51
|
Arnesano F, Natile G. Mechanistic insight into the cellular uptake and processing of cisplatin 30 years after its approval by FDA. Coord Chem Rev 2009. [DOI: 10.1016/j.ccr.2009.01.028] [Citation(s) in RCA: 183] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
52
|
Reaction of human metallothionein-3 with cisplatin and transplatin. J Biol Inorg Chem 2009; 14:1129-38. [PMID: 19536566 DOI: 10.1007/s00775-009-0557-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 06/08/2009] [Indexed: 10/20/2022]
Abstract
Human metallothioneins, small cysteine- and metal-rich proteins, play an important role in the acquired resistance to platinum-based anticancer drugs. These proteins contain a M(II)4(CysS)11 cluster and a M(II)3(CysS)9 cluster localized in the alpha-domain and the beta-domain, respectively. The noninducible isoform metallothionein-3 (Zn7MT-3) is mainly expressed in the brain, but was found overexpressed in a number of cancer tissues. Since the structural properties of this isoform substantially differ from those of the ubiquitously occurring Zn7MT-1/Zn7MT-2 isoforms, the reactions of cis-diamminedichloridoplatinum(II) (cisplatin) and trans-diamminedichloridoplatinum(II) (transplatin) with human Zn7MT-3 were investigated and the products characterized. A comparison of the reaction kinetics revealed that transplatin reacts with cysteine ligands of Zn7MT-3 faster than cisplatin. In both binding processes, stoichiometric amounts of Zn(II) were released from the protein. Marked differences between the reaction rates of cisplatin and transplatin binding to Zn7MT-3 and the formation of the Pt-S bonds suggest that the binding of both Pt(II) compounds is a complex process, involving at least two subsequent binding steps. The electrospray ionization mass spectrometry characterization of the products showed that whereas all ligands in cisplatin were replaced by cysteine thiolates, transplatin retained its carrier ammine ligands. The 113Cd NMR studies of Pt1 113Cd6MT-3 revealed that cisplatin binds preferentially to the beta-domain of the protein. The rates of reaction of cisplatin and transplatin with Zn7MT-3 were much faster than those of cisplatin and transplatin with Zn7MT-2. The biological consequences of a substantially higher reactivity of cisplatin toward Zn7MT-3 than Zn7MT-2 in the acquired resistance to platinum-based drugs are discussed.
Collapse
|
53
|
Moreno-Gordaliza E, Cañas B, Palacios MA, Gómez-Gómez MM. Top-Down Mass Spectrometric Approach for the Full Characterization of Insulin−Cisplatin Adducts. Anal Chem 2009; 81:3507-16. [DOI: 10.1021/ac900046v] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Estefanía Moreno-Gordaliza
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, Avenida Complutense s/n, 28040 Madrid, Spain
| | - Benito Cañas
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, Avenida Complutense s/n, 28040 Madrid, Spain
| | - María A. Palacios
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, Avenida Complutense s/n, 28040 Madrid, Spain
| | - M. Milagros Gómez-Gómez
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, Avenida Complutense s/n, 28040 Madrid, Spain
| |
Collapse
|
54
|
The soluble metal-binding domain of the copper transporter ATP7B binds and detoxifies cisplatin. Biochem J 2009; 419:51-6, 3 p following 56. [PMID: 19173677 DOI: 10.1042/bj20081359] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Wilson disease ATPase (ATP7B) has been implicated in the resistance of cancer cells to cisplatin. Using a simple in vivo assay in bacterial culture, in the present study we demonstrate that ATP7B can confer resistance to cisplatin by sequestering the drug in its N-terminal metal-binding domain without active drug extrusion from the cell. Expression of a protein fragment containing four N-terminal MBRs (metal-binding repeats) of ATP7B (MBR1-4) protects cells from the toxic effects of cisplatin. One MBR1-4 molecule binds up to three cisplatin molecules at the copper-binding sites in the MBRs. The findings of the present study suggest that suppressing enzymatic activity of ATP7B may not be an effective way of combating cisplatin resistance. Rather, the efforts should be directed at preventing cisplatin binding to the protein.
Collapse
|
55
|
Petering DH, Krezoski S, Tabatabai NM. Metallothionein Toxicology: Metal Ion Trafficking and Cellular Protection. METALLOTHIONEINS AND RELATED CHELATORS 2009. [DOI: 10.1039/9781847559531-00353] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The literature is replete with reports about the involvement of metallothionein in host defense against injurious chemical, biological, and physical agents. Yet, metallothionein's functional roles are still being debated. This review addresses the issues that have left the physiological significance of metallothionein in doubt and moves on to assess the MT's importance in cell toxicology. It is evident that the protein is broadly involved in protecting cells from injury due to toxic metal ions, oxidants, and electrophiles. Attention is focused on MT's structural and chemical properties that confer this widespread role in cell protection. Particular emphasis is placed on the implications of finding that metal ion unsaturated metallothionein is commonly present in many cells and tissues and the question, how does selectivity of reaction with metallothionein take place in the cellular environment that includes large numbers of competing metal binding sites and high concentrations of protein and glutathione sulfhydryl groups?
Collapse
Affiliation(s)
- David H. Petering
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee Milwaukee WI 53201 USA
| | - Susan Krezoski
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee Milwaukee WI 53201 USA
| | - Niloofar M. Tabatabai
- Division of Endocrinology, Metabolism and Clinical Nutrition and Kidney Disease Center Medical College of Wisconsin Milwaukee WI 53226 USA
| |
Collapse
|
56
|
Pedersen MØ, Larsen A, Stoltenberg M, Penkowa M. The role of metallothionein in oncogenesis and cancer prognosis. ACTA ACUST UNITED AC 2008; 44:29-64. [PMID: 19348910 DOI: 10.1016/j.proghi.2008.10.001] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Accepted: 10/02/2008] [Indexed: 12/12/2022]
Abstract
The antiapoptotic, antioxidant, proliferative, and angiogenic effects of metallothionein (MT)-I+II has resulted in increased focus on their role in oncogenesis, tumor progression, therapy response, and patient prognosis. Studies have reported increased expression of MT-I+II mRNA and protein in various human cancers; such as breast, kidney, lung, nasopharynx, ovary, prostate, salivary gland, testes, urinary bladder, cervical, endometrial, skin carcinoma, melanoma, acute lymphoblastic leukemia (ALL), and pancreatic cancers, where MT-I+II expression is sometimes correlated to higher tumor grade/stage, chemotherapy/radiation resistance, and poor prognosis. However, MT-I+II are downregulated in other types of tumors (e.g. hepatocellular, gastric, colorectal, central nervous system (CNS), and thyroid cancers) where MT-I+II is either inversely correlated or unrelated to mortality. Large discrepancies exist between different tumor types, and no distinct and reliable association exists between MT-I+II expression in tumor tissues and prognosis and therapy resistance. Furthermore, a parallel has been drawn between MT-I+II expression as a potential marker for prognosis, and MT-I+II's role as oncogenic factors, without any direct evidence supporting such a parallel. This review aims at discussing the role of MT-I+II both as a prognostic marker for survival and therapy response, as well as for the hypothesized role of MT-I+II as causal oncogenes.
Collapse
Affiliation(s)
- Mie Ø Pedersen
- Section of Neuroprotection, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, The Panum Institute, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark.
| | | | | | | |
Collapse
|
57
|
Karotki AV, Vasák M. Interaction of metallothionein-2 with platinum-modified 5'-guanosine monophosphate and DNA. Biochemistry 2008; 47:10961-9. [PMID: 18803406 DOI: 10.1021/bi801253x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human metallothioneins (MTs), a family of cysteine- and metal-rich metalloproteins, play an important role in the acquired resistance to platinum drugs. MTs occur in the cytosol and the nucleus of the cells and sequester platinum drugs through interaction with their zinc-thiolate clusters. Herein, we investigate the ability of human Zn 7MT-2 to form DNA-Pt-MT cross-links using the cisplatin- and transplatin-modified plasmid DNA pSP73. Immunochemical analysis of MT-2 showed that the monofunctional platinum-DNA adducts formed DNA- cis/ trans-Pt-MT cross-links and that platinated MT-2 was released from the DNA- trans-Pt-MT cross-links with time. The DNA- cis/ trans-Pt-MT cross-links were also formed in the presence of 2 mM glutathione, a strong S-donor ligand. Independently, we used 5'-guanosine monophosphate (5'-GMP) platinated at the N7 position as a model of monofunctional platinum-DNA adducts. Comparison of reaction kinetics revealed that the formation of ternary complexes between Zn 7MT-2 and cis-Pt-GMP was faster than that of the trans isomer. The analysis of the reaction products with time showed that while the formation of ternary GMP- trans-Pt-MT complex(es) is accompanied by 5'-GMP release, a stable ternary GMP- cis-Pt-MT complex is formed. In the latter complex, a fast initial formation of two Pt-S bonds was followed by a slow formation of an additional Pt-S bond yielding an unusual Pt(II)S 3N coordination with N7-GMP as the only N-donor ligand. The ejection of negligible zinc from the zinc-thiolate clusters implies the initial formation of Zn-(mu-SCys)-Pt bridges involving the terminal thiolate ligands. The biological implications of these studies are discussed.
Collapse
Affiliation(s)
- Andrei V Karotki
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | | |
Collapse
|
58
|
Suchánková T, Vojtísková M, Reedijk J, Brabec V, Kaspárková J. DNA and glutathione interactions in cell-free media of asymmetric platinum(II) complexes cis- and trans-[PtCl2(isopropylamine)(1-methylimidazole)]: relations to their different antitumor effects. J Biol Inorg Chem 2008; 14:75-87. [PMID: 18777181 DOI: 10.1007/s00775-008-0425-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2008] [Accepted: 08/24/2008] [Indexed: 11/25/2022]
Abstract
The global modification of mammalian and plasmid DNAs by the novel platinum compounds cis-[PtCl(2)(isopropylamine)(1-methylimidazole)] and trans-[PtCl(2)(isopropylamine)(1-methylimidazole)] and the reactivity of these compounds with reduced glutathione (GSH) were investigated in cell-free media using various biochemical and biophysical methods. Earlier cytotoxicity studies had revealed that the replacement of the NH(3) groups in cisplatin by the azole and isopropylamine ligands lowers the activity of cisplatin in both sensitive and resistant cell lines. The results of the present work show that this replacement does not considerably affect the DNA modifications by this drug, recognition of these modifications by HMGB1 protein, their repair, and reactivity of the platinum complex with GSH. These results were interpreted to mean that the reduced activity of this analog of cisplatin in tumor cell lines is due to factors that do not operate at the level of the target DNA. In contrast, earlier studies had shown that the replacement of the NH(3) groups in the clinically ineffective trans isomer (transplatin) by the azole and isopropylamine ligands results in a radical enhancement of its activity in tumor cell lines. Importantly, this replacement also markedly alters the DNA binding mode of transplatin, which is distinctly different from that of cisplatin, but does not affect reactivity with GSH. Hence, the results of the present work are consistent with the view and support the hypothesis systematically tested by us and others that platinum drugs that bind to DNA in a fundamentally different manner from that of conventional cisplatin may have altered pharmacological properties.
Collapse
Affiliation(s)
- Tereza Suchánková
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 61265, Brno, Czech Republic
| | | | | | | | | |
Collapse
|
59
|
Fabrik I, Krizkova S, Huska D, Adam V, Hubalek J, Trnkova L, Eckschlager T, Kukacka J, Prusa R, Kizek R. Employment of Electrochemical Techniques for Metallothionein Determination in Tumor Cell Lines and Patients with a Tumor Disease. ELECTROANAL 2008. [DOI: 10.1002/elan.200704215] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
60
|
Investigation of interaction between human hemoglobin A0 and platinum anticancer drugs by capillary isoelectric focusing with whole column imaging detection. J Sep Sci 2008; 31:1803-9. [DOI: 10.1002/jssc.200700418] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
61
|
Halámiková A, Heringová P, Kašpárková J, Intini FP, Natile G, Nemirovski A, Gibson D, Brabec V. Cytotoxicity, mutagenicity, cellular uptake, DNA and glutathione interactions of lipophilic trans-platinum complexes tethered to 1-adamantylamine. J Inorg Biochem 2008; 102:1077-89. [DOI: 10.1016/j.jinorgbio.2007.12.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2007] [Revised: 12/07/2007] [Accepted: 12/14/2007] [Indexed: 11/26/2022]
|
62
|
Ragunathan N, Dairou J, Pluvinage B, Martins M, Petit E, Janel N, Dupret JM, Rodrigues-Lima F. Identification of the xenobiotic-metabolizing enzyme arylamine N-acetyltransferase 1 as a new target of cisplatin in breast cancer cells: molecular and cellular mechanisms of inhibition. Mol Pharmacol 2008; 73:1761-8. [PMID: 18310302 DOI: 10.1124/mol.108.045328] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Arylamine N-acetyltransferase 1 (NAT1) is a phase II xenobiotic-metabolizing enzyme that plays an important role in the biotransformation of aromatic drugs and carcinogens. NAT1 activity has long been associated with susceptibility to various cancers. Evidence for a role of NAT1 in malignant progression has also been obtained, particularly for breast and prostate cancer. Cisplatin is widely used in chemotherapy against human cancers, and it is thought to act principally by forming DNA adducts. However, recent studies have suggested that some of the pharmacological and/or toxicological effects of cisplatin may be due to the direct targeting and inhibition of certain cellular enzymes. We show here that the exposure of breast cancer cells, known to express functional NAT1 enzyme, to therapeutically relevant concentrations of cisplatin impairs the catalytic activity of endogenous NAT1. Endogenous NAT1 was also found to be inactivated, in vivo, in the tissues of mice treated with cisplatin. Mechanistic studies with purified human NAT1 indicated that this inhibition resulted from the irreversible formation of a cisplatin adduct with the active-site cysteine residue of the enzyme. Kinetic studies suggested that NAT1 interacts rapidly with cisplatin, with a second-order rate inhibition constant of 700 M(-1) min(-1). This rate constant is one the highest ever reported for the reaction of cisplatin with a biological macromolecule. Few enzymes have been clearly shown to be inactivated by cisplatin. We provide here molecular and cellular evidence suggesting that NAT1 is one of the targets of cisplatin in cells.
Collapse
Affiliation(s)
- Nilusha Ragunathan
- Laboratoire de Cytophysiologie et Toxicologie Cellulaire (EA 1553), Université Paris Diderot-Paris 7, 75005, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
63
|
Mellor HR, Callaghan R. Resistance to chemotherapy in cancer: a complex and integrated cellular response. Pharmacology 2008; 81:275-300. [PMID: 18259091 DOI: 10.1159/000115967] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Accepted: 09/10/2007] [Indexed: 12/30/2022]
Abstract
Inherent and acquired resistance pathways account for the high rate of failure in cancer chemotherapy. The mechanisms or pathways mediating resistance may be classified as pharmacokinetic (i.e. alter intratumour drug exposue) or pharmacodynamic (i.e. failure to elicit cytotoxicity). More often than not, the resistant phenotype is characterised by alterations in multiple pathways. Consequently, the pathways may act synergistically or generate a broad spectrum of resistance to anticancer drugs. There has been a great deal of systematic characterisation of drug resistance in vitro. However, translating this greater understanding into clinical efficacy has rarely been achieved. This review explores the phenomenon of drug resistance in cancer and highlights the gap between in vitro and in vivo observations. This gap presents a major obstacle in overcoming drug resistance and restoring sensitivity to chemotherapy.
Collapse
Affiliation(s)
- Howard R Mellor
- Growth Factor Group, Weatherall Institute of Molecular Medicine, Oxford, UK
| | | |
Collapse
|
64
|
de Mier-Vinué J, Gay M, Montaña ÁM, Sáez RI, Moreno V, Kasparkova J, Vrana O, Heringova P, Brabec V, Boccarelli A, Coluccia M, Natile G. Synthesis, Biophysical Studies, and Antiproliferative Activity of Platinum(II) Complexes Having 1,2-Bis(aminomethyl)carbobicyclic Ligands. J Med Chem 2008; 51:424-31. [DOI: 10.1021/jm070844u] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Jordi de Mier-Vinué
- Department of Organic Chemistry and Department of Inorganic Chemistry, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., CZ-61265 Brno, Czech Republic, and Dipartimento Farmaco Chimico and Dipartimento di Scienze Biomediche e Oncologia Umana, Universitá degli Studi di Bari, Bari, Italy
| | - Marina Gay
- Department of Organic Chemistry and Department of Inorganic Chemistry, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., CZ-61265 Brno, Czech Republic, and Dipartimento Farmaco Chimico and Dipartimento di Scienze Biomediche e Oncologia Umana, Universitá degli Studi di Bari, Bari, Italy
| | - Ángel M. Montaña
- Department of Organic Chemistry and Department of Inorganic Chemistry, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., CZ-61265 Brno, Czech Republic, and Dipartimento Farmaco Chimico and Dipartimento di Scienze Biomediche e Oncologia Umana, Universitá degli Studi di Bari, Bari, Italy
| | - Rosa-Isabel Sáez
- Department of Organic Chemistry and Department of Inorganic Chemistry, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., CZ-61265 Brno, Czech Republic, and Dipartimento Farmaco Chimico and Dipartimento di Scienze Biomediche e Oncologia Umana, Universitá degli Studi di Bari, Bari, Italy
| | - Virtudes Moreno
- Department of Organic Chemistry and Department of Inorganic Chemistry, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., CZ-61265 Brno, Czech Republic, and Dipartimento Farmaco Chimico and Dipartimento di Scienze Biomediche e Oncologia Umana, Universitá degli Studi di Bari, Bari, Italy
| | - Jana Kasparkova
- Department of Organic Chemistry and Department of Inorganic Chemistry, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., CZ-61265 Brno, Czech Republic, and Dipartimento Farmaco Chimico and Dipartimento di Scienze Biomediche e Oncologia Umana, Universitá degli Studi di Bari, Bari, Italy
| | - Oldrich Vrana
- Department of Organic Chemistry and Department of Inorganic Chemistry, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., CZ-61265 Brno, Czech Republic, and Dipartimento Farmaco Chimico and Dipartimento di Scienze Biomediche e Oncologia Umana, Universitá degli Studi di Bari, Bari, Italy
| | - Pavla Heringova
- Department of Organic Chemistry and Department of Inorganic Chemistry, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., CZ-61265 Brno, Czech Republic, and Dipartimento Farmaco Chimico and Dipartimento di Scienze Biomediche e Oncologia Umana, Universitá degli Studi di Bari, Bari, Italy
| | - Viktor Brabec
- Department of Organic Chemistry and Department of Inorganic Chemistry, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., CZ-61265 Brno, Czech Republic, and Dipartimento Farmaco Chimico and Dipartimento di Scienze Biomediche e Oncologia Umana, Universitá degli Studi di Bari, Bari, Italy
| | - Angela Boccarelli
- Department of Organic Chemistry and Department of Inorganic Chemistry, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., CZ-61265 Brno, Czech Republic, and Dipartimento Farmaco Chimico and Dipartimento di Scienze Biomediche e Oncologia Umana, Universitá degli Studi di Bari, Bari, Italy
| | - Mauro Coluccia
- Department of Organic Chemistry and Department of Inorganic Chemistry, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., CZ-61265 Brno, Czech Republic, and Dipartimento Farmaco Chimico and Dipartimento di Scienze Biomediche e Oncologia Umana, Universitá degli Studi di Bari, Bari, Italy
| | - Giovanni Natile
- Department of Organic Chemistry and Department of Inorganic Chemistry, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., CZ-61265 Brno, Czech Republic, and Dipartimento Farmaco Chimico and Dipartimento di Scienze Biomediche e Oncologia Umana, Universitá degli Studi di Bari, Bari, Italy
| |
Collapse
|
65
|
Casini A, Guerri A, Gabbiani C, Messori L. Biophysical characterisation of adducts formed between anticancer metallodrugs and selected proteins: new insights from X-ray diffraction and mass spectrometry studies. J Inorg Biochem 2008; 102:995-1006. [PMID: 18289690 DOI: 10.1016/j.jinorgbio.2007.12.022] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 12/21/2007] [Accepted: 12/24/2007] [Indexed: 10/22/2022]
Abstract
There is considerable interest today for the reactions of anticancer metallodrugs with proteins as these interactions might feature processes that are crucial for the biodistribution, the toxicity and even the mechanism of action of this important group of anticancer agents. We survey here the results of research activities carried out in our "Laboratory of Metals in Medicine" (Department of Chemistry, University of Florence) during the last three years, concerning the molecular characterisation of adducts formed between platinum, ruthenium and gold metallodrugs and a few model proteins. Valuable structural and functional information on these adducts could be derived from several biophysical studies mainly relying on the application of X-ray diffraction and ESI MS techniques. The value and the limitations of both approaches are outlined through a number of examples. Remarkably, the structural and functional information achieved on the respective metallodrug-protein adducts allowed us to identify some general trends in the reactivity of anticancer metallodrugs with protein targets.
Collapse
Affiliation(s)
- Angela Casini
- Laboratory of Metals in Medicine, Department of Chemistry, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Firenze, Italy.
| | | | | | | |
Collapse
|
66
|
Abstract
When the antitumor activity of cisplatin was discovered, no one would have thought of the existence of specific proteins able to transport Pt across the cell membrane or to specifically recognize DNA modified by this drug. However, such proteins do exist and, furthermore, are specific for the Pt substrate considered. It follows that proteins are deeply involved in managing the biological activity of cisplatin. It is expected that, after the first 20 years in which most of the efforts were devoted to understanding its mode of interaction with DNA and consequent structural and functional alterations, the role of proteins will be more deeply scavenged. How cisplatin can survive the attack of the many platinophiles present in the extracellular and intracellular media is the issue addressed in this article. Significantly, differences are observed between cisplatin, carboplatin, and oxaliplatin.
Collapse
|
67
|
Knipp M, Karotki AV, Chesnov S, Natile G, Sadler PJ, Brabec V, Vasák M. Reaction of Zn7metallothionein with cis- and trans-[Pt(N-donor)2Cl2] anticancer complexes: trans-Pt(II) complexes retain their N-donor ligands. J Med Chem 2007; 50:4075-86. [PMID: 17665893 DOI: 10.1021/jm070271l] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Intrinsic and acquired resistance are major drawbacks of platinum-based cancer therapy. The protein superfamily of cysteine- and ZnII-rich proteins, metallothioneins (MT), efficiently inactivate these antitumor drugs because of the strong reactivity of platinum compounds with S-donor molecules. In this study the reactions of human Zn7MT-2 with twelve cis/trans-[Pt(N-donor)2Cl2] compounds and [Pt(dien)Cl]Cl, including new generation drugs, were investigated and the products characterized. A comparison of reaction kinetics revealed that trans-PtII compounds react faster with Zn7MT-2 than cis-PtII compounds. The characterization of the products showed that while all ligands in cis-PtII compounds were replaced by cysteine thiolates, trans-PtII compounds retained their N-donor ligands, thus remaining in a potentially active form. These results provide an increased understanding of the role of MT in the acquired resistance to platinum-based anticancer drugs.
Collapse
Affiliation(s)
- Markus Knipp
- Department of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
68
|
Timerbaev AR, Hartinger CG, Aleksenko SS, Keppler BK. Interactions of antitumor metallodrugs with serum proteins: advances in characterization using modern analytical methodology. Chem Rev 2007; 106:2224-48. [PMID: 16771448 DOI: 10.1021/cr040704h] [Citation(s) in RCA: 523] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Andrei R Timerbaev
- Institute of Inorganic Chemistry, University of Vienna, Währinger Strasse 42, A-1090 Vienna, Austria
| | | | | | | |
Collapse
|
69
|
ESTEBANFERNANDEZ D, GOMEZGOMEZ M, CANAS B, VERDAGUER J, RAMIREZ R, PALACIOS M. Speciation analysis of platinum antitumoral drugs in impacted tissues. Talanta 2007; 72:768-73. [DOI: 10.1016/j.talanta.2006.12.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Revised: 11/21/2006] [Accepted: 12/04/2006] [Indexed: 10/23/2022]
|
70
|
Tao Z, Goodisman J, Penefsky HS, Souid AK. Caspase activation by anticancer drugs: the caspase storm. Mol Pharm 2007; 4:583-95. [PMID: 17439154 DOI: 10.1021/mp070002r] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This study measures the time-dependence of cellular caspase activation by anticancer drugs and compares it with that of cellular respiration. Intracellular caspase activation and cellular respiration were measured during continuous exposure of Jurkat, HL-60, and HL-60/MX2 (deficient in topoisomerase-II) cells to dactinomycin, doxorubicin, and the platinum (Pt) compounds cisplatin, carboplatin, and oxaliplatin. Caspase activation was measured using the fluorogenic compound N-acetyl-asp-glu-val-asp-7-amino-4-trifluoromethyl coumarin (Ac-DEVD-AFC). We show that this substrate rapidly enters cells where it is efficiently cleaved at the aspartate residue by specific caspases, yielding the fluorescent compound 7-amino-4-trifluoromethyl coumarin (AFC). Following cell disruption, released AFC was separated on HPLC and detected by fluorescence. The appearance of AFC in cells was blocked by the pancaspase inhibitor benzyloxycarbonyl-val-ala-asp-fluoromethylketone, thus establishing that intracellular caspases were responsible for the cleavage. Caspase activity was first noted after about 2 h of incubation with doxorubicin or dactinomycin, the production of AFC being linear with time afterward. Caspase activation by doxorubicin was delayed in HL-60/MX2 cells, reflecting the critical role of topoisomerase-II in doxorubicin cytotoxicity. For both drugs, caspase activity increased rapidly between approximately 2 and approximately 6 h, went through a maximum, and decreased after approximately 8 h ("caspase storm"). Cisplatin treatment induced noticeable caspase activity only after approximately 14 h of incubation, and the fluorescent intensity of AFC became linear with time at approximately 16 h. Exposure of the cells to all of the drugs studied led to impaired cellular respiration and decreased cellular ATP, concomitant with caspase activation. Thus, the mitochondria are rapidly targeted by active caspases.
Collapse
Affiliation(s)
- Zhimin Tao
- Department of Chemistry, Syracuse University, 1-014 CST, Syracuse, New York 13244, USA
| | | | | | | |
Collapse
|
71
|
Surowiak P, Materna V, Maciejczyk A, Pudełko M, Markwitz E, Spaczyński M, Dietel M, Zabel M, Lage H. Nuclear metallothionein expression correlates with cisplatin resistance of ovarian cancer cells and poor clinical outcome. Virchows Arch 2007; 450:279-85. [PMID: 17235562 DOI: 10.1007/s00428-006-0362-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Revised: 12/07/2006] [Accepted: 12/18/2006] [Indexed: 11/26/2022]
Abstract
Elevated metallothionein (MT) expression in ovarian cancers treated with cisplatin-based schemes represents an unfavorable prognostic index. MT expression is significantly higher in tumor samples obtained after chemotherapy. The present study aimed at examining MT expression in ovarian carcinoma cells sensitive (A2780) or resistant (A2780RCIS) against platinum drug treatment as well as examining effects of exposure to cisplatin on MT expression. Subcellular expression of MT was evaluated also in samples originating from 73 ovarian tumors. Cisplatin-resistant A2780RCIS cells were exposed to increasing cisplatin concentrations, and the subcellular expression of MT was determined by immunocytochemistry. The studies demonstrated that cisplatin-resistant A2780RCIS cells exposed to cisplatin typically manifested a nuclear MT expression. The study demonstrated also that exposure to cisplatin was paralleled by growing MT expression in cell nuclei. The nuclear expression of MT was also found to be specific for ovarian cancers of poor clinical outcome. No relationship could be demonstrated between cytoplasmic expression of MT and clinical variables. Nuclear MT expression is induced by cisplatin and seems to protect DNA in the cells from toxic effects of the drug.
Collapse
Affiliation(s)
- Paweł Surowiak
- Institute of Pathology, Charité Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Kaspárková J, Nováková O, Vrána O, Intini F, Natile G, Brabec V. Molecular Aspects of Antitumor Effects of a New Platinum(IV) Drug. Mol Pharmacol 2006; 70:1708-19. [PMID: 16896071 DOI: 10.1124/mol.106.027730] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The new platinum(IV) complex cis,trans,cis-[PtCl(2)(CH(3)COO)(2)-(NH(3))(1-adamantylamine)] [adamplatin(IV)] seems promising for the perspective application in therapy of corresponding tumors. It is therefore of great interest to understand details of mechanisms underlying its biological efficacy. Cellular uptake of the drug, alterations in the target DNA induced by platinum drugs along with processing of platinum-induced damage to DNA and drug inactivation by sulfur-containing compounds belong to major pharmacological factors affecting antitumor effects of platinum compounds. We examined in the present work the significance of these factors in the mechanism of antitumor effects of adamplatin(IV) and compared the results with those of the parallel studies performed with "conventional" cisplatin. The results show that deactivation of adamplatin(IV) by sulfur-containing compounds (such as glutathione or metallothioneins) is likely to play a less significant role in the mechanism of resistance of tumor cells to adamplatin(IV) in contrast to the role of these reactions in the effects of cisplatin. Moreover, the treatment of tumor cells with adamplatin(IV) does not result in DNA modifications that would be markedly different from those produced by cisplatin. In contrast, the effects of other factors, such as enhanced accumulation of the drug in cells, strong inhibition of DNA polymerization by these adducts, lowered DNA repair, and DNA-protein cross-linking are different from the effects of these factors in the mechanism underlying activity of cisplatin. Hence, the differences between effects of adamplatin(IV) and cisplatin observed in the present work on molecular level may help understand the unique activity of adamplatin(IV).
Collapse
Affiliation(s)
- Jana Kaspárková
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, CZ-61265 Brno, Czech Republic
| | | | | | | | | | | |
Collapse
|
73
|
Waalkes MP, Liu J, Kasprzak KS, Diwan BA. Hypersusceptibility to cisplatin carcinogenicity in metallothionein-I/II double knockout mice: production of hepatocellular carcinoma at clinically relevant doses. Int J Cancer 2006; 119:28-32. [PMID: 16432836 DOI: 10.1002/ijc.21245] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Metallothionein (MT) is a high-affinity metal binding protein thought to mitigate the toxicity of various metals. Cisplatin is a widely used cancer chemotherapeutic that is a rodent carcinogen and may have carcinogenic potential in humans. MT seems to reduce cisplatin toxicity by binding the metal compound but how MT deficiency might impact the carcinogenic effects of cisplatin is unknown. Thus, groups (n = 25) of male MT-I/II double knockout (MT-null) or MT wild-type (WT) mice were exposed to a single treatment of cisplatin (5 or 10 mg/kg, i.p.), or left untreated (control) and observed over the next 104 weeks. The doses of cisplatin used equate to only a fraction of the total dose used typically in clinical settings. In cisplatin-treated MT-null mice, a dose-related increase in hepatocellular carcinoma (HCC) occurred (control, 0%; 5 mg/kg, 17%; 10 mg/kg, 36%) that was not seen in WT mice. Similarly, liver carcinoma multiplicity (HCC/liver) was increased markedly by cisplatin but only in MT-null mice, indicating the formation of multiple primaries in MT deficient mice. Harderian gland carcinoma incidence was also increased by cisplatin treatment in MT-null mice but not WT mice. Our results indicate that MT-null mice are hypersusceptible to the hepatocarcinogenic effects of cisplatin, and poor MT expression may be a predisposing factor for cisplatin-induced secondary tumors after chemotherapy.
Collapse
Affiliation(s)
- Michael P Waalkes
- Inorganic Carcinogenesis Section, Laboratory of Comparative Carcinogenesis, National Cancer Institute at the National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| | | | | | | |
Collapse
|
74
|
Ramos-Lima FJ, Vrána O, Quiroga AG, Navarro-Ranninger C, Halámiková A, Rybnícková H, Hejmalová L, Brabec V. Structural characterization, DNA interactions, and cytotoxicity of new transplatin analogues containing one aliphatic and one planar heterocyclic amine ligand. J Med Chem 2006; 49:2640-51. [PMID: 16610807 DOI: 10.1021/jm0602514] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We report in the present work new analogues of clinically ineffective transplatin in which one ammine group was replaced by aliphatic and the other by a planar heterocyclic ligand, namely trans-[PtCl(2)(isopropylamine)(3-(hydroxymethyl)-pyridine)], 1, and trans-[PtCl(2)(isopropylamine)(4-(hydroxymethyl)-pyridine)], 2. The new compounds, in comparison with parent transplatin, exhibit radically enhanced activity in tumor cell lines both sensitive and in particular resistant to cisplatin. Concomitantly, the DNA binding mode of 1 and 2 compared to parent transplatin and other antitumor analogues of transplatin in which only one ammine group was replaced is also different. The results also suggest that the reactions of glutathione and metallothionein-2 with compounds 1 and 2 do not play a crucial role in their overall biological effects. In addition, the monofunctional adducts of 1 and 2 are quenched by glutathione considerably less than the adducts of transplatin, which may potentiate cytotoxic effects of these new platinum complexes.
Collapse
Affiliation(s)
- Francisco J Ramos-Lima
- Institute of Biophysics, Academy of Sciences of the Czech Republic, CZ-61265 Brno, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Goodisman J, Hagrman D, Tacka KA, Souid AK. Analysis of cytotoxicities of platinum compounds. Cancer Chemother Pharmacol 2005; 57:257-67. [PMID: 16028101 DOI: 10.1007/s00280-005-0041-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2005] [Accepted: 04/05/2005] [Indexed: 10/25/2022]
Abstract
Extent of DNA platination, loss of cell viability, DNA fragmentation, and impairment of cellular mitochondrial oxygen consumption are measures of drug cytotoxicity. We measured and compared these effects for cisplatin, oxaliplatin and carboplatin. Because reaction with intracellular thiols may be responsible for drug resistance, we also determined the rates of Pt drug reactions with metallothionein. Jurkat cells were exposed at 37 degrees C to 25 microM Pt drugs for 3 h. Pt-DNA adducts were determined at the end of the incubation period by atomic absorption spectroscopy. Viability, DNA fragmentation, and cellular respiration (microM O2/min/10(6) cells) were determined 24 h post drug exposure. The average amount of Pt-DNA adducts (Pt atoms/10(6) nucleotides) produced by cisplatin was 43.4, by oxaliplatin 4.8 and by carboplatin 1.5. Cisplatin decreased the rate of respiration by approximately 63% and oxaliplatin by approximately 37%. DNA fragmentation by cisplatin and oxaliplatin was very similar. Carboplatin produced an unnoticeable effect on cellular respiration, and only approximately 10% of the DNA fragmentation was produced by cisplatin or oxaliplatin. Although, for a given drug, all four measures of cytotoxicity were proportional, this did not hold for comparisons between the drugs. The rate constants (M-1 s-1) for reaction of cisplatin, oxaliplatin and carboplatin with Cd/Zn thionein were 0.75, 0.44 and 0.012, respectively. For comparison, the rate constants (M-1 s-1) for reaction of cisplatin, oxaliplatin and carboplatin with glutathione were 0.027, 0.038 and 0.0012, respectively. The low reactivity of carboplatin with metallothionein and glutathione suggests that its low cytotoxic activities are not due to reaction of Pt2+ with cellular thiols. Despite a tenfold difference in Pt-DNA adducts between cisplatin and oxaliplatin, the cytotoxicities of these compounds are very similar, suggesting that oxaliplatin lesions are more potent than cisplatin lesions. The results demonstrate a large influence of the ligands occupying Pt coordination spheres on the chemical and biologic activities of Pt drugs.
Collapse
Affiliation(s)
- Jerry Goodisman
- Department of Chemistry, Syracuse University, 1-014 CST, Syracuse, NY 13244, USA
| | | | | | | |
Collapse
|
76
|
Surowiak P, Materna V, Kaplenko I, Spaczyński M, Dietel M, Lage H, Zabel M. Augmented expression of metallothionein and glutathione S-transferase pi as unfavourable prognostic factors in cisplatin-treated ovarian cancer patients. Virchows Arch 2005; 447:626-33. [PMID: 15968547 DOI: 10.1007/s00428-005-1228-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2004] [Accepted: 02/09/2005] [Indexed: 11/25/2022]
Abstract
Resistance to cis- or carboplatin represents the principal cause of therapeutic failures in ovarian carcinoma. The phenomenon of resistance to platinum-based drugs is partly related to expression of metallothionein (MT) and of glutathione S-transferase pi (GST-pi), but opinion on the subject is discordant. Documentation of a negative predictive effect of MT and GST-pi expression for the therapy employing platinum-based drugs would permit to select resistant cases in which other therapeutic approaches could be employed. The present study aimed at examining the relation between intensities of MT and GST-pi expressions in ovarian carcinomas and dynamics of the clinical course in the neoplastic disease in a group of cisplatin-treated patients. The analyses were performed on samples of ovarian carcinoma originating from 43 first-look laparotomies (FLLs) and, in 30 cases, from second-look laparotomies (SLL) from the same patients. Immunohistochemical reactions were performed on paraffin sections of studied tumors, using monoclonal antibodies to MT and GST-pi. The calculations showed that in cases with augmented expression of MT, mortality was higher. On the other hand, augmented expression of GST-pi predisposed to more frequent relapses, deaths and progression of the tumor. Kaplan-Meier analysis showed that a significantly shorter survival time was linked to cases of higher expression of MT at FLL and of higher expression of GST-pi at FLL, whereas a shorter progression-free time was manifested by cases with higher expression of GST-pi at FLL. The performed investigations indicate that augmented expressions of MT at FLL and GST-pi at FLL in ovarian cancer represent an unfavourable predictive factor in cisplatin-treated patients.
Collapse
Affiliation(s)
- Paweł Surowiak
- Department of Histology and Embryology, University School of Medicine, ul. Chałubińskiego 6a, 50-356 Wrocław, Poland
| | | | | | | | | | | | | |
Collapse
|
77
|
Vickers AEM, Rose K, Fisher R, Saulnier M, Sahota P, Bentley P. Kidney slices of human and rat to characterize cisplatin-induced injury on cellular pathways and morphology. Toxicol Pathol 2005; 32:577-90. [PMID: 15603542 DOI: 10.1080/01926230490508821] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Kidney slices represent an in vitro model that has the cellular complexity of in vivo tissue to provide insights into mechanisms of organ injury, as shown in this study with the model nephrotoxicant cisplatin. Cell pathways altered by cisplatin exposure are assessed by gene expression analysis, cell function, and morphology in human and rat kidney slices in comparison to rat kidney from an in vivo study. The acute nephrosis of the tubular epithelium induced by cisplatin in vivo was reproduced in both human and rat kidney slices, while the glomerulus appeared resistant even at high concentrations. Kidney gene expression changes of in vivo and in vitro samples were indicative of transcription, DNA damage, cell cycle, proliferation, and apoptosis that are in agreement with the mechanism of cisplatin causing DNA damage, growth arrest, and apoptosis; while genes indicative of protein damage, the disruption of transport and calcium homeostasis, cellular metabolism, and oxidative stress are pathways linked with cisplatin binding to various cellular proteins and macromolecules. Both concentration and time-dependent gene expression changes evident in the in vitro model preceded a change in tissue morphology. Functional assays confirming cell dysfunction and increased apoptosis revealed the rat kidney to be more sensitive to the effects of cisplatin than human kidney as demonstrated by significant decreases in slice ATP and GSH levels, significant increases in caspase 9 and 3 activity, p53 protein levels, and increased DNA laddering. The regional markers of proximal and distal tubular injury, alpha- and pi-glutathione S-transferases, were shown for the human kidney slices to be significantly increased by cisplatin. In this study, cisplatin-induced nephrotoxicity was demonstrated morphologically in rat and human kidney slices, and the associated gene expression and functional changes characterized the cellular pathways involved.
Collapse
Affiliation(s)
- Alison E M Vickers
- Preclinical Safety, Novartis Pharmaceuticals Corporation, One Health Plaza, E. Hanover, New Jersey 07936, USA.
| | | | | | | | | | | |
Collapse
|
78
|
Hasinoff BB, Wu X, Krokhin OV, Ens W, Standing KG, Nitiss JL, Sivaram T, Giorgianni A, Yang S, Jiang Y, Yalowich JC. Biochemical and proteomics approaches to characterize topoisomerase IIalpha cysteines and DNA as targets responsible for cisplatin-induced inhibition of topoisomerase IIalpha. Mol Pharmacol 2004; 67:937-47. [PMID: 15602006 DOI: 10.1124/mol.104.004416] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cisplatin was shown to strongly inhibit the decatenation and relaxation activity of isolated human DNA topoisomerase IIalpha. This inhibition was not accompanied by stabilization of a covalent topoisomerase IIalpha-DNA intermediate. Pretreatment of kinetoplast plasmid DNA (kDNA) or pBR322 DNA with submicromolar concentrations of cisplatin quickly rendered these substrates incompetent in the topoisomerase IIalpha catalytic assay. Cisplatin nearly equally inhibited growth of a parental K562 and an etoposide-resistant K/VP.5 cell line that contained decreased topoisomerase IIalpha levels, a result consistent with isolated enzyme experiments demonstrating that cisplatin was not a topoisomerase IIalpha poison. Because cisplatin is known to react with protein sulfhydryl groups, the 13 cysteine groups in the topoisomerase IIalpha monomer were evaluated by mass spectrometry to determine which cysteines were free and disulfide-bonded to identify possible sites of cisplatin adduction. High-pressure liquid chromatography-matrix-assisted laser desorption ionization mass spectrometry showed that topoisomerase IIalpha contained at least five free cysteines (170, 216, 300, 392, and 405) and two disulfide-bonded cysteine pairs (427-455 and 997-1008). Cysteine 733 was also disulfide-bonded, but its partner cysteine could not be identified. Cisplatin antagonized the formation of a fluorescence adduct between topoisomerase IIalpha and the sulfhydryl-reactive maleimide reagent 10-(2,5-dihydro-2,5-dioxo-1H-pyrrol-1-yl)-9-methoxy-3-oxo-3H-naphtho[2,1-b]pyran-2-carboxylic acid methyl ester (ThioGlo-1). Dithiothreitol, which was shown by spectrophotometry to react rapidly with cisplatin (6-min half-time), diminished the capacity of cisplatin to interfere with ThioGlo-1 binding to topoisomerase IIalpha. The results of this study suggest that cisplatin may exert some of its cell growth inhibitory and antitumor activity by inhibition of topoisomerase IIalpha through reaction with critical enzyme sulfhydryl groups and/or by forming DNA adducts that render the DNA substrate refractory to topoisomerase IIalpha.
Collapse
Affiliation(s)
- Brian B Hasinoff
- Faculty of Pharmacy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Tacka KA, Dabrowiak JC, Goodisman J, Penefsky HS, Souid AK. Effects of Cisplatin on Mitochondrial Function in Jurkat Cells. Chem Res Toxicol 2004; 17:1102-11. [PMID: 15310242 DOI: 10.1021/tx0499564] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In this work, we measured the effects of pharmacological concentrations of cisplatin (cis-diaminedichloroplatinum II) on mitochondrial function, cell viability, and DNA fragmentation in Jurkat cells. The exposure of cells to 0-25 microM cisplatin for 3 h had no immediate effect on cellular mitochondrial oxygen consumption, measured using a palladium-porphyrin oxygen sensing phosphor. Similarly, the cell viability as measured by trypan blue staining was unchanged immediately following exposure to the drug, and no small DNA fragments, characteristic of drug-induced apoptosis, appeared. At 24 h after exposure to cisplatin, cellular respiration and viability decreased relative to controls and the amount of small DNA fragments, measured using quantitative agarose gel electrophoresis, was proportional to the concentration of cisplatin present during the drug exposure period. The small DNA fragments showed the banding pattern (with a spacing of approximately 300 bp) characteristic of drug-induced cell death by apoptosis. The changes in respiration and DNA fragmentation correlated linearly with the amount of platinum bound to DNA, determined by atomic absorption spectroscopy immediately following drug exposure. The oxygen consumption by beef heart mitochondria was not affected 0-24 h after exposure to 25 microM cisplatin or to solutions containing the monoaquated form of the drug, suggesting that the drug does not attack the mitochondrial respiratory chain directly. Cells exposed to the peptide benzyloxycarbonyl-val-ala-asp-fluoromethyl ketone, which blocks apoptosis by the caspase pathway, showed a decrease in cisplatin-induced DNA fragmentation but not in the impairment of cellular respiration. Thus, although apoptosis is caspase-dependent, the impairment of cellular respiration is independent of the caspase system. Collectively, these results suggest that alteration in mitochondrial function is a secondary effect of cisplatin cytotoxicity in Jurkat cells.
Collapse
Affiliation(s)
- Kirk A Tacka
- Department of Chemistry, Syracuse University, CST 1-014, Syracuse, New York 13244-4100, USA
| | | | | | | | | |
Collapse
|
80
|
Wilson LA, Yamamoto H, Singh G. Role of the transcription factor Ets-1 in cisplatin resistance. Mol Cancer Ther 2004. [DOI: 10.1158/1535-7163.823.3.7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Cisplatin is a DNA damaging agent widely used as a chemotherapeutic agent. A major limitation of the use of this agent is the development of drug resistance within tumors. Several in vitro models exist which enable the investigation of resistance mechanisms, including 2008/C13* ovarian carcinoma cells. C13* cells are variants of 2008 cells, displaying cisplatin resistance following 13 consecutive cisplatin treatments. This model system has led to the identification of several mechanisms that play parts in the multifactorial nature of cisplatin resistance. In this study, we have examined the contribution of a transcription factor, Ets-1, to the cisplatin resistance of C13* cells. Ets-1 is up-regulated in C13* cells as compared with the cisplatin-sensitive 2008 cells and overexpression of this protein in 2008 cells led to a 7-fold increase in resistance. Further studies on a colorectal carcinoma cell line overexpressing Ets-1 indicated that this phenomenon is not cell specific—increased cisplatin resistance correlated to Ets-1 expression. The mechanism of cisplatin resistance elicited by Ets-1 is potentially via transcriptional activation of genes whose products have well-described functions in reducing cisplatin toxicity. Examples, identified via microarray analysis, include metallothioneins and DNA repair enzymes. This is the first report to our knowledge associating expression of Ets-1, a transcription factor whose expression often signals poor prognosis in various cancer types, to cisplatin resistance.
Collapse
Affiliation(s)
- Leigh A. Wilson
- 1Juravinski Cancer Center and Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada and
| | - Hirotaka Yamamoto
- 2The Chicago Institute of Neurosurgery and Neuroresearch, Chicago, Illinois
| | - Gurmit Singh
- 1Juravinski Cancer Center and Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada and
| |
Collapse
|
81
|
Hagrman D, Goodisman J, Souid AK. Kinetic Study on the Reactions of Platinum Drugs with Glutathione. J Pharmacol Exp Ther 2003; 308:658-66. [PMID: 14610218 DOI: 10.1124/jpet.103.059410] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The binding of platinum (Pt) drugs (oxaliplatin, carboplatin, and cisplatin) to glutathione (GSH, 6.75 mM) was investigated at 37 degrees C in Hepes (100 mM, pH approximately 7.4) or Tris-NO(3) (60 mM, pH 7.4) buffer and NaCl (4.62, 6.63, or 7.82 mM). The conditions were chosen to mimic passage of clinical concentrations of the drugs (135 microM) through the cytosol. The reactions were monitored by UV-absorption spectroscopy, high-performance liquid chromatography (HPLC), and atomic absorption spectroscopy. The initial rates, detected by UV absorbance, were similar for oxaliplatin and cisplatin reacting with GSH and were more than 5-fold faster than for carboplatin reacting with GSH. The Pt contents in HPLC eluates corresponding to unbound drug decreased exponentially with time, confirming that the reactions were first order in [Pt drug] and allowing determination of the pseudo first-order rate constants (k(1)). The second-order rate constants (k(2)) were calculated as k(1) divided by [GSH]. The k(2) value for oxaliplatin reacting with GSH was approximately 3.8 x 10(-2) M(-1) s(-1), for cisplatin reacting with GSH approximately 2.7 x 10(-2) M(-1) s(-1), and for carboplatin reacting with GSH approximately 1.2 x 10(-3) M(-1) s(-1) (approximately 32-fold slower than that of oxaliplatin and approximately 23-fold slower than that of cisplatin). These results demonstrate an influence of ligands surrounding the Pt coordination sphere on the reactivity of Pt(2+) with GSH.
Collapse
Affiliation(s)
- Douglas Hagrman
- Department of Pediatrics, Upstate Medical University, State University of New York, 750 East Adams St., Syracuse, NY 13210, USA
| | | | | |
Collapse
|