Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Hum Mutat ; 38(2): 193-203, 2017 02.
Article in English | MEDLINE | ID: mdl-27859906

ABSTRACT

Heritable loss of function mutations in the human RECQ helicase genes BLM, WRN, and RECQL4 cause Bloom, Werner, and Rothmund-Thomson syndromes, cancer predispositions with additional developmental or progeroid features. In order to better understand RECQ pathogenic and population variation, we systematically analyzed genetic variation in all five human RECQ helicase genes. A total of 3,741 unique base pair-level variants were identified, across 17,605 potential mutation sites. Direct counting of BLM, RECQL4, and WRN pathogenic variants was used to determine aggregate and disease-specific carrier frequencies. The use of biochemical and model organism data, together with computational prediction, identified over 300 potentially pathogenic population variants in RECQL and RECQL5, the two RECQ helicases that are not yet linked to a heritable deficiency syndrome. Despite the presence of these predicted pathogenic variants in the human population, we identified no individuals homozygous for any biochemically verified or predicted pathogenic RECQL or RECQL5 variant. Nor did we find any individual heterozygous for known pathogenic variants in two or more of the disease-associated RECQ helicase genes BLM, RECQL4, or WRN. Several postulated RECQ helicase deficiency syndromes-RECQL or RECQL5 loss of function, or compound haploinsufficiency for the disease-associated RECQ helicases-may remain missing, as they likely incompatible with life.


Subject(s)
Genetic Association Studies , Genetics, Population , Mutation , RecQ Helicases/genetics , Computational Biology/methods , DNA Mutational Analysis , Databases, Nucleic Acid , Gene Frequency , Genetic Predisposition to Disease , Genotype , Humans , ROC Curve , Software , Web Browser
2.
Sci Rep ; 6: 24165, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27087647

ABSTRACT

Galectin-1 (Gal-1) dimers crosslink carbohydrates on cell surface receptors. Carbohydrate-derived inhibitors have been developed for cancer treatment. Intracellularly, Gal-1 was suggested to interact with the farnesylated C-terminus of Ras thus specifically stabilizing GTP-H-ras nanoscale signalling hubs in the membrane, termed nanoclusters. The latter activity may present an alternative mechanism for how overexpressed Gal-1 stimulates tumourigenesis. Here we revise the current model for the interaction of Gal-1 with H-ras. We show that it indirectly forms a complex with GTP-H-ras via a high-affinity interaction with the Ras binding domain (RBD) of Ras effectors. A computationally generated model of the Gal-1/C-Raf-RBD complex is validated by mutational analysis. Both cellular FRET as well as proximity ligation assay experiments confirm interaction of Gal-1 with Raf proteins in mammalian cells. Consistently, interference with H-rasG12V-effector interactions basically abolishes H-ras nanoclustering. In addition, an intact dimer interface of Gal-1 is required for it to positively regulate H-rasG12V nanoclustering, but negatively K-rasG12V nanoclustering. Our findings suggest stacked dimers of H-ras, Raf and Gal-1 as building blocks of GTP-H-ras-nanocluster at high Gal-1 levels. Based on our results the Gal-1/effector interface represents a potential drug target site in diseases with aberrant Ras signalling.


Subject(s)
Galectin 1/metabolism , raf Kinases/metabolism , ras Proteins/metabolism , Animals , Cell Line , Computer Simulation , Cricetinae , Dimerization , Galectin 1/chemistry , Galectin 1/genetics , Humans , Mutation , Protein Binding , Signal Transduction , raf Kinases/genetics , ras Proteins/chemistry , ras Proteins/genetics
3.
Elife ; 4: e08905, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26274561

ABSTRACT

Hotspot mutations of Ras drive cell transformation and tumorigenesis. Less frequent mutations in Ras are poorly characterized for their oncogenic potential. Yet insight into their mechanism of action may point to novel opportunities to target Ras. Here, we show that several cancer-associated mutations in the switch III region moderately increase Ras activity in all isoforms. Mutants are biochemically inconspicuous, while their clustering into nanoscale signaling complexes on the plasma membrane, termed nanocluster, is augmented. Nanoclustering dictates downstream effector recruitment, MAPK-activity, and tumorigenic cell proliferation. Our results describe an unprecedented mechanism of signaling protein activation in cancer.


Subject(s)
Cell Transformation, Neoplastic , Mutation , Neoplasms/pathology , ras Proteins/genetics , ras Proteins/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cell Proliferation , Humans , Protein Multimerization , Signal Transduction
4.
Anal Chem ; 87(6): 3527-34, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25707436

ABSTRACT

GTPases are central cellular signaling proteins, which cycle between a GDP-bound inactive and a GTP-bound active conformation in a controlled manner. Ras GTPases are frequently mutated in cancer and so far only few experimental inhibitors exist. The most common methods for monitoring GTP hydrolysis rely on luminescent GDP- or GTP-analogs. In this study, the first GTP-specific Fab fragment and its application are described. We selected Fab fragments using the phage display technology. Six Fab fragments were found against 2'/3'-GTP-biotin and 8-GTP-biotin. Selected antibody fragments allowed specific detection of endogenous, free GTP. The most potent Fab fragment (2A4(GTP)) showed over 100-fold GTP-specificity over GDP, ATP, or CTP and was used to develop a heterogeneous time-resolved luminescence based assay for the monitoring of GTP concentration. The method allows studying the GEF dependent H-Ras activation (GTP binding) and GAP-catalyzed H-Ras deactivation (GTP hydrolysis) at nanomolar protein concentrations.


Subject(s)
Antibody Specificity , Enzyme Assays/methods , GTP Phosphohydrolases/metabolism , Guanosine Triphosphate/immunology , Guanosine Triphosphate/metabolism , Immunoglobulin Fab Fragments/immunology , Enzyme Activation , Enzyme Inhibitors/pharmacology , GTP Phosphohydrolases/antagonists & inhibitors , GTPase-Activating Proteins/metabolism , Humans , Hydrolysis
5.
Anal Bioanal Chem ; 406(17): 4147-56, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24760397

ABSTRACT

A quenching resonance energy transfer (QRET) assay for small GTPase nucleotide exchange kinetic monitoring is demonstrated using nanomolar protein concentrations. Small GTPases are central signaling proteins in all eukaryotic cells acting as a "molecular switches" that are active in the GTP-state and inactive in the GDP-state. GTP-loading is highly regulated by guanine nucleotide exchange factors (GEFs). In several diseases, most prominently cancer, this process in misregulated. The kinetics of the nucleotide exchange reaction reports on the enzymatic activity of the GEF reaction system and is, therefore, of special interest. We determined the nucleotide exchange kinetics using europium-labeled GTP (Eu-GTP) in the QRET assay for small GTPases. After GEF catalyzed GTP-loading of a GTPase, a high time-resolved luminescence signal was found to be associated with GTPase bound Eu-GTP, whereas the non-bound Eu-GTP fraction was quenched by soluble quencher. The association kinetics of the Eu-GTP was measured after GEF addition, whereas the dissociation kinetics could be determined after addition of unlabeled GTP. The resulting association and dissociation rates were in agreement with previously published values for H-Ras(Wt), H-Ras(Q61G), and K-Ras(Wt), respectively. The broader applicability of the QRET assay for small GTPases was demonstrated by determining the kinetics of the Ect2 catalyzed RhoA(Wt) GTP-loading. The QRET assay allows the use of nanomolar protein concentrations, as more than 3-fold signal-to-background ratio was achieved with 50 nM GTPase and GEF proteins. Thus, small GTPase exchange kinetics can be efficiently determined in a HTS compatible 384-well plate format.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , GTP Phosphohydrolases/chemistry , Nucleotides/chemistry , Animals , Energy Transfer , Humans
6.
J Biol Chem ; 289(14): 9519-33, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24569991

ABSTRACT

Solution structures and biochemical data have provided a wealth of mechanistic insight into Ras GTPases. However, information on how much the membrane organization of these lipid-modified proteins impacts on their signaling is still scarce. Ras proteins are organized into membrane nanoclusters, which are necessary for Ras-MAPK signaling. Using quantitative conventional and super-resolution fluorescence methods, as well as mathematical modeling, we investigated nanoclustering of H-ras helix α4 and hypervariable region mutants that have different bona fide conformations on the membrane. By following the emergence of conformer-specific nanoclusters in the plasma membrane of mammalian cells, we found that conformers impart distinct nanoclustering responses depending on the cytoplasmic levels of the nanocluster scaffold galectin-1. Computational modeling revealed that complexes containing H-ras conformers and galectin-1 affect both the number and lifetime of nanoclusters and thus determine the specific Raf effector recruitment. Our results show that mutations in Ras can affect its nanoclustering response and thus allosterically effector recruitment and downstream signaling. We postulate that cancer- and developmental disease-linked mutations that are associated with the Ras membrane conformation may exhibit so far unrecognized Ras nanoclustering and therefore signaling alterations.


Subject(s)
Cell Membrane/enzymology , Models, Biological , Oncogene Protein p21(ras)/metabolism , Protein Multimerization , Signal Transduction , raf Kinases/metabolism , Animals , Cell Line , Cell Membrane/genetics , Cricetinae , Galectin 1/genetics , Galectin 1/metabolism , Mice , Mice, Knockout , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/pathology , Oncogene Protein p21(ras)/genetics , Protein Structure, Secondary , raf Kinases/genetics
7.
Int J Oncol ; 43(4): 1269-80, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23903781

ABSTRACT

Berberine is a natural isoquinoline alkaloid with significant antitumor activity against many types of cancer cells, including ovarian tumors. This study investigated the molecular mechanisms by which berberine differently affects cell growth of cisplatin (cDDP)-sensitive and -resistant and polyamine analogue cross-resistant human ovarian cancer cells. The results show that berberine suppresses the growth of cDDP-resistant cells more than the sensitive counterparts, by interfering with the expression of folate cycle enzymes, dihydrofolate reductase (DHFR) and thymidylate synthase (TS). In addition, the impairment of the folate cycle also seems partly ascribable to a reduced accumulation of folate, a vitamin which plays an essential role in the biosynthesis of nucleic acids and amino acids. This effect was observed in both lines, but especially in the resistant cells, correlating again with the reduced tolerance to this isoquinoline alkaloid. The data also indicate that berberine inhibits cellular growth by affecting polyamine metabolism, in particular through the upregulation of the key catabolic enzyme, spermidine/spermine N1-acetyltransferase (SSAT). In this regard, berberine is shown to stimulate the SSAT induction by the spermine analogue N1, N12 bisethylspermine (BESpm), which alone was also able to downregulate DHFR mRNA more than TS mRNA. We report that the sensitivity of resistant cells to cisplatin or to BESpm is reverted to the levels of sensitive cells by the co-treatment with berberine. These data confirm the intimate inter-relationships between folate cycle and polyamine pathways and suggest that this isoquinoline plant alkaloid could be a useful adjuvant therapeutic agent in the treatment of ovarian carcinoma.


Subject(s)
Acetyltransferases/metabolism , Berberine/administration & dosage , Ovarian Neoplasms/drug therapy , Tetrahydrofolate Dehydrogenase/metabolism , Thymidylate Synthase/metabolism , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/biosynthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Polyamines/metabolism , Tetrahydrofolate Dehydrogenase/biosynthesis , Thymidylate Synthase/biosynthesis
8.
Nucleic Acids Res ; 41(7): 4159-70, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23423353

ABSTRACT

Resistance to drugs targeting human thymidylate synthase (TS) poses a major challenge in the field of anti-cancer therapeutics. Overexpression of the TS protein has been implicated as one of the factors leading to the development of resistance. Therefore, repressing translation by targeting the TS mRNA could help to overcome this problem. In this study, we report that the compound Hoechst 33258 (HT) can reduce cellular TS protein levels without altering TS mRNA levels, suggesting that it modulates TS expression at the translation level. We have combined nuclear magnetic resonance, UV-visible and fluorescence spectroscopy methods with docking and molecular dynamics simulations to study the interaction of HT with a region in the TS mRNA. The interaction predominantly involves intercalation of HT at a CC mismatch in the region near the translational initiation site. Our results support the use of HT-like compounds to guide the design of therapeutic agents targeting TS mRNA.


Subject(s)
Antineoplastic Agents/chemistry , Bisbenzimidazole/chemistry , Bisbenzimidazole/pharmacology , Gene Expression Regulation/drug effects , Protein Biosynthesis/drug effects , RNA, Messenger/drug effects , Thymidylate Synthase/genetics , Antineoplastic Agents/pharmacology , Base Pair Mismatch , Binding Sites , Cell Line, Tumor , Humans , Intercalating Agents/chemistry , Intercalating Agents/pharmacology , Models, Molecular , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Thymidylate Synthase/metabolism
9.
PLoS One ; 7(10): e47318, 2012.
Article in English | MEDLINE | ID: mdl-23056627

ABSTRACT

BACKGROUND: 5-fluorouracil, a commonly used chemotherapeutic agent, up-regulates expression of human thymidylate synthase (hTS). Several different regulatory mechanisms have been proposed to mediate this up-regulation in distinct cell lines, but their specific contributions in a single cell line have not been investigated to date. We have established the relative contributions of these previously proposed regulatory mechanisms in the ovarian cancer cell line 2008 and the corresponding cisplatin-resistant and 5-FU cross-resistant-subline C13*. METHODOLOGY/PRINCIPAL FINDINGS: Using RNA polymerase II inhibitor DRB treated cell cultures, we showed that 70-80% of up-regulation of hTS results from transcriptional activation of TYMS mRNA. Moreover, we report that 5-FU compromises the cell cycle by blocking the 2008 and C13* cell lines in the S phase. As previous work has established that TYMS mRNA is synthesized in the S and G(1) phase and hTS is localized in the nuclei during S and G(2)-M phase, the observed cell cycle changes are also expected to affect the intracellular regulation of hTS. Our data also suggest that the inhibition of the catalytic activity of hTS and the up-regulation of the hTS protein level are not causally linked, as the inactivated ternary complex, formed by hTS, deoxyuridine monophosphate and methylenetetrahydrofolate, was detected already 3 hours after 5-FU exposure, whereas substantial increase in global TS levels was detected only after 24 hours. CONCLUSIONS/SIGNIFICANCE: Altogether, our data indicate that constitutive TYMS mRNA transcription, cell cycle-induced hTS regulation and hTS enzyme stability are the three key mechanisms responsible for 5-fluorouracil induced up-regulation of human thymidylate synthase expression in the two ovarian cancer cell lines studied. As these three independent regulatory phenomena occur in a precise order, our work provides a feasible rationale for earlier observed synergistic combinations of 5-FU with other drugs and may suggest novel therapeutic strategies.


Subject(s)
Cell Cycle/drug effects , Fluorouracil/pharmacology , Thymidylate Synthase/metabolism , Blotting, Western , Cell Cycle/genetics , Cell Line, Tumor , Humans , Immunoprecipitation , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Thymidylate Synthase/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
10.
J Biol Chem ; 287(19): 15648-60, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22431731

ABSTRACT

Pyrococcus abyssi NucS is the founding member of a new family of structure-specific DNA endonucleases that interact with the replication clamp proliferating cell nuclear antigen (PCNA). Using a combination of small angle x-ray scattering and surface plasmon resonance analyses, we demonstrate the formation of a stable complex in solution, in which one molecule of the PabNucS homodimer binds to the outside surface of the PabPCNA homotrimer. Using fluorescent labels, PCNA is shown to increase the binding affinity of NucS toward single-strand/double-strand junctions on 5' and 3' flaps, as well as to modulate the cleavage specificity on the branched DNA structures. Our results indicate that the presence of a single major contact between the PabNucS and PabPCNA proteins, together with the complex-induced DNA bending, facilitate conformational flexibility required for specific cleavage at the single-strand/double-strand DNA junction.


Subject(s)
Archaeal Proteins/chemistry , Endonucleases/chemistry , Proliferating Cell Nuclear Antigen/chemistry , Pyrococcus abyssi/enzymology , Algorithms , Amino Acid Sequence , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Binding, Competitive , DNA Replication/genetics , DNA, Archaeal/chemistry , DNA, Archaeal/genetics , DNA, Archaeal/metabolism , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Protein Structure, Tertiary , Pyrococcus abyssi/genetics , Pyrococcus abyssi/metabolism , Scattering, Small Angle , Surface Plasmon Resonance , X-Ray Diffraction
11.
Amino Acids ; 42(2-3): 641-53, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21814787

ABSTRACT

Acquired resistance to cisplatin (cDDP) is a multifactorial process that represents one of the main problems in ovarian cancer therapy. Distamycin A is a minor groove DNA binder whose toxicity has limited its use and prompted the synthesis of derivatives such as NAX001 and NAX002, which have a carbamoyl moiety and different numbers of pyrrolamidine groups. Their interaction with a B-DNA model and with an extended-TATA box model, [Polyd(AT)], was investigated using isothermal titration calorimetry (ITC) to better understand their mechanism of interaction with DNA and therefore better explain their cellular effects. Distamycin A interactions with Dickerson and Poly[d(AT)(6)] oligonucleotides show a different thermodynamic with respect to NAX002. The bulkier distamycin A analogue shows a non optimal binding to DNA due to its additional pyrrolamidine group. Cellular assays performed on cDDP-sensitive and -resistant cells showed that these compounds, distamycin A in particular, affect the expression of folate cycle enzymes even at cellular level. The optimal interaction of distamycin A with DNA may account for the down-regulation of both dihydrofolate reductase (DHFR) and thymidylate synthase (TS) and the up-regulation of spermidine/spermine N1-acetyltransferase (SSAT) caused by this compound. These effects seem differently modulated by the cDDP-resistance phenotype. NAX002 which presents a lower affinity to DNA and slightly affected these enzymes, showed a synergic inhibition profile in combination with cDDP. In addition, their combination with cDDP or polyamine analogues increased cell sensitivity to the drugs suggesting that these interactions may have potential for development in the treatment of ovarian carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Distamycins/pharmacology , Ovarian Neoplasms/pathology , Base Sequence , Cell Line, Tumor , DNA Primers , Drug Resistance, Neoplasm , Drug Synergism , Female , Humans , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
12.
Proc Natl Acad Sci U S A ; 108(34): E542-9, 2011 Aug 23.
Article in English | MEDLINE | ID: mdl-21795601

ABSTRACT

Human thymidylate synthase is a homodimeric enzyme that plays a key role in DNA synthesis and is a target for several clinically important anticancer drugs that bind to its active site. We have designed peptides to specifically target its dimer interface. Here we show through X-ray diffraction, spectroscopic, kinetic, and calorimetric evidence that the peptides do indeed bind at the interface of the dimeric protein and stabilize its di-inactive form. The "LR" peptide binds at a previously unknown binding site and shows a previously undescribed mechanism for the allosteric inhibition of a homodimeric enzyme. It inhibits the intracellular enzyme in ovarian cancer cells and reduces cellular growth at low micromolar concentrations in both cisplatin-sensitive and -resistant cells without causing protein overexpression. This peptide demonstrates the potential of allosteric inhibition of hTS for overcoming platinum drug resistance in ovarian cancer.


Subject(s)
Enzyme Inhibitors/pharmacology , Molecular Targeted Therapy , Ovarian Neoplasms/enzymology , Peptides/metabolism , Peptides/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Cisplatin/therapeutic use , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/therapeutic use , Female , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Peptides/chemistry , Peptides/therapeutic use , Protein Binding/drug effects , Protein Conformation , Protein Multimerization/drug effects , Thermodynamics , Thymidylate Synthase/chemistry , Thymidylate Synthase/metabolism
13.
Biochem Soc Trans ; 39(1): 145-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21265762

ABSTRACT

Branched DNA structures that occur during DNA repair and recombination must be efficiently processed by structure-specific endonucleases in order to avoid cell death. In the present paper, we summarize our screen for new interaction partners for the archaeal replication clamp that led to the functional characterization of a novel endonuclease family, dubbed NucS. Structural analyses of Pyrococcus abyssi NucS revealed an unexpected binding site for ssDNA (single-stranded DNA) that directs, together with the replication clamp, the nuclease activity of this protein towards ssDNA-dsDNA (double-stranded DNA) junctions. Our studies suggest that understanding the detailed architecture and dynamic behaviour of the NucS (nuclease specific for ssDNA)-PCNA (proliferating-cell nuclear antigen) complex with DNA will be crucial for identification of its physiologically relevant activities.


Subject(s)
DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , Endodeoxyribonucleases/metabolism , Nucleic Acid Conformation , Amino Acid Sequence , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , DNA Replication , Endodeoxyribonucleases/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Proliferating Cell Nuclear Antigen/metabolism , Protein Conformation , Pyrococcus abyssi/genetics , Pyrococcus abyssi/metabolism , Ribonucleases/chemistry , Ribonucleases/metabolism , Sequence Alignment
14.
Invest New Drugs ; 29(1): 73-86, 2011 Feb.
Article in English | MEDLINE | ID: mdl-19834646

ABSTRACT

The cellular effects of a novel DNA-intercalating agent, the bipyridyl complex of platinum(II) with diphenyl thiourea, [Pt(bipy)(Ph(2)-tu)(2)]Cl(2), has been analyzed in the cisplatin (cDDP)-sensitive human ovarian carcinoma cell line, 2008, and its -resistant variant, C13* cells, in which the highest accumulation and cytotoxicity was found among six related bipyridyl thiourea complexes. We also show here that this complex causes reactive oxygen species to form and inhibits topoisomerase II activity to a greater extent in the sensitive than in the resistant line. The impairment of this enzyme led to DNA damage, as shown by the comet assay. As a consequence, cell cycle distribution has also been greatly perturbed in both lines. Morphological analysis revealed deep cellular derangement with the presence of cellular masses, together with increased membrane permeability and depolarization of the mitochondrial membrane. Some of these effects, sometimes differentially evident between the two cell lines, might also be related to the decrease of total cell magnesium content caused by this thiourea complex both in sensitive and resistant cells, though the basal content of this ion was higher in the cDDP-resistant line. Altogether these results suggest that this compound exerts its cytotoxicity by mechanisms partly mediated by the resistance phenotype. In particular, cDDP-sensitive cells were affected mostly by impairing topoisomerase II activity and by increasing membrane permeability and the formation of reactive oxygen species; conversely, mitochondrial impairment appeared to play the most important role in the action of complex F in resistant cells.


Subject(s)
2,2'-Dipyridyl/pharmacology , Cisplatin/pharmacology , DNA, Neoplasm/metabolism , Drug Resistance, Neoplasm/drug effects , Intercalating Agents/pharmacology , Organoplatinum Compounds/pharmacology , Ovarian Neoplasms/pathology , 2,2'-Dipyridyl/chemistry , Cell Cycle/drug effects , Cell Fusion , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Topoisomerases, Type II/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Intercalating Agents/chemistry , Intracellular Space/drug effects , Intracellular Space/metabolism , Magnesium/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Organoplatinum Compounds/chemistry , Ovarian Neoplasms/enzymology , Reactive Oxygen Species/metabolism
15.
Bioorg Med Chem ; 18(22): 7773-85, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20951595

ABSTRACT

In the process of drug discovery the lead-identification phase may be critical due to the likely poor safety profile of the candidates, causing the delay or even the abandonment of a certain project. Nowadays, combining molecular modeling and in vivo cellular evaluation can help to identify compounds with an enhanced safety profile. Previously, two quinoxalines have been identified as inhibitors of the folate-dependent proteins belonging to the thymidylate synthase cycle. Unfortunately, cytotoxic activity against a panel of cisplatin(cDDP)-sensitive ovarian carcinoma cell lines and their resistant counterparts was coupled with toxicity to non-tumorigenic Vero cells. Here we describe the application of a ligand-based virtual screening, and several [1,2,4]triazolo[4,3-a]quinoxalines were optimized to improve their ADME-tox profile. The resulting 4-(trifluoromethyl)-1-p-tolyl-[1,2,4]triazolo[4,3-a]quinoxaline (24), which interferes intracellularly with DHFR and TS reducing the protein levels like 5-FU, but without inducing TS ternary complex formation, was 2-times less toxic in vitro than cisplatin and 5-FU.


Subject(s)
Folic Acid Antagonists/chemistry , Folic Acid/metabolism , Ligands , Quinoxalines/chemistry , Triazoles/chemistry , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/toxicity , Folic Acid/chemistry , Folic Acid Antagonists/pharmacokinetics , Folic Acid Antagonists/toxicity , Humans , Quinoxalines/pharmacokinetics , Quinoxalines/toxicity , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/metabolism , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/metabolism , Triazoles/pharmacokinetics , Triazoles/toxicity
16.
Gynecol Oncol ; 117(2): 202-10, 2010 May.
Article in English | MEDLINE | ID: mdl-20031193

ABSTRACT

OBJECTIVE: Polyamines have been shown to play a role in the growth and survival of several solid tumors, including ovarian cancer. Intracellular polyamine depletion by the inhibition of biosynthesis enzymes or by the induction of the catabolic pathway leads to antiproliferative effects in many different tumor cell lines. Recent studies showed that the thymidylate synthase inhibitor 5-fluorouracil (5-FU) affects polyamine metabolism in colon carcinoma cells through the induction of the key catabolic enzyme spermidine/spermine N1-acetyltransferase (SSAT). METHODS: We therefore examined whether combinations of novel folate cycle inhibitors with quinoxaline structure and drugs that specifically target polyamine metabolism, such as diethylderivatives of norspermine (DENSPM) or spermine (BESpm), have synergistic effect in killing cisplatin-sensitive and drug-resistant daughter human ovarian cell lines. RESULTS: Our results showed that simultaneous drug combination or quinoxaline pre-treatment synergistically increased SSAT expression, depleted polyamines, increased reactive oxygen species production, and produced synergistic tumor cell killing in both cell lines. Of note, this combined therapy increased the chemosensitivity of cisplatin-resistant cells and cross-resistant to the polyamine analogues. On the contrary, some pre-treatment regimens of Spm analogues were antagonistic. CONCLUSIONS: These results show that SSAT plays an important role in novel folate cycle inhibitors effects and suggest that their combination with analogues has potential for development as therapy for ovarian carcinoma based on SSAT modulation.


Subject(s)
Acetyltransferases/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cisplatin/pharmacology , Folic Acid Antagonists/pharmacology , Spermine/analogs & derivatives , Acetyltransferases/biosynthesis , Acetyltransferases/deficiency , Cell Growth Processes/drug effects , Cell Line, Tumor , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/enzymology , Cystadenocarcinoma, Serous/pathology , Drug Resistance, Neoplasm , Drug Synergism , Female , Folic Acid Antagonists/administration & dosage , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Quinoxalines/administration & dosage , Reactive Oxygen Species/metabolism , Spermine/administration & dosage , Spermine/metabolism , Spermine/pharmacology
17.
Eur J Pharmacol ; 615(1-3): 17-26, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19446547

ABSTRACT

The cytotoxicity of two novel folate cycle inhibitors with quinoxalinic structure, 3-methyl-7-trifluoromethyl-2(R)-[3,4,5-trimethoxyanilino]-quinoxaline (453R) and 3-piperazinilmethyl-2[4(oxymethyl)-phenoxy]quinoxaline (311S), was tested against a panel of both cisplatin(cDDP)-sensitive and -resistant carcinoma cell lines. Interestingly, the cisplatin-resistant human ovarian line, C13 cells, exhibited collateral sensitivity towards the two compounds when compared to its sensitive parental 2008 cells. In this resistant line, which showed elevated expression of the folate cycle enzymes, thymidylate synthase (TS) and dihydrofolate reductase (DHFR), due to cisplatin-resistance phenotype, collateral sensitivity correlated with the greater reduction of enzyme expression. In addition, TS and DHFR expression of the other resistant lines, the human ovarian carcinoma A2780/CP cells and the human breast cancer MDA/CH cells, were decreased in accordance with the similar sensitivity or the low level of cross-resistance to these compounds in comparison to their respective parental lines. Noteworthy, unlike 5-fluorouracil, both drugs reduced the level of TS without inducing ternary complex formation with the co-substrate and the nucleotide analogue. Median effect analysis of the interactive effects of cisplatin with the two quinoxalines mainly showed additive or synergistic cell killing, depending on schedules of drug combinations. In particular, synergistic effects were more often obtained, even on the resistant cells, when cisplatin was added at the beginning of the treatment. These results indicate that, despite the possibility of other mechanisms being involved, inhibition of TS cycle enzymes plays an important role in the pharmacology of these compounds, which might also represent a useful component in drug treatment protocols against cDDP-resistant cells.


Subject(s)
Cisplatin/pharmacology , Folic Acid Antagonists/pharmacology , Tetrahydrofolate Dehydrogenase/biosynthesis , Thymidylate Synthase/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cystadenocarcinoma, Serous , Drug Resistance, Neoplasm , Female , Fluorouracil/pharmacology , Humans , Ovarian Neoplasms , Quinoxalines/pharmacology , Tetrahydrofolate Dehydrogenase/genetics , Thymidylate Synthase/biosynthesis
18.
J Inorg Biochem ; 102(4): 699-712, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18082268

ABSTRACT

Six bipyridyl complexes of platinum(II) with thiourea, with different substituents on thiourea moiety [Pt(bipy)(R,R'NCSNR'',R''')(2)]Cl(2) (bipy=2,2'-bipyridine: R=R'=R''=R''' =H; R=Me, R'=R''=R'''=H; R=n-Bu, R'=R''=R'''=H; R=Et, R'=H, R''=Et, R'''=H; R=p-tolyl, R'=R''=R'''=H; R=phenyl, R'=H, R''=phenyl, R'''=H), rationally designed to intercalate into DNA, have been tested against a cisplatin (cDDP)-sensitive human ovarian carcinoma cell line (2008) and its -resistant variant (C13( *)). We show here that the anti-proliferative efficacy of these drugs was dependent on molecular structure, since it increased with ancillary ligand bulkiness and hydrophobicity of substituents on thiourea moiety. In particular, the presence of two phenyl groups on thiourea moiety confers an outstanding cytotoxicity. The increasing cell growth inhibition along the series of complexes partially paralleled with drug accumulation, particularly in resistant cells, but not with drug intercalation into DNA since all compounds exerted comparable ethidium bromide displacement ability. The cDDP-resistant phenotype seems, at least in part, to be involved in the action of these compounds, since the level of cross-resistance established for most complexes appeared to be in agreement with the observed impairment of drug accumulation in the resistant subline. These findings indicate that resistance to alkylating agents such as cDDP confers low level of cross-resistance to this class of DNA intercalators, which, however, depending on substituents on thiourea moiety may present remarkable cell growth inhibition even of resistant cells.


Subject(s)
2,2'-Dipyridyl/chemistry , Cell Proliferation/drug effects , Cisplatin/pharmacology , DNA/drug effects , Intercalating Agents/pharmacology , Ovarian Neoplasms/pathology , Platinum Compounds/pharmacology , Thiourea/chemistry , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Humans , Intercalating Agents/chemistry , Magnetic Resonance Spectroscopy , Platinum Compounds/chemistry
19.
Carcinogenesis ; 26(10): 1677-86, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15905201

ABSTRACT

The limited induction of spermidine/spermine N1-acetyltransferase (SSAT) activity has been implicated as an important determinant of the reduced response to the spermine analogue N1,N12-bis(ethyl)spermine (BESpm) by the cisplatin or cis-diamminedichloroplatinum(II) (cDDP)-resistant human ovarian carcinoma cell line (C13*). We checked whether or not under conditions of SSAT overexpression, enzyme induction and cell sensitivity to both, BESpm and cDDP, were restored to levels comparable with those of more responsive cDDP-sensitive 2008 cells. We transiently transfected the SSAT repressed C13* cells with two expression vectors driving human SSAT overexpression by diverse promoters. We then analysed their responses in the absence and in the presence of BESpm. SSAT activity was promptly, but briefly, expressed by transfection with both pOP/SSAT and pCMV-SSAT plasmids. However, only in the presence of BESpm, did SSAT activity reach the highest levels of induction for longer duration, with different time-courses for the two vectors, that paralleled the effect on cell growth. Under these conditions, growth sensitivity to BESpm of the less-responsive C13* cells was 25% reverted to cell growth inhibition displayed by 2008 cells. More interestingly, the sensitivity to cDDP cytotoxicity also increased in parallel to SSAT overexpression. BESpm induction of pCMV-SSAT-transfected cells caused a further 20-30% reduction of cell survival induced by cDDP, almost recovering the sensitivity of 2008 cells. The enhanced effectiveness of cDDP was also confirmed by the comet assay, showing an increase in the number and length of tails of damaged DNA. These findings confirm that SSAT overexpression inhibits cell growth and enhances growth sensitivity to BESpm in C13* cells, showing for the first time that restoring high inducibility of SSAT activity subverts the reduced sensitivity to cDDP of SSAT-deficient cells, making them almost indistinguishable from the responsive parental 2008 cells.


Subject(s)
Acetyltransferases/genetics , Cisplatin/toxicity , Spermine/analogs & derivatives , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cystadenocarcinoma , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Spermine/toxicity , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL