Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 811
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Biochem ; 82: 139-70, 2013.
Article in English | MEDLINE | ID: mdl-23495937

ABSTRACT

DNA topoisomerases are nature's tools for resolving the unique problems of DNA entanglement that occur owing to unwinding and rewinding of the DNA helix during replication, transcription, recombination, repair, and chromatin remodeling. These enzymes perform topological transformations by providing a transient DNA break, formed by a covalent adduct with the enzyme, through which strand passage can occur. The active site tyrosine is responsible for initiating two transesterifications to cleave and then religate the DNA backbone. The cleavage reaction intermediate is exploited by cytotoxic agents, which have important applications as antibiotics and anticancer drugs. The reactions mediated by these enzymes can also be regulated by their binding partners; one example is a DNA helicase capable of modulating the directionality of strand passage, enabling important functions like reannealing denatured DNA and resolving recombination intermediates. In this review, we cover recent advances in mechanistic insights into topoisomerases and their various cellular functions.


Subject(s)
DNA Replication , DNA Topoisomerases, Type II , DNA Topoisomerases, Type I , Antineoplastic Agents/pharmacology , Catalytic Domain , DNA/genetics , DNA/metabolism , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Humans , Protein Structure, Tertiary
2.
Nucleic Acids Res ; 51(17): 9475-9490, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37587714

ABSTRACT

African swine fever virus (ASFV) is highly contagious and can cause lethal disease in pigs. Although it has been extensively studied in the past, no vaccine or other useful treatment against ASFV is available. The genome of ASFV encodes more than 170 proteins, but the structures and functions for the majority of the proteins remain elusive, which hindered our understanding on the life cycle of ASFV and the development of ASFV-specific inhibitors. Here, we report the structural and biochemical studies of the highly conserved C962R protein of ASFV, showing that C962R is a multidomain protein. The N-terminal AEP domain is responsible for the DNA polymerization activity, whereas the DNA unwinding activity is catalyzed by the central SF3 helicase domain. The middle PriCT2 and D5_N domains and the C-terminal Tail domain all contribute to the DNA unwinding activity of C962R. C962R preferentially works on forked DNA, and likely functions in Base-excision repair (BER) or other repair pathway in ASFV. Although it is not essential for the replication of ASFV, C962R can serve as a model and provide mechanistic insight into the replicative primase proteins from many other species, such as nitratiruptor phage NrS-1, vaccinia virus (VACV) and other viruses.


Subject(s)
African Swine Fever Virus , Viral Proteins , Animals , African Swine Fever/virology , African Swine Fever Virus/enzymology , Swine , Viral Proteins/chemistry , Viral Proteins/metabolism , DNA Topoisomerases, Type I/chemistry , DNA Replication
3.
Annu Rev Biochem ; 78: 31-54, 2009.
Article in English | MEDLINE | ID: mdl-19489720

ABSTRACT

I was born in China and would have remained there but for the tumultuous events that led many of my generation to the United States for graduate studies. Norman Davidson introduced me to DNA when I became a postdoctoral fellow in his group at the California Institute of Technology in 1964, and a fortuitous conversation there ignited my interest in DNA ring formation, which later led me to study different topological forms of DNA rings-catenanes, knots, and supercoils. In 1968, a chance observation led me to identify a new enzyme capable of converting one DNA ring form to another, an enzyme now known as a DNA topoisomerase. My interest in DNA rings and DNA topoisomerases continued throughout my years at the University of California, Berkeley, and Harvard. The fascinating ability of the topoisomerases in passing DNA strands or double helices through one another and their importance in cellular processes have kept me and many others excited in their studies.


Subject(s)
Biochemistry/history , DNA Topoisomerases, Type I/chemistry , DNA/chemistry , China , DNA, Superhelical , History, 20th Century , Taiwan , United States
4.
Chemistry ; 30(39): e202401400, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38736421

ABSTRACT

Coumestan represents a biologically relevant structural motif distributed in a number of natural products, and the rapid construction of related derivatives as well as the characterization of targets would accelerate lead compound discovery in medicinal chemistry. In this work, a general and scalable approach to 8,9-dihydroxycoumestans via two-electrode constant current electrolysis was developed. The application of a two-phase (aqueous/organic) system plays a crucial role for success, protecting the sensitive o-benzoquinone intermediates from over-oxidation. Based on the structurally diverse products, a primary SAR study on coumestan scaffold was completed, and compound 3 r exhibited potent antiproliferative activities and a robust topoisomerase I (Top1) inhibitory activity. Further mechanism studies demonstrates that compound 3 r was a novel Top1 poison, which might open an avenue for the development of Top1-targeted antitumor agent.


Subject(s)
Antineoplastic Agents , Coumarins , DNA Topoisomerases, Type I , Topoisomerase I Inhibitors , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/chemical synthesis , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type I/chemistry , Humans , Structure-Activity Relationship , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Coumarins/chemistry , Coumarins/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Oxidation-Reduction , Umbelliferones/chemistry , Umbelliferones/pharmacology , Drug Screening Assays, Antitumor
5.
J Chem Inf Model ; 64(15): 6062-6071, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39024468

ABSTRACT

Human topoisomerase III beta (hTOP3B) is the only topoisomerase in the human cell that can act on both DNA and RNA substrates. Recent findings have emphasized the physiological importance of hTOP3B and consolidated it as a valuable drug target for antiviral and anticancer therapeutics. Although type IA topoisomerases of different organisms have been studied over the years, the step-by-step interaction of hTOP3B and nucleic acid substrates is still not well understood. Due to the lack of hTOP3B-RNA structures as well as DNA/RNA covalent complexes, computational investigations have been limited. In our study, we utilized molecular dynamics (MD) simulations to study the interactions between hTOP3B and nucleic acids to get a closer look into the residues that play a role in binding DNA or RNA and facilitate catalysis, along with the differences and similarities when hTOP3B interacts with DNA compared to RNA. For this, we generated multiple models of hTOP3B complexed with DNA and RNA sequences using the hTOP3B crystal structure and 8-mer single-stranded DNA and RNA sequences. These models include both covalent and noncovalent complexes, which are then subjected to MD simulations and analyzed. Our findings highlight the complexes' stability, sequence preference, and interactions of the binding pocket residues with different nucleotides. Our work demonstrates that hTOP3B forms stable complexes with both DNA and RNA and provides a better understanding of the enzyme's interaction with different nucleic acid substrate sequences.


Subject(s)
DNA Topoisomerases, Type I , DNA , RNA , Humans , DNA/metabolism , DNA/chemistry , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type I/chemistry , Molecular Dynamics Simulation , Nucleic Acid Conformation , Protein Binding , RNA/metabolism , RNA/chemistry
6.
Bioorg Chem ; 147: 107412, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38696845

ABSTRACT

The development of novel topoisomerase I (TOP1) inhibitors is crucial for overcoming the drawbacks and limitations of current TOP1 poisons. Here, we identified two potential TOP1 inhibitors, namely, FTY720 (a sphingosine 1-phosphate antagonist) and COH29 (a ribonucleotide reductase inhibitor), through experimental screening of known active compounds. Biological experiments verified that FTY720 and COH29 were nonintercalative TOP1 catalytic inhibitors that did not induce the formation of DNA-TOP1 covalent complexes. Molecular docking revealed that FTY720 and COH29 interacted favorably with TOP1. Molecular dynamics simulations revealed that FTY720 and COH29 could affect the catalytic domain of TOP1, thus resulting in altered DNA-binding cavity size. The alanine scanning and interaction entropy identified Arg536 as a hotspot residue. In addition, the bioinformatics analysis predicted that FTY720 and COH29 could be effective in treating malignant breast tumors. Biological experiments verified their antitumor activities using MCF-7 breast cancer cells. Their combinatory effects with TOP1 poisons were also investigated. Further, FTY720 and COH29 were found to cause less DNA damage compared with TOP1 poisons. The findings provide reliable lead compounds for the development of novel TOP1 catalytic inhibitors and offer new insights into the potential clinical applications of FTY720 and COH29 in targeting TOP1.


Subject(s)
Antineoplastic Agents , DNA Topoisomerases, Type I , Fingolimod Hydrochloride , Molecular Docking Simulation , Topoisomerase I Inhibitors , Humans , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/chemical synthesis , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type I/chemistry , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/chemical synthesis , Molecular Structure , Structure-Activity Relationship , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Cell Proliferation/drug effects , Molecular Dynamics Simulation , MCF-7 Cells
7.
Int J Mol Sci ; 25(19)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39409108

ABSTRACT

Cancer poses a substantial global health challenge, driving the need for innovative therapeutic solutions that offer improved effectiveness and fewer side effects. Topoisomerase I (Topo I) has emerged as a validated molecular target in the pursuit of developing anticancer drugs due to its critical role in DNA replication and transcription. (+)-Pancratistatin (PST), a naturally occurring compound found in various Amaryllidaceae plants, exhibits promising anticancer properties by inhibiting Topo I activity. However, its clinical utility is hindered by issues related to limited chemical availability and aqueous solubility. To address these challenges, molecular modelling techniques, including virtual screening, molecular docking, molecular mechanics with generalised born and surface area solvation (MM-GBSA) calculations, and molecular dynamics simulations were utilised to evaluate the binding interactions and energetics of PST analogues with Topo I, comparing them with the well-known Topo I inhibitor, Camptothecin. Among the compounds screened for this study, nitrogenated analogues emerged as the most encouraging drug candidates, exhibiting improved binding affinities, favourable interactions with the active site of Topo I, and stability of the protein-ligand complex. Structural analysis pinpointed key molecular determinants responsible for the heightened potency of nitrogenated analogues, shedding light on essential structural modifications for increased activity. Moreover, in silico absorption, distribution, metabolism, excretion, and toxicity (ADMET) predictions highlighted favourable drug-like properties and reduced toxicity profiles for the most prominent nitrogenated analogues, further supporting their potential as effective anticancer agents. In summary, this screening study underscores the significance of nitrogenation in augmenting the anticancer efficacy of PST analogues targeting Topo I. The identified lead compounds exhibit significant potential for subsequent experimental validation and optimisation, thus facilitating the development of novel and efficacious anticancer therapeutics with enhanced pharmacological profiles.


Subject(s)
Amaryllidaceae Alkaloids , Antineoplastic Agents , DNA Topoisomerases, Type I , Isoquinolines , Molecular Docking Simulation , Molecular Dynamics Simulation , Topoisomerase I Inhibitors , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/chemistry , Amaryllidaceae Alkaloids/pharmacology , Amaryllidaceae Alkaloids/chemistry , Humans , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type I/chemistry , Isoquinolines/chemistry , Isoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Protein Binding
8.
J Mol Evol ; 91(2): 192-203, 2023 04.
Article in English | MEDLINE | ID: mdl-36651963

ABSTRACT

Type IB topoisomerases relax the torsional stress associated with DNA metabolism in the nucleus and mitochondria and constitute important molecular targets of anticancer drugs. Vertebrates stand out among eukaryotes by having two Type IB topoisomerases acting specifically in the nucleus (TOP1) and mitochondria (TOP1MT). Despite their major importance, the origin and evolution of these paralogues remain unknown. Here, we examine the molecular evolutionary processes acting on both TOP1 and TOP1MT in Chordata, taking advantage of the increasing number of available genome sequences. We found that both TOP1 and TOP1MT evolved under strong purifying selection, as expected considering their essential biological functions. Critical active sites, including those associated with resistance to anticancer agents, were found particularly conserved. However, TOP1MT presented a higher rate of molecular evolution than TOP1, possibly related with its specialized activity on the mitochondrial genome and a less critical role in cells. We could place the duplication event that originated the TOP1 and TOP1MT paralogues early in the radiation of vertebrates, most likely associated with the first round of vertebrate tetraploidization (1R). Moreover, our data suggest that cyclostomes present a specialized mitochondrial Type IB topoisomerase. Interestingly, we identified two missense mutations replacing amino acids in the Linker region of TOP1MT in Neanderthals, which appears as a rare event when comparing the genome of both species. In conclusion, TOP1 and TOP1MT differ in their rates of evolution, and their evolutionary histories allowed us to better understand the evolution of chordates.


Subject(s)
Chordata , DNA, Mitochondrial , Animals , DNA, Mitochondrial/genetics , Chordata/genetics , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type I/metabolism , Mitochondria/genetics , Cell Nucleus/genetics
9.
Org Biomol Chem ; 21(21): 4518-4528, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37199703

ABSTRACT

The human topoisomerase IB (hTopoIB) enzyme is a monomeric protein that relaxes the supercoils on double-stranded DNA by forming a covalent DNA/hTopoIB complex by introducing a nick on the DNA strand. Inhibition of hTopoIB results in cell death, which makes this protein a strong target for the treatment of various cancer types, including small-cell lung cancers and ovarian cancers. Camptothecin (CPT) and indenoisoquinoline (IQN) classes of compounds inhibit the hTopoIB activity by intercalating to nicked DNA pairs; however, these inhibitors show different preferences towards DNA bases when bound to the DNA/hTopoIB complex. Here, we investigated the affinities of CPT and one IQN derivative towards different DNA base pairs. The two inhibitors showed different stacking behaviors in the intercalation site and interaction pattern with binding pocket residues, indicating that they have different inhibition mechanisms in the binding pocket that affects the base-pair selectivity. The results obtained from this study are expected to guide researchers in designing gene-specific and more potent compounds to fight cancer through hTopoIB poisoning.


Subject(s)
Neoplasms , Topoisomerase I Inhibitors , Humans , Topoisomerase I Inhibitors/pharmacology , DNA/chemistry , DNA Topoisomerases, Type I/chemistry , Base Pairing , Camptothecin/chemistry , Enzyme Inhibitors/pharmacology
10.
Nucleic Acids Res ; 49(10): 5470-5492, 2021 06 04.
Article in English | MEDLINE | ID: mdl-33963870

ABSTRACT

Topoisomerases are essential enzymes that regulate DNA topology. Type 1A family topoisomerases are found in nearly all living organisms and are unique in that they require single-stranded (ss)DNA for activity. These enzymes are vital for maintaining supercoiling homeostasis and resolving DNA entanglements generated during DNA replication and repair. While the catalytic cycle of Type 1A topoisomerases has been long-known to involve an enzyme-bridged ssDNA gate that allows strand passage, a deeper mechanistic understanding of these enzymes has only recently begun to emerge. This knowledge has been greatly enhanced through the combination of biochemical studies and increasingly sophisticated single-molecule assays based on magnetic tweezers, optical tweezers, atomic force microscopy and Förster resonance energy transfer. In this review, we discuss how single-molecule assays have advanced our understanding of the gate opening dynamics and strand-passage mechanisms of Type 1A topoisomerases, as well as the interplay of Type 1A topoisomerases with partner proteins, such as RecQ-family helicases. We also highlight how these assays have shed new light on the likely functional roles of Type 1A topoisomerases in vivo and discuss recent developments in single-molecule technologies that could be applied to further enhance our understanding of these essential enzymes.


Subject(s)
DNA Topoisomerases, Type I , DNA , DNA/chemistry , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type I/physiology , Humans , Molecular Structure , RecQ Helicases/chemistry
11.
Nucleic Acids Res ; 48(8): 4448-4462, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32232337

ABSTRACT

Type IA topoisomerases interact with G-strand and T-strand ssDNA to regulate DNA topology. However, simultaneous binding of two ssDNA segments to a type IA topoisomerase has not been observed previously. We report here the crystal structure of a type IA topoisomerase with ssDNA segments bound in opposite polarity to the N- and C-terminal domains. Titration of small ssDNA oligonucleotides to Mycobacterium smegmatis topoisomerase I with progressive C-terminal deletions showed that the C-terminal region has higher affinity for ssDNA than the N-terminal active site. This allows the C-terminal domains to capture one strand of underwound negatively supercoiled DNA substrate first and position the N-terminal domains to bind and cleave the opposite strand in the relaxation reaction. Efficiency of negative supercoiling relaxation increases with the number of domains that bind ssDNA primarily with conserved aromatic residues and possibly with assistance from polar/basic residues. A comparison of bacterial topoisomerase I structures showed that a conserved transesterification unit (N-terminal toroid structure) for cutting and rejoining of a ssDNA strand can be combined with two different types of C-terminal ssDNA binding domains to form diverse bacterial topoisomerase I enzymes that are highly efficient in their physiological role of preventing excess negative supercoiling in the genome.


Subject(s)
DNA Topoisomerases, Type I/chemistry , DNA, Single-Stranded/metabolism , Mycobacterium smegmatis/enzymology , Crystallography, X-Ray , DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , Models, Molecular , Protein Domains , Sequence Deletion
12.
Nucleic Acids Res ; 48(2): 761-769, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31777935

ABSTRACT

Identifying the molecular mechanisms that give rise to genetic variation is essential for the understanding of evolutionary processes. Previously, we have used adaptive laboratory evolution to enable biomass synthesis from CO2 in Escherichia coli. Genetic analysis of adapted clones from two independently evolving populations revealed distinct enrichment for insertion and deletion mutational events. Here, we follow these observations to show that mutations in the gene encoding for DNA topoisomerase I (topA) give rise to mutator phenotypes with characteristic mutational spectra. Using genetic assays and mutation accumulation lines, we find that point mutations in topA increase the rate of sequence deletion and duplication events. Interestingly, we observe that a single residue substitution (R168C) results in a high rate of head-to-tail (tandem) short sequence duplications, which are independent of existing sequence repeats. Finally, we show that the unique mutation spectrum of topA mutants enhances the emergence of antibiotic resistance in comparison to mismatch-repair (mutS) mutators, and leads to new resistance genotypes. Our findings highlight a potential link between the catalytic activity of topoisomerases and the fundamental question regarding the emergence of de novo tandem repeats, which are known modulators of bacterial evolution.


Subject(s)
Carbon Dioxide/metabolism , DNA Topoisomerases, Type I/genetics , Escherichia coli Proteins/genetics , Escherichia coli/genetics , MutS DNA Mismatch-Binding Protein/genetics , Biomass , Carbon Dioxide/chemistry , DNA Topoisomerases, Type I/chemistry , Drug Resistance, Bacterial/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Evolution, Molecular , Gene Duplication/genetics , Genotype , MutS DNA Mismatch-Binding Protein/chemistry , Mutation , Point Mutation/genetics
13.
J Biol Chem ; 295(12): 3990-4000, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32029477

ABSTRACT

DNA double-stranded breaks (DSBs) are strongly associated with active transcription, and promoter-proximal pausing of RNA polymerase II (Pol II) is a critical step in transcriptional regulation. Mapping the distribution of DSBs along actively expressed genes and identifying the location of DSBs relative to pausing sites can provide mechanistic insights into transcriptional regulation. Using genome-wide DNA break mapping/sequencing techniques at single-nucleotide resolution in human cells, we found that DSBs are preferentially located around transcription start sites of highly transcribed and paused genes and that Pol II promoter-proximal pausing sites are enriched in DSBs. We observed that DSB frequency at pausing sites increases as the strength of pausing increases, regardless of whether the pausing sites are near or far from annotated transcription start sites. Inhibition of topoisomerase I and II by camptothecin and etoposide treatment, respectively, increased DSBs at the pausing sites as the concentrations of drugs increased, demonstrating the involvement of topoisomerases in DSB generation at the pausing sites. DNA breaks generated by topoisomerases are short-lived because of the religation activity of these enzymes, which these drugs inhibit; therefore, the observation of increased DSBs with increasing drug doses at pausing sites indicated active recruitment of topoisomerases to these sites. Furthermore, the enrichment and locations of DSBs at pausing sites were shared among different cell types, suggesting that Pol II promoter-proximal pausing is a common regulatory mechanism. Our findings support a model in which topoisomerases participate in Pol II promoter-proximal pausing and indicated that DSBs at pausing sites contribute to transcriptional activation.


Subject(s)
DNA Breaks, Double-Stranded , RNA Polymerase II/metabolism , Camptothecin/metabolism , Camptothecin/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/metabolism , Etoposide/metabolism , Etoposide/pharmacology , HeLa Cells , Humans , Transcription Initiation Site , Transcriptional Activation/drug effects
14.
Biochem Biophys Res Commun ; 569: 193-198, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34256188

ABSTRACT

Visceral leishmaniasis (VL) is a fatal infectious disease caused by viscerotropic parasitic species of Leishmania. Current treatment options are often ineffective and toxic, and more importantly, there are no clinically validated drug targets available to develop next generation therapeutics against VL. Topoisomerase IB (TopIB) is an essential enzyme for Leishmania survival. The enzyme is organized as a bi-subunit that is distinct from the monomeric topoisomerase I of human. Based on this unique feature, we synthesized peptides composed of partial amino acid sequences of small subunit of Leishmania donovani (Ld) TopIB to confirm a decrease in catalytic activity by interfering the interaction between the two subunits. One of the synthetic peptides, covering essential amino acids for catalytic activity of LdTopIB, interrupted the enzymatic activity. Next, we examined 151 compounds selected from virtual screening in a functional assay and identified three LRL-TP compounds with a significant decrease in LdTopIB activity (IC50 of LRL-TP-85: 1.3 µM; LRL-TP-94: 2.9 µM; and LRL-TP-101: 35.3 µM) and no effects on Homo sapiens (Hs) TopIB activity. Based on molecular docking, the protonated tertiary amine of inhibitors formed key interactions with S415 of the large subunit. The EC50 values of LRL-TP-85, LRL-TP-94, and LRL-TP-101 were respectively 4.9, 1.4, and 27.8 µM in extracellular promastigote assay and 34.0, 53.7, and 11.4 µM in intracellular amastigote assay. Overall, we validated the protein-protein interaction site of LdTopIB as a potential drug target and identified small molecule inhibitors with anti-leishmanial activity.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Leishmania donovani/enzymology , Protein Interaction Maps/drug effects , Protozoan Proteins/metabolism , Topoisomerase I Inhibitors/pharmacology , Animals , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacology , Cells, Cultured , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type I/genetics , Humans , Leishmania donovani/drug effects , Leishmania donovani/genetics , Leishmaniasis, Visceral/parasitology , Leishmaniasis, Visceral/prevention & control , Mice , Models, Molecular , Molecular Structure , Nucleic Acid Conformation , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Binding/drug effects , Protein Domains , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , THP-1 Cells , Topoisomerase I Inhibitors/chemistry
15.
Exp Mol Pathol ; 123: 104687, 2021 12.
Article in English | MEDLINE | ID: mdl-34592199

ABSTRACT

BACKGROUND: The JC polyomavirus has been blamed to contribute in colorectal cancer (CRC), however, the topic is still controversial. Varying detection rate of JCPyV genome has been reported mainly due to technical reasons. Here, we provide summative data on the topic, with emphasize on technical issues. METHODS: Formalin-fixed paraffin-embedded tissue samples from 50 patients with CRC, consisting of tumoral and non-cancerous marginal tissue (totally 100 samples) were included in the study. After DNA extraction, specific JCPyV T-Ag sequences were targeted using Real-time PCR. To unwind the supercoiled JCPyV genome, pretreatment with topoisomerase I, was applied. Immunohistochemical (IHC) staining was performed using an anti-T-Ag monoclonal antibody. RESULTS: In the first attempts, no samples were found to be positive in Real-time PCR assays. However, JCPyV sequences were found in 60% of CRC tissues and 38% of non-cancerous colorectal mucosa after application of pre-treatment step with topoisomerase I enzyme (P = 0.028). T-Ag protein was found in the nuclear compartment of the stained cells in IHC assays. CONCLUSIONS: The presence of JCPyV in CRC tissues, as well as T-Ag localization in the nucleolus, where its oncogenic effect takes place, may provide supporting evidence for JCPyV involvement in CRC development. The study highlights the importance of using topoisomerase I to enhance JCPyV genome detection. Also, colorectal tissue is one of the permissive human tissue for JC resistance after preliminary infection.


Subject(s)
Colorectal Neoplasms/virology , DNA Topoisomerases, Type I/pharmacology , Genome, Viral/genetics , JC Virus/isolation & purification , Cell Nucleolus/genetics , Cell Nucleolus/virology , Colorectal Neoplasms/complications , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Topoisomerases, Type I/chemistry , Female , Humans , JC Virus/genetics , JC Virus/pathogenicity , Male , Middle Aged , Polyomavirus Infections/complications , Polyomavirus Infections/genetics , Polyomavirus Infections/pathology , Polyomavirus Infections/virology , Virus Replication/genetics
16.
Bioorg Chem ; 107: 104631, 2021 02.
Article in English | MEDLINE | ID: mdl-33476866

ABSTRACT

In this account we present NMR based results of the interaction of 7-ethyl-9-hydroxymethyl-10-hydroxycamptothecin (1), a derivative of SN38, with a model nicked DNA decamer mimicking the wild type DNA target of Topoisomerase I inhibitors from the camptothecin family. The title compound 1 can be considered a main metabolite of phase I in the metabolic pathway of camptothecin derivatives bearing the alkylamino substituent. Therefore, its pharmacodynamic properties are of interest. It was established by DOSY (Diffusion Ordered Spectroscopy) that compound 1 forms a fairly stable molecular complex with a model nicked DNA decamer with affinity constant Ka 3.02 mM-1. The analysis of NOESY experiments revealed intermolecular cross peaks and mutual induced shifts on both interacting components allowing the conclusion that guest molecule 1 is stacking the nitrogen bases inside the nick. MD (Molecular Dynamics) analysis of four possible inclusions of 1 inside the nick allows establishing the detailed geometry of a complex. Two conformations are suggested as the ones best representing the results of molecular modeling reconciled with experimental NOESY results. The aromatic core of both structures is stacking the nitrogen bases in a nick facing the unbroken strand with ring A. The protons in ring E interact with ribose protons of edge bases of a nick. In conclusion, it can be asserted that SN38 derivative 1 can effectively bind the molecular target of Topo I enzyme and play a role as a Topo I inhibitor.


Subject(s)
Camptothecin/chemistry , DNA Topoisomerases, Type I/chemistry , DNA/chemistry , Topoisomerase I Inhibitors/chemistry , Binding Sites , Camptothecin/metabolism , Cell Line, Tumor , Cell Survival/drug effects , DNA/metabolism , DNA Topoisomerases, Type I/metabolism , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , Topoisomerase I Inhibitors/metabolism
17.
Bioorg Chem ; 107: 104637, 2021 02.
Article in English | MEDLINE | ID: mdl-33454505

ABSTRACT

Cancer has become an important public problem in worldwide since cancer incidence and mortality are growing rapidly. In this study, water soluble and non-aggregated silicon (IV) phthalocyanines and naphthalocyanines containing (3,5-bis{3-[3-(diethylamino)phenoxy]propoxy}phenyl)methoxy groups have been synthesized and characterized to investigate their anticancer potential. Their DNA binding/nuclease, topoisomerases inhibition were investigated using UV-Vis absorption, thermal denaturation and agarose gel electrophoresis. The in vitro cytotoxic properties of the compounds on human lung (A549), breast (BT-20), liver (SNU-398), prostate (DU-145), melanoma (SK-Mel 128) carcinoma and human fibroblast (HFC) normal cell lines were evaluated by using MTT assay. In order to determine the mechanism of cancer cell growth suppression, cell cycle analysis was carried out using flow cytometer on A549 cell line. The Kb values of SiPc1a and SiNc2a were 6.85 ± (0.35) × 106 and 1.72 ± (0.16) × 104 M-1 and Tm values of ct-DNA were calculated as 82.02 °C and 78.07 °C, respectively in the presence of both compounds. The ΔTm values of SiPc1a and SiNc2a were calculated as 6.45 and 2.50 °C, respectively. The nuclease effects of SiPc1a and SiNc2a with supercoiled plasmid pBR322 DNA demonstrated that both compounds did not trigger any DNA nuclease effects at the lowest concentrations without irradiation whereas both compounds in the presence of activating agent (H2O2) showed significant plasmid DNA nuclease actions under irradiation (22.5 J/cm2). SiPc1a and SiNc2a inhibited to topoisomerase I on increasing concentrations whilst they had lower inhibition action toward topoisomerase II that of topoisomerase I. The in vitro cytotoxicity studies displayed that SiPc1a had the highest cytotoxic effects among the tested compounds against A549, SNU-398, SK-MEL128, DU-145, BT-20 and HFC cell lines with CC50 values ranged from 0.49 to 2.99 µM. Furthermore, SiPc1a inhibited cell proliferation by cell cycle arrest in G0/G1 phase. All of these results suggested that SiPc1a is a promising candidate as an anticancer agent.


Subject(s)
Antineoplastic Agents/chemical synthesis , Drug Design , Indoles/chemistry , Organosilicon Compounds/chemistry , Topoisomerase I Inhibitors/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/chemistry , DNA/metabolism , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/metabolism , Deoxyribonucleases/antagonists & inhibitors , Deoxyribonucleases/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Hydrogen Peroxide/pharmacology , Indoles/metabolism , Indoles/pharmacology , Organosilicon Compounds/metabolism , Organosilicon Compounds/pharmacology , Solubility , Topoisomerase I Inhibitors/metabolism , Topoisomerase I Inhibitors/pharmacology , Water/chemistry
18.
Bioorg Chem ; 106: 104422, 2021 01.
Article in English | MEDLINE | ID: mdl-33248713

ABSTRACT

A novel series of urea-linked ciprofloxacin (CP)-chalcone hybrids 3a-j were synthesized and screened by NCI-60 cancer cell lines as potential cytotoxic agents. Interestingly, compounds 3c and 3j showed remarkable antiproliferative activities against both colon HCT-116 and leukemia SR cancer cells compared to camptothecin, topotecan and staurosporine with IC50 = 2.53, 2.01, 17.36, 12.23 and 3.1 µM for HCT-116 cells, respectively and IC50 = 0.73, 0.64, 3.32, 13.72 and 1.17 µM for leukemia SR cells, respectively. Also, compounds 3c and 3j exhibited inhibitory activities against Topoisomerase (Topo) I with % inhibition = 51.19% and 56.72%, respectively, compared to camptothecin (% inhibition = 60.05%) and Topo IIß with % inhibition = 60.81% and 60.06%, respectively, compared to topotecan (% inhibition = 71.09%). Furthermore, compound 3j arrested the cell cycle of leukemia SR cells at G2/M phase. It induced apoptosis both intrinsically and extrinsically via activation of proteolytic caspases cascade (caspases-3, -8, and -9), release of cytochrome C from mitochondria, upregulation of proapoptotic Bax and down-regulation of Bcl-2 protein level. Thus, the new ciprofloxacin derivative 3j could be considered as a potential lead for further optimization of antitumor agent against leukemia and colorectal carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Chalcones/pharmacology , Ciprofloxacin/analogs & derivatives , Ciprofloxacin/pharmacology , Topoisomerase Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Caspases/metabolism , Catalytic Domain , Cell Line, Tumor , Chalcones/chemical synthesis , Chalcones/metabolism , Ciprofloxacin/chemical synthesis , Ciprofloxacin/metabolism , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/metabolism , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Molecular Docking Simulation , Molecular Structure , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/metabolism , Phenylurea Compounds/pharmacology , Poly-ADP-Ribose Binding Proteins/chemistry , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Binding , Proto-Oncogene Proteins c-bcl-2/metabolism , Structure-Activity Relationship , Topoisomerase Inhibitors/chemical synthesis , Topoisomerase Inhibitors/metabolism , bcl-2-Associated X Protein/metabolism
19.
Nucleic Acids Res ; 47(11): 5511-5521, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31073604

ABSTRACT

Inspired by DNA mimic proteins, we have introduced aromatic foldamers bearing phosphonate groups as synthetic mimics of the charge surface of B-DNA and competitive inhibitors of some therapeutically relevant DNA-binding enzymes: the human DNA Topoisomerase 1 (Top1) and the human HIV-1 integrase (HIV-1 IN). We now report on variants of these anionic foldamers bearing carboxylates instead of phosphonates. Several new monomers have been synthesized with protecting groups suitable for solid phase synthesis (SPS). Six hexadecaamides have been prepared using SPS. Proof of their resemblance to B-DNA was brought by the first crystal structure of one of these DNA-mimic foldamers in its polyanionic form. While some of the foldamers were found to be as active as, or even more active than, the original phosphonate oligomers, others had no activity at all or could even stimulate enzyme activity in vitro. Some foldamers were found to have differential inhibitory effects on the two enzymes. These results demonstrate a strong dependence of inhibitory activity on foldamer structure and charge distribution. They open broad avenues for the development of new classes of derivatives that could inhibit the interaction of specific proteins with their DNA target thereby influencing the cellular pathways in which they are involved.


Subject(s)
Amides/chemistry , DNA Topoisomerases, Type I/chemistry , DNA, B-Form/chemistry , HIV Integrase Inhibitors/chemistry , HIV Integrase/chemistry , Biocatalysis , Biomimetic Materials/chemistry , Crystallography, X-Ray , HIV Integrase Inhibitors/chemical synthesis , HIV-1/enzymology , Humans , Molecular Structure , Protein Conformation , Solid-Phase Synthesis Techniques
20.
Int J Mol Sci ; 22(14)2021 Jul 12.
Article in English | MEDLINE | ID: mdl-34299090

ABSTRACT

The compounds 7-ethyl-9-(N-methylamino)methyl-10-hydroxycamptothecin (2) and 7-ethyl-9-(N-morpholino)methyl-10-hydroxycamptothecin (3) are potential topoisomerase I poisons. Moreover, they were shown to have favorable anti-neoplastic effects on several tumor cell lines. Due to these properties, the compounds are being considered for advancement to the preclinical development stage. To gain better insights into the molecular mechanism with the biological target, here, we conducted an investigation into their interactions with model nicked DNA (1) using different techniques. In this work, we observed the complexity of the mechanism of action of the compounds 2 and 3, in addition to their decomposition products: compound 4 and SN38. Using DOSY experiments, evidence of the formation of strongly bonded molecular complexes of SN38 derivatives with DNA duplexes was provided. The molecular modeling based on cross-peaks from the NOESY spectrum also allowed us to assign the geometry of a molecular complex of DNA with compound 2. Confirmation of the alkylation reaction of both compounds was obtained using MALDI-MS. Additionally, in the case of 3, alkylation was confirmed in the recording of cross-peaks in the 1H/13C HSQC spectrum of 13C-enriched compound 3. In this work, we showed that the studied compounds-parent compounds 2 and 3, and their potential metabolite 4 and SN38-interact inside the nick of 1, either forming the molecular complex or alkylating the DNA nitrogen bases. In order to confirm the influence of the studied compounds on the topoisomerase I relaxation activity of supercoiled DNA, the test was performed based upon the measurement of the fluorescence of DNA stain which can differentiate between supercoiled and relaxed DNA. The presented results confirmed that studied SN38 derivatives effectively block DNA relaxation mediated by Topo I, which means that they stop the machinery of Topo I activity.


Subject(s)
Camptothecin/analogs & derivatives , Camptothecin/metabolism , DNA Breaks, Single-Stranded , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type I/metabolism , DNA, Superhelical , Topoisomerase II Inhibitors/pharmacology , Alkylation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL