-strand break formation and chromosomal aberrations in etoposide-treated cells. Strikingly, this function of FANCD2 is independent of FANCD2 foci formation and of FANCA, which is a member of the FA core complex upstream of FANCD2 mono-ubiquitination. Thus, FANCD2 appears to promote HRR in a mono-ubiquitination-independent manner in conjunction with BRCA1/2. These data add to an emerging body of evidence indicating that the FA pathway is not linear and that several protein subcomplexes with different functions exist. Our findings are potentially relevant for predicting the sensitivity of lung and breast cancers to etoposide and doxorubicin, respectively.
Collapse
|
627
|
Kennedy S, Dicesare JC, Sheaff RJ. Topoisomerase I inactivation by a novel thiol reactive naphthoquinone. Biochem Biophys Res Commun 2011; 410:152-8. [PMID: 21651895 DOI: 10.1016/j.bbrc.2011.05.131] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 05/21/2011] [Indexed: 01/17/2023]
Abstract
The naphthoquinone adduct 12,13-dihydro-N-methyl-6,11,13-trioxo-5H-benzo[4,5]cyclohepta[1,2-b]naphthalen-5,12-imine (hereafter called TU100) contains structural features of both the anthracycline and isoquinone chemotherapeutics. An initial characterization showed TU100 is cytotoxic to mammalian cells and can inhibit topoisomerase I and II. Analysis using topoisomerase I now reveals TU100 is a slow acting inhibitor targeting the enzyme in the absence of DNA. Diluting pre-incubated TU100 and topoisomerase I failed to alleviate inhibition, suggesting the enzyme is being covalently modified. Critical cysteine thiols were identified as the possible target based on the ability of reducing agents to reverse TU100 inhibition. Consistent with this idea, TU100 protected topoisomerase I from inactivation by the sulfhydryl modifying agent N-ethylmaleimide (NEM). Unlike agents nonspecifically reacting with thiols, however, TU100 is specific for topoisomerase because it failed to inhibit a cysteine dependent protease. These results indicate TU100 is a novel naphthoquinone that inactivates free topoisomerase I via alkylation of cysteine residues.
Collapse
Affiliation(s)
- Steven Kennedy
- Department of Chemistry and Biochemistry, The University of Tulsa, Tulsa, OK 74104, United States
| | | | | |
Collapse
|
628
|
Wiśniewski JR, Ostasiewicz P, Mann M. High recovery FASP applied to the proteomic analysis of microdissected formalin fixed paraffin embedded cancer tissues retrieves known colon cancer markers. J Proteome Res 2011; 10:3040-9. [PMID: 21526778 DOI: 10.1021/pr200019m] [Citation(s) in RCA: 239] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Proteomic analysis of samples isolated by laser capture microdissection from clinical specimens requires sample preparation and fractionation methods suitable for small amounts of protein. Here we describe a streamlined filter-aided sample preparation (FASP) workflow that allows efficient analysis of lysates from low numbers of cells. Addition of carrier substances such as polyethylene glycol or dextran to the processed samples improves the peptide yields in the low to submicrogram range. In a single LC-MS/MS run, analyses of 500, 1000, and 3000 cells allowed identification of 905, 1536, and 2055 proteins, respectively. Incorporation of an additional SAX fractionation step at somewhat higher amounts enabled the analysis of formalin fixed and paraffin embedded human tissues prepared by LCM to a depth of 3600-4400 proteins per single experiment. We applied this workflow to compare archival neoplastic and matched normal colonic mucosa cancer specimens for three patients. Label-free quantification of more than 6000 proteins verified this technology through the differential expression of 30 known colon cancer markers. These included Carcino-Embryonic Antigen (CEA), the most widely used colon cancer marker, complement decay accelerating factor (DAF, CD55) and Metastasis-associated in colon cancer protein 1 (MACC1). Concordant with literature knowledge, mucin 1 was overexpressed and mucin 2 underexpressed in all three patients. These results show that FASP is suitable for the low level analysis of microdissected tissue and that it has the potential for exploration of clinical samples for biomarker and drug target discovery.
Collapse
Affiliation(s)
- Jacek R Wiśniewski
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, D-82152 Martinsried, Germany.
| | | | | |
Collapse
|
629
|
Kennedy S, DiCesare JC, Sheaff RJ. Topoisomerase I/II inhibition by a novel naphthoquinone containing a modified anthracycline ring system. Biochem Biophys Res Commun 2011; 408:94-8. [DOI: 10.1016/j.bbrc.2011.03.126] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 03/26/2011] [Indexed: 10/18/2022]
|
630
|
Tabassum S, Khan RA, Arjmand F, Sen S, Kayal J, Juvekar AS, Zingde SM. Synthesis and characterization of glycoconjugate tin(IV) complexes: In vitro DNA binding studies, cytotoxicity, and cell death. J Organomet Chem 2011; 696:1600-1608. [DOI: 10.1016/j.jorganchem.2011.01.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
631
|
Chen CF, He X, Arslan AD, Mo YY, Reinhold WC, Pommier Y, Beck WT. Novel regulation of nuclear factor-YB by miR-485-3p affects the expression of DNA topoisomerase IIα and drug responsiveness. Mol Pharmacol 2011; 79:735-41. [PMID: 21252292 PMCID: PMC3063729 DOI: 10.1124/mol.110.069633] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 01/20/2011] [Indexed: 12/22/2022] Open
Abstract
Nuclear factor (NF)-YB, a subunit of the transcription factor nuclear factor Y (NF-Y) complex, binds and activates CCAAT-containing promoters. Our previous work suggested that NF-YB may be a mediator of topoisomerase IIα (Top2α), working through the Top2α promoter. DNA topoisomerase II (Top2) is an essential nuclear enzyme and the primary target for several clinically important anticancer drugs. Our teniposide-resistant human lymphoblastic leukemia CEM cells (CEM/VM-1-5) express reduced Top2α protein compared with parental CEM cells. To study the regulation of Top2α during the development of drug resistance, we found that NF-YB protein expression is increased in CEM/VM-1-5 cells compared with parental CEM cells. This further suggests that increased NF-YB may be a negative regulator of Top2α in CEM/VM-1-5 cells. We asked what causes the up-regulation of NF-YB in CEM/VM-1-5 cells. We found by microRNA profiling that hsa-miR-485-3p is lower in CEM/VM-1-5 cells compared with CEM cells. MicroRNA target prediction programs revealed that the 3'-untranslated region (3'-UTR) of NF-YB harbors a putative hsa-miR-485-3p binding site. We thus hypothesized that hsa-miR-485-3p mediates drug responsiveness by decreasing NF-YB expression, which in turn negatively regulates Top2α expression. To test this, we overexpressed miR-485-3p in CEM/VM-1-5 cells and found that this led to reduced expression of NF-YB, a corresponding up-regulation of Top2α, and increased sensitivity to the Top2 inhibitors. Results in CEM cells were replicated in drug-sensitive and -resistant human rhabdomyosarcoma Rh30 cells, suggesting that our findings represent a general phenomenon. Ours is the first study to show that miR-485-3p mediates Top2α down-regulation in part by altered regulation of NF-YB.
Collapse
Affiliation(s)
- Cheng-Fen Chen
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
632
|
Bug M, Dobbelstein M. Anthracyclines induce the accumulation of mutant p53 through E2F1-dependent and -independent mechanisms. Oncogene 2011; 30:3612-24. [DOI: 10.1038/onc.2011.72] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
633
|
Dobrovolskaia IV, Kenward M, Arya G. Twist propagation in dinucleosome arrays. Biophys J 2011; 99:3355-64. [PMID: 21081084 DOI: 10.1016/j.bpj.2010.09.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 09/13/2010] [Accepted: 09/28/2010] [Indexed: 12/01/2022] Open
Abstract
We present a Monte Carlo simulation study of the distribution and propagation of twist from one DNA linker to another for a two-nucleosome array subjected to externally applied twist. A mesoscopic model of the array that incorporates nucleosome geometry along with the bending and twisting mechanics of the linkers is employed and external twist is applied in stepwise increments to mimic quasistatic twisting of chromatin fibers. Simulation results reveal that the magnitude and sign of the imposed and induced twist on contiguous linkers depend strongly on their relative orientation. Remarkably, the relative direction of the induced and applied twist can become inverted for a subset of linker orientations-a phenomenon we refer to as "twist inversion". We characterize the twist inversion, as a function of relative linker orientation, in a phase diagram and explain its key features using a simple model based on the geometry of the nucleosome/linker complex. In addition to twist inversion, our simulations reveal "nucleosome flipping", whereby nucleosomes may undergo sudden flipping in response to applied twist, causing a rapid bending of the linker and a significant change in the overall twist and writhe of the array. Our findings shed light on the underlying mechanisms by which torsional stresses impact chromatin organization.
Collapse
Affiliation(s)
- Irina V Dobrovolskaia
- Department of NanoEngineering, University of California at San Diego, La Jolla, CA, USA
| | | | | |
Collapse
|
634
|
Bau JT, Kurz EU. Sodium salicylate is a novel catalytic inhibitor of human DNA topoisomerase II alpha. Biochem Pharmacol 2011; 81:345-54. [DOI: 10.1016/j.bcp.2010.10.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 10/08/2010] [Accepted: 10/12/2010] [Indexed: 01/04/2023]
|
635
|
Wang P, Leung CH, Ma DL, Lu W, Che CM. Organoplatinum(II) complexes with nucleobase motifs as inhibitors of human topoisomerase II catalytic activity. Chem Asian J 2011; 5:2271-80. [PMID: 20730852 DOI: 10.1002/asia.201000451] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Platinum(II) complexes bearing acetylide ligands containing nucleobase motifs are prepared and their impact on human topoisomerase II (TopoII) is evaluated. Both platinum(II) complexes [Pt(II)(C^N^N)(C≡CCH₂R)] (1a-c) and [Pt(II)(tBu₃terpy)(C≡CCH₂R)](+) (2a-c) (C^N^N=6-phenyl-2,2'-bipyridyl, tBu₃terpy=4,4',4''-tri-tert-butyl-2,2':6',2''-terpyridyl, and R=(a) adenine, (b) thymine, and (c) 2-amino-6-chloropurine) are stable in aqueous solutions for 48 hours at room temperature. The binding constants (K) for the platinum(II) complexes towards calf thymus DNA are in the order of 10⁵ dm³ mol⁻¹ as estimated by using UV/Vis absorption spectroscopy. Of the complexes examined, only complexes 1a-c are found to behave as intercalators. Both complexes 1a-c and 2a-c inhibit TopoII-induced relaxation of supercoiled DNA, while 2c is the most potent TopoII inhibitors among the tested compounds. Inhibition of DNA relaxation is detected at nanomolar concentrations of 2c. All of the platinum(II) complexes are cytotoxic to human cancer cells with IC₅₀ values of 0.5-13.7 μM, while they are less toxic against normal cells CCD-19 Lu.
Collapse
Affiliation(s)
- Ping Wang
- Department of Chemistry and Open Laboratory of Chemical Biology of the Institute of Molecular Technology for Drug Discovery and Synthesis, The University of Hong Kong, Pokfulam Road, Hong Kong, P.R. China
| | | | | | | | | |
Collapse
|
636
|
Jun KY, Lee EY, Jung MJ, Lee OH, Lee ES, Park Choo HY, Na Y, Kwon Y. Synthesis, biological evaluation, and molecular docking study of 3-(3'-heteroatom substituted-2'-hydroxy-1'-propyloxy) xanthone analogues as novel topoisomerase IIα catalytic inhibitor. Eur J Med Chem 2011; 46:1964-71. [PMID: 21419530 DOI: 10.1016/j.ejmech.2011.01.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 12/23/2010] [Accepted: 01/10/2011] [Indexed: 11/19/2022]
Abstract
Epoxide ring-opened xanthone derivatives were synthesized and tested for their topoisomerase inhibitory activity and cytotoxicity. Most of the compounds showed topo IIα specific inhibitory activity. To clarify the mechanism of action of these compounds, the most potent compound (compound 14) of the synthesized analogues was further studied by testing its ATPase inhibitory activity and through molecular docking experiments. The results showed that the topo IIα inhibitory activity of compound 14 was inversely proportional to ATP concentration. In the ATPase inhibitory test, ATP hydrolysis was reduced less efficiently by compound 14 (28.5±4.6%) than novobiocin (60.4±8.1%). Molecular docking study revealed compound 14 to have a stable binding pattern to the ATP-binding domain of human topo II.
Collapse
Affiliation(s)
- Kyu-Yeon Jun
- College of Pharmacy & Division of Life & Pharmaceutical Sciences, Ewha Womans University, and Severance Hospital Integrative Research Institute for Cerebral & Cardiovascular Disease, Seoul 120-750, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
637
|
Kundukad B, van der Maarel JRC. Control of the flow properties of DNA by topoisomerase II and its targeting inhibitor. Biophys J 2011; 99:1906-15. [PMID: 20858436 DOI: 10.1016/j.bpj.2010.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 07/04/2010] [Accepted: 07/09/2010] [Indexed: 11/27/2022] Open
Abstract
The flow properties of DNA are important for understanding cell division and, indirectly, cancer therapy. DNA topology controlling enzymes such as topoisomerase II are thought to play an essential role. We report experiments showing how double-strand passage facilitated by topoisomerase II controls DNA rheology. For this purpose, we have measured the elastic storage and viscous loss moduli of a model system comprising bacteriophage λ-DNA and human topoisomerase IIα using video tracking of the Brownian motion of colloidal probe particles. We found that the rheology is critically dependent on the formation of temporal entanglements among the DNA molecules with a relaxation time of ∼1 s. We observed that topoisomerase II effectively removes these entanglements and transforms the solution from an elastic physical gel to a viscous fluid depending on the consumption of ATP. A second aspect of this study is the effect of the generic topoisomerase II inhibitor adenylyl-imidodiphosphate (AMP-PNP). In mixtures of AMP-PNP and ATP, the double-strand passage reaction gets blocked and progressively fewer entanglements are relaxed. A total replacement of ATP by AMP-PNP results in a temporal increase in elasticity at higher frequencies, but no transition to an elastic gel with fixed cross-links.
Collapse
Affiliation(s)
- Binu Kundukad
- Biophysics and Complex Fluids Group, Department of Physics, National University of Singapore, Singapore
| | | |
Collapse
|
638
|
Nakamura H, Shoji A, Takeuchi A, Ban HS, Lee JD, Yamori T, Kang SO. Discovery of ortho-Carborane-Conjugated Triazines as Selective Topoisomerase I/II Inhibitors. Aust J Chem 2011. [DOI: 10.1071/ch11295] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The cell growth inhibition profile of 2,4-(2-methyl-ortho-carboranyl)-4-(dimethylamino)-1,3,5-triazine (TAZ-6) was found to be similar to that of ICRF-193, a topoisomerase II inhibitor, as revealed by COMPARE analysis (correlation coefficient (r) = 0.724). Various mono- and di-ortho-carborane-substituted 1,3,5-triazines were synthesized based on the structure of TAZ-6 and tested for their ability to inhibit cell growth and the activities of topoisomerases I and II. Among the compounds synthesized, 3c, 4c, and 4f completely inhibited topoisomerase I activity without affecting topoisomerase II activity, whereas 3a and 3d completely inhibited topoisomerase II activity without affecting topoisomerase I activity, at 100 μM.
Collapse
|
639
|
Arita Y, Nishimura S, Matsuyama A, Yashiroda Y, Usui T, Boone C, Yoshida M. Microarray-based target identification using drug hypersensitive fission yeast expressing ORFeome. MOLECULAR BIOSYSTEMS 2011; 7:1463-72. [DOI: 10.1039/c0mb00326c] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
640
|
Haggerty TJ, Dunn IS, Rose LB, Newton EE, Martin S, Riley JL, Kurnick JT. Topoisomerase inhibitors modulate expression of melanocytic antigens and enhance T cell recognition of tumor cells. Cancer Immunol Immunother 2011; 60:133-44. [PMID: 21052994 PMCID: PMC3108190 DOI: 10.1007/s00262-010-0926-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 10/05/2010] [Indexed: 12/14/2022]
Abstract
While there are many obstacles to immune destruction of autologous tumors, there is mounting evidence that tumor antigen recognition does occur. Unfortunately, immune recognition rarely controls clinically significant tumors. Even the most effective immune response will fail if tumors fail to express target antigens. Importantly, reduced tumor antigen expression often results from changes in gene regulation rather than irrevocable loss of genetic information. Such perturbations are often reversible by specific compounds or biological mediators, prompting a search for agents with improved antigen-enhancing properties. Some recent findings have suggested that certain conventional chemotherapeutic agents may have beneficial properties for cancer treatment beyond their direct cytotoxicities against tumor cells. Accordingly, we screened an important subset of these agents, topoisomerase inhibitors, for their effects on antigen levels in tumor cells. Our analyses demonstrate upregulation of antigen expression in a variety of melanoma cell lines and gliomas in response to nanomolar levels of certain specific topoisomerase inhibitors. To demonstrate the ability of CD8+ T cells to recognize tumors, we assayed cytokine secretion in T cells transfected with T cell receptors directed against Melan-A/MART-1 antigen. Three days of daunorubicin treatment resulted in enhanced antigen expression by tumor cells, in turn inducing co-cultured antigen-specific T cells to secrete Interleukin-2 and Interferon-γ. These results demonstrate that specific topoisomerase inhibitors can augment melanoma antigen production, suggesting that a combination of chemotherapy and immunotherapy may be of potential value in the treatment of otherwise insensitive cancers.
Collapse
|
641
|
Abstract
Autoimmune disease affects a significant proportion of the population. The etiology of most autoimmune diseases is largely unknown, but it is thought to be multifactorial with both environmental and genetic influences. Rare monogenic autoimmune diseases, however, offer an invaluable window into potential disease mechanisms. In this review, we will discuss the autoimmune polyglandular syndrome (APS1), the immunedysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX), and autoimmune lymphoproliferative syndrome (ALPS). Significantly, the information gained from the study of these diseases has provided new insights into more common autoimmune disease and have yielded new diagnostics and therapeutic opportunities.
Collapse
Affiliation(s)
- Michael Waterfield
- Diabetes Center – University of California San Francisco
- Department of Pediatrics- University of California San Francisco
| | | |
Collapse
|
642
|
Williamson LM, Lees-Miller SP. Estrogen receptor α-mediated transcription induces cell cycle-dependent DNA double-strand breaks. Carcinogenesis 2010; 32:279-85. [PMID: 21112959 DOI: 10.1093/carcin/bgq255] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Prolonged exposure to estrogen increases breast cancer risk. Estrogen is known to induce chromosomal aberrations, yet the mechanisms by which estrogen promotes genomic instability are not fully understood. Here, we show that exposure of MCF-7 cells to 17β-estradiol (E2) induces DNA double-strand breaks (DSBs), as determined by the formation of γH2AX foci. Foci formation was dependent upon estrogen receptor-α (ERα) and the catalytic activity of the type II topoisomerase, topoisomerase IIβ (topoIIβ). Moreover, we show by chromatin immunoprecipitation that topoIIβ-dependent E2-induced γH2AX localizes to the promoter of the estrogen-inducible gene, trefoil factor 1. E2-induced foci were associated with cyclin A expression and inhibited by pre-incubation with the DNA polymerase inhibitor aphidicolin suggesting that E2-induced DSBs are mediated by progression through S phase. Furthermore, E2-induced γH2AX foci colocalized with Rad51, suggesting that E2-induced DSBs are repaired by homologous recombination. We propose that DNA DSBs formed by the strand-cleaving activity of the topoIIβ-DNA cleavage complex at estrogen-inducible genes can present a barrier to DNA replication, leading to persistent DNA DSBs in ERα-positive breast cancer cells.
Collapse
Affiliation(s)
- Laura M Williamson
- Department of Biochemistry and Molecular Biology, Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
643
|
Liu J, Leung CH, Chow ALF, Sun RWY, Yan SC, Che CM. Cyclometalated platinum(II) complexes as topoisomerase IIα poisons. Chem Commun (Camb) 2010; 47:719-21. [PMID: 21072335 DOI: 10.1039/c0cc03641b] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A platinum(II)-based major groove binder [Pt(II)(C^N)(C≡NR)(2)](+) (HC^N = 2-phenylpyridine (phpy), R = 2-naphthyl) was identified as a potent human topoisomerase IIα poison. It stabilizes the covalent TopoIIα-DNA cleavage complex and induces cancer cell death with potency significantly higher than the widely clinically used TopoIIα poison Vp-16.
Collapse
Affiliation(s)
- Jia Liu
- Department of Chemistry and Open Laboratory of Chemical Biology of the Institute of Molecular Technology for Drug Discovery and Synthesis, The University of Hong Kong, Pokfulam Road, Hong Kong
| | | | | | | | | | | |
Collapse
|
644
|
Jewell R, Conway C, Mitra A, Randerson-Moor J, Lobo S, Nsengimana J, Harland M, Marples M, Edward S, Cook M, Powell B, Boon A, de Kort F, Parker KA, Cree IA, Barrett JH, Knowles MA, Bishop DT, Newton-Bishop J. Patterns of expression of DNA repair genes and relapse from melanoma. Clin Cancer Res 2010; 16:5211-21. [PMID: 20705614 PMCID: PMC2972254 DOI: 10.1158/1078-0432.ccr-10-1521] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE To use gene expression profiling of formalin-fixed primary melanoma samples to detect expression patterns that are predictive of relapse and response to chemotherapy. EXPERIMENTAL DESIGN Gene expression profiles were identified in samples from two studies (472 tumors). Gene expression data for 502 cancer-related genes from these studies were combined for analysis. RESULTS Increased expression of DNA repair genes most strongly predicted relapse and was associated with thicker tumors. Increased expression of RAD51 was the most predictive of relapse-free survival in unadjusted analysis (hazard ratio, 2.98; P = 8.80 × 10(-6)). RAD52 (hazard ratio, 4.73; P = 0.0004) and TOP2A (hazard ratio, 3.06; P = 0.009) were independent predictors of relapse-free survival in multivariable analysis. These associations persisted when the analysis was further adjusted for demographic and histologic features of prognostic importance (RAD52 P = 0.01; TOP2A P = 0.02). Using principal component analysis, expression of DNA repair genes was summarized into one variable. Genes whose expression correlated with this variable were predominantly associated with the cell cycle and DNA repair. In 42 patients treated with chemotherapy, DNA repair gene expression was greater in tumors from patients who progressed on treatment. Further data supportive of a role for increased expression of DNA repair genes as predictive biomarkers are reported, which were generated using multiplex PCR. CONCLUSIONS Overexpression of DNA repair genes (predominantly those involved in double-strand break repair) was associated with relapse. These data support the hypothesis that melanoma progression requires maintenance of genetic stability and give insight into mechanisms of melanoma drug resistance and potential therapies.
Collapse
Affiliation(s)
- Rosalyn Jewell
- Section of Epidemiology and Biostatistics, Leeds Institute of Molecular Medicine, St James's University Hospital, Leeds, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
645
|
Hawtin RE, Stockett DE, Wong OK, Lundin C, Helleday T, Fox JA. Homologous recombination repair is essential for repair of vosaroxin-induced DNA double-strand breaks. Oncotarget 2010; 1:606-619. [PMID: 21317456 PMCID: PMC3248135 DOI: 10.18632/oncotarget.195] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 11/22/2010] [Indexed: 11/25/2022] Open
Abstract
Vosaroxin (formerly voreloxin) is a first-in-class anticancer quinolone derivative that intercalates DNA and inhibits topoisomerase II, inducing site-selective double-strand breaks (DSB), G2 arrest and apoptosis. Objective responses and complete remissions were observed in phase 2 studies of vosaroxin in patients with solid and hematologic malignancies, and responses were seen in patients whose cancers were resistant to anthracyclines. The quinolone-based scaffold differentiates vosaroxin from the anthracyclines and anthracenediones, broadly used DNA intercalating topoisomerase II poisons. Here we report that vosaroxin induces a cell cycle specific pattern of DNA damage and repair that is distinct from the anthracycline, doxorubicin. Both drugs stall replication and preferentially induce DNA damage in replicating cells, with damage in G2 / M > S >> G1. However, detectable replication fork collapse, as evidenced by DNA fragmentation and long tract recombination during S phase, is induced only by doxorubicin. Furthermore, vosaroxin induces less overall DNA fragmentation. Homologous recombination repair (HRR) is critical for recovery from DNA damage induced by both agents, identifying the potential to clinically exploit synthetic lethality.
Collapse
Affiliation(s)
| | - David Elliot Stockett
- Sunesis Pharmaceuticals, Inc. 395 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| | - Oi Kwan Wong
- Sunesis Pharmaceuticals, Inc. 395 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| | - Cecilia Lundin
- Gray Institute for Radiation Oncology & Biology, University of Oxford. Old Road Campus Research Building, Roosevelt Drive. Oxford, OX3 7DQ, UK
| | - Thomas Helleday
- Gray Institute for Radiation Oncology & Biology, University of Oxford. Old Road Campus Research Building, Roosevelt Drive. Oxford, OX3 7DQ, UK
- Dept. of Genetics Microbiology and Toxicology, Stockholm University. Arrhenius Laboratory, Svante Arrhenius väg 16 E4. S-106 91 Stockholm, Sweden
| | - Judith Ann Fox
- Sunesis Pharmaceuticals, Inc. 395 Oyster Point Boulevard, South San Francisco, CA 94080, USA
| |
Collapse
|
646
|
Jäämaa S, Af Hällström TM, Sankila A, Rantanen V, Koistinen H, Stenman UH, Zhang Z, Yang Z, De Marzo AM, Taari K, Ruutu M, Andersson LC, Laiho M. DNA damage recognition via activated ATM and p53 pathway in nonproliferating human prostate tissue. Cancer Res 2010; 70:8630-41. [PMID: 20978201 DOI: 10.1158/0008-5472.can-10-0937] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
DNA damage response (DDR) pathways have been extensively studied in cancer cell lines and mouse models, but little is known about how DNA damage is recognized by different cell types in nonmalignant, slowly replicating human tissues. Here, we assess, using ex vivo cultures of human prostate tissue, DDR caused by cytotoxic drugs (camptothecin, doxorubicin, etoposide, and cisplatin) and ionizing radiation (IR) in the context of normal tissue architecture. Using specific markers for basal and luminal epithelial cells, we determine and quantify cell compartment-specific damage recognition. IR, doxorubicin, and etoposide induced the phosphorylation of H2A.X on Ser(139) (γH2AX) and DNA damage foci formation. Surprisingly, luminal epithelial cells lack the prominent γH2AX response after IR when compared with basal cells, although ATM phosphorylation on Ser(1981) and 53BP1 foci were clearly detectable in both cell types. The attenuated γH2AX response seems to result from low levels of total H2A.X in the luminal cells. Marked increase in p53, a downstream target of the activated ATM pathway, was detected only in response to camptothecin and doxorubicin. These findings emphasize the diversity of pathways activated by DNA damage in slowly replicating tissues and reveal an unexpected deviation in the prostate luminal compartment that may be relevant in prostate tumorigenesis. Detailed mapping of tissue and cell type differences in DDR will provide an outlook of relevant responses to therapeutic strategies.
Collapse
Affiliation(s)
- Sari Jäämaa
- Biomedicum Helsinki and Department of Virology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
647
|
Ye J, Lenain C, Bauwens S, Rizzo A, Saint-Léger A, Poulet A, Benarroch D, Magdinier F, Morere J, Amiard S, Verhoeyen E, Britton S, Calsou P, Salles B, Bizard A, Nadal M, Salvati E, Sabatier L, Wu Y, Biroccio A, Londoño-Vallejo A, Giraud-Panis MJ, Gilson E. TRF2 and apollo cooperate with topoisomerase 2alpha to protect human telomeres from replicative damage. Cell 2010; 142:230-42. [PMID: 20655466 DOI: 10.1016/j.cell.2010.05.032] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 12/21/2009] [Accepted: 05/17/2010] [Indexed: 10/19/2022]
Abstract
Human telomeres are protected from DNA damage by a nucleoprotein complex that includes the repeat-binding factor TRF2. Here, we report that TRF2 regulates the 5' exonuclease activity of its binding partner, Apollo, a member of the metallo-beta-lactamase family that is required for telomere integrity during S phase. TRF2 and Apollo also suppress damage to engineered interstitial telomere repeat tracts that were inserted far away from chromosome ends. Genetic data indicate that DNA topoisomerase 2alpha acts in the same pathway of telomere protection as TRF2 and Apollo. Moreover, TRF2, which binds preferentially to positively supercoiled DNA substrates, together with Apollo, negatively regulates the amount of TOP1, TOP2alpha, and TOP2beta at telomeres. Our data are consistent with a model in which TRF2 and Apollo relieve topological stress during telomere replication. Our work also suggests that cellular senescence may be caused by topological problems that occur during the replication of the inner portion of telomeres.
Collapse
Affiliation(s)
- Jing Ye
- Shanghai Ruijin Hospital, School of Medicine of Shanghai Jiaotong University, 200025 Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
648
|
Cultured cerebellar granule neurons as an in vitro aging model: topoisomerase IIβ as an additional biomarker in DNA repair and aging. Toxicol In Vitro 2010; 24:1935-45. [PMID: 20708677 DOI: 10.1016/j.tiv.2010.08.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 07/27/2010] [Accepted: 08/09/2010] [Indexed: 01/11/2023]
Abstract
Aging in the brain is a multicellular process manifesting as neurodegeneration and associated functional impairment. In the present study, we report that cerebellar granule neurons (CGNs) in culture show senescence-mediated molecular changes indicating establishment of aging processes in vitro. CGNs were viable for 5 weeks followed by cellular degeneration. Molecular changes correlated with cellular senescence and aging include the elevation of senescence-mediated beta galactosidase (SA-β-gal) activity and intracellular Ca(2+) levels. Decreased base excision repair (BER) as well as non-homologous end joining (NHEJ) activities in CGNs were also observed upon aging in vitro. The decrease in NHEJ activity was shown correlated with corresponding decrease in the levels of topoisomerase IIβ (topo IIβ), Ku 70 and Ku 80 suggesting a crucial role for topo IIβ in repair capacity of CGNs. These studies, besides establishing that CGNs would serve as a good in vitro model for analysis of aging phenomena, also brought out that topo IIβ, by virtue of its significant role in controlling NHEJ activity, would serve as an additional biomarker for studying aging process.
Collapse
|
649
|
Giraud-Panis MJ, Pisano S, Poulet A, Le Du MH, Gilson E. Structural identity of telomeric complexes. FEBS Lett 2010; 584:3785-99. [PMID: 20696167 DOI: 10.1016/j.febslet.2010.08.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 08/02/2010] [Accepted: 08/02/2010] [Indexed: 02/01/2023]
Abstract
A major issue in telomere research is to understand how the integrity of chromosome ends is controlled. Although several nucleoprotein complexes have been described at the telomeres of different organisms, it is still unclear how they confer a structural identity to chromosome ends in order to mask them from DNA repair and to ensure their proper replication. In this review, we describe how telomeric nucleoprotein complexes are structured, comparing different organisms and trying to link these structures to telomere biology. It emerges that telomeres are formed by a complex and specific network of interactions between DNA, RNA and proteins. The fact that these interactions and associated activities are reinforcing each other might help to guaranty the robustness of telomeric functions across the cell cycle and in the event of cellular perturbations. We propose that telomeric nucleoprotein complexes orient cell fate through dynamic transitions in their structures and their organization.
Collapse
Affiliation(s)
- Marie-Josèphe Giraud-Panis
- University de Nice, Laboratory of Biology and Pathology of Genomes, UMR 6267 CNRS U998 INSERM, Faculté de Médecine, Nice, France
| | | | | | | | | |
Collapse
|
650
|
Podhorecka M, Skladanowski A, Bozko P. H2AX Phosphorylation: Its Role in DNA Damage Response and Cancer Therapy. J Nucleic Acids 2010; 2010. [PMID: 20811597 PMCID: PMC2929501 DOI: 10.4061/2010/920161] [Citation(s) in RCA: 396] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 06/28/2010] [Accepted: 07/05/2010] [Indexed: 11/20/2022] Open
Abstract
Double-strand breaks (DSBs) are the most deleterious DNA lesions, which, if left unrepaired, may have severe consequences for cell survival, as they lead to chromosome aberrations, genomic instability, or cell death. Various physical, chemical, and biological factors are involved in DSB induction. Cells respond to DNA damage by activating the so-called DNA damage response (DDR), a complex molecular mechanism developed to detect and repair DNA damage. The formation of DSBs triggers activation of many factors, including phosphorylation of the histone variant H2AX, producing gammaH2AX. Phosphorylation of H2AX plays a key role in DDR and is required for the assembly of DNA repair proteins at the sites containing damaged chromatin as well as for activation of checkpoints proteins which arrest the cell cycle progression. In general, analysis of gammaH2AX expression can be used to detect the genotoxic effect of different toxic substances. When applied to clinical samples from cancer patients, evaluation of gammaH2AX levels may allow not only to monitor the efficiency of anticancer treatment but also to predict of tumor cell sensitivity to DNA damaging anticancer agents and toxicity of anticancer treatment toward normal cells.
Collapse
Affiliation(s)
- Monika Podhorecka
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20081 Lublin, Poland
| | | | | |
Collapse
|