Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 43(5): 114214, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38761375

RESUMO

TDP1 removes transcription-blocking topoisomerase I cleavage complexes (TOP1ccs), and its inactivating H493R mutation causes the neurodegenerative syndrome SCAN1. However, the molecular mechanism underlying the SCAN1 phenotype is unclear. Here, we generate human SCAN1 cell models using CRISPR-Cas9 and show that they accumulate TOP1ccs along with changes in gene expression and genomic distribution of R-loops. SCAN1 cells also accumulate transcriptional DNA double-strand breaks (DSBs) specifically in the G1 cell population due to increased DSB formation and lack of repair, both resulting from abortive removal of transcription-blocking TOP1ccs. Deficient TDP1 activity causes increased DSB production, and the presence of mutated TDP1 protein hampers DSB repair by a TDP2-dependent backup pathway. This study provides powerful models to study TDP1 functions under physiological and pathological conditions and unravels that a gain of function of the mutated TDP1 protein, which prevents DSB repair, rather than a loss of TDP1 activity itself, could contribute to SCAN1 pathogenesis.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Mutação , Doenças Neurodegenerativas , Diester Fosfórico Hidrolases , Humanos , Diester Fosfórico Hidrolases/metabolismo , Diester Fosfórico Hidrolases/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Mutação/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , DNA Topoisomerases Tipo I/metabolismo , DNA Topoisomerases Tipo I/genética , Transcrição Gênica , Estruturas R-Loop , Sistemas CRISPR-Cas/genética
2.
Sci Adv ; 10(21): eadm8196, 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38787953

RESUMO

DNA topoisomerase I can contribute to cancer genome instability. During catalytic activity, topoisomerase I forms a transient intermediate, topoisomerase I-DNA cleavage complex (Top1cc) to allow strand rotation and duplex relaxation, which can lead to elevated levels of DNA-RNA hybrids and micronuclei. To comprehend the underlying mechanisms, we have integrated genomic data of Top1cc-triggered hybrids and DNA double-strand breaks (DSBs) shortly after Top1cc induction, revealing that Top1ccs increase hybrid levels with different mechanisms. DSBs are at highly transcribed genes in early replicating initiation zones and overlap with hybrids downstream of accumulated RNA polymerase II (RNAPII) at gene 5'-ends. A transcription factor IIS mutant impairing transcription elongation further increased RNAPII accumulation likely due to backtracking. Moreover, Top1ccs can trigger micronuclei when occurring during late G1 or early/mid S, but not during late S. As micronuclei and transcription-replication conflicts are attenuated by transcription factor IIS, our results support a role of RNAPII arrest in Top1cc-induced transcription-replication conflicts leading to DSBs and micronuclei.


Assuntos
Quebras de DNA de Cadeia Dupla , Replicação do DNA , DNA Topoisomerases Tipo I , Instabilidade Genômica , Estruturas R-Loop , RNA Polimerase II , Humanos , DNA Topoisomerases Tipo I/metabolismo , DNA Topoisomerases Tipo I/genética , RNA Polimerase II/metabolismo , RNA Polimerase II/genética , Transcrição Gênica
3.
Methods Enzymol ; 695: 103-118, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38521582

RESUMO

A large variety of non-B secondary structures can be formed between DNA and RNA. In this chapter, we focus on G-quadruplexes (G4) and R-loops, which can have a close structural interplay. In recent years, increasing evidence pointed to the fact that they can strongly influence each other in vivo, both having physiological and pathological roles in normal and cancer cells. Here, we detail specific and accurate methods for purification of BG4 and S9.6 antibodies, and their subsequent use in immunofluorescence microscopy, enabling single-cell analysis of extent and localization of G4s and R-loops.


Assuntos
Quadruplex G , Estruturas R-Loop , DNA/química , RNA/química , Microscopia de Fluorescência
4.
Mol Cancer ; 21(1): 180, 2022 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-36114513

RESUMO

G-quadruplex (G4) binders have been investigated to discover new anticancer drugs worldwide in past decades. As these ligands are generally not highly cytotoxic, the discovery rational was mainly based on increasing the cell-killing potency. Nevertheless, no G4 binder has been shown yet to be effective in cancer patients. Here, G4 binder activity at low dosages will be discussed as a critical feature to discover ligands with therapeutic effects in cancer patients. Specific effects of G4 binders al low doses have been reported to occur in cancer and normal cells. Among them, genome instability and the stimulation of cytoplasmic processes related to autophagy and innate immune response open to the use of G4 binders as immune-stimulating agents. Thus, we propose a new rational of drug discovery, which is not based on cytotoxic potency but rather on immune gene activation at non-cytotoxic dosage.


Assuntos
Antineoplásicos , Quadruplex G , Neoplasias , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Instabilidade Genômica , Humanos , Ligantes , Neoplasias/tratamento farmacológico , Neoplasias/genética
5.
J Med Chem ; 65(18): 12055-12067, 2022 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-36074772

RESUMO

G-quadruplex (G4) ligands are investigated to discover new anticancer drugs with increased cell-killing potency. These ligands can induce genome instability and activate innate immune genes at non-cytotoxic doses, opening the discovery of cytostatic immune-stimulating ligands. However, the interplay of G4 affinity/selectivity with cytotoxicity and immune gene activation is not well-understood. We investigated a series of closely related hydrazone derivatives to define the molecular bases of immune-stimulation activity. Although they are closely related to each other, such derivatives differ in G4 affinity, cytotoxicity, genome instability, and immune gene activation. Our findings show that G4 affinity of ligands is a critical feature for immune gene activation, whereas a high cytotoxic potency interferes with it. The balance of G4 stabilization versus cytotoxicity can determine the level of immune gene activation in cancer cells. Thus, we propose a new rationale based on low cell-killing potency and high immune stimulation to discover effective anticancer G4 ligands.


Assuntos
Antineoplásicos , Citostáticos , Quadruplex G , Neoplasias , Antineoplásicos/farmacologia , Instabilidade Genômica , Humanos , Hidrazonas/farmacologia , Interferon beta/genética , Ligantes , Neoplasias/genética
6.
Br J Cancer ; 127(7): 1214-1225, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35794238

RESUMO

BACKGROUND: Current immunotherapy strategies have contrasting clinical results in human lung cancer patients as small-cell lung cancers (SCLC) often show features of immunological cold tumours. Topoisomerase 1 (TOP1) poisons are effective antitumor drugs with good efficacy against lung cancers. METHODS: We used molecular, genetic and bioinformatic approaches to determine the mechanism of micronuclei formation induced by two TOP1 poisons in different human cancer cells, including SCLC cell lines. RESULTS: TOP1 poisons stimulate similar levels of micronuclei in all tested cell lines but downstream effects can vary markedly. TOP1 poisons increase micronuclei levels with a mechanism involving R-loops as overexpression of RNaseH1 markedly reduces or abolishes both H2AX phosphorylation and micronuclei formation. TOP1 poison-induced micronuclei activate the cGAS/STING pathway leading to increased expression of immune genes in HeLa cells, but not in human SCLC cell lines, mainly due to lack of STING and/or cGAS expression. Moreover, the expression of STING and antigen-presenting machinery genes is generally downregulated in patient tumours of human lung cancer datasets. CONCLUSIONS: Altogether, our data reveal an immune signalling mechanism activated by TOP1 poisons, which is often impaired in human SCLC tumours.


Assuntos
Antineoplásicos , Neoplasias Pulmonares , Venenos , Carcinoma de Pequenas Células do Pulmão , Antineoplásicos/uso terapêutico , DNA Topoisomerases Tipo I/genética , DNA Topoisomerases Tipo I/metabolismo , Células HeLa , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/uso terapêutico , Venenos/uso terapêutico , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Ativação Transcricional
7.
Methods Mol Biol ; 2528: 203-213, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35704193

RESUMO

Topoisomerase 1-DNA cleavage complexes (Top1ccs) form at DNA sites of Top1 activity and are increased by a highly specific anticancer drug (camptothecin, CPT) in living cells. Various methods are available to detect Top1ccs in cultured cells, including protocols based on the use of specific antibodies. Here, we describe a protocol to isolate Top1ccs at high purity, which does not depend on antibodies.


Assuntos
Antineoplásicos , Inibidores da Topoisomerase I , Camptotecina/farmacologia , DNA , Clivagem do DNA , DNA Topoisomerases Tipo I/metabolismo
8.
J Clin Nurs ; 29(15-16): 2999-3011, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32350946

RESUMO

AIMS AND OBJECTIVES: To explore the day-to-day experiences of family caregivers who are caring for children with Osteogenesis Imperfecta (OI). BACKGROUND: Osteogenesis Imperfecta is a rare genetic condition known to cause bone fragility. Family caregivers of children with OI play an important role in helping these children live well at home. DESIGN: A qualitative descriptive design was used. METHODS: A qualitative descriptive study was conducted in accordance with the COREQ guidelines. Adult family caregivers (n = 18) of children with OI were recruited from a university-affiliated, paediatric orthopaedic hospital in Montreal, Canada. Individual interviews were conducted, transcribed verbatim and inductively thematically analysed. RESULTS: Osteogenesis Imperfecta family caregiving entailed: (a) managing regular day-to-day caregiving activities, including morning routines, evening routines and the facilitation of their child's mobilisation; (b) coping with periods that made the caregiving routine more challenging, such as fractures, surgeries and pain; and (c) devising long-term strategies to support day-to-day care, such as managing the environment, accessing medical and school resources, and coordinating care and respite. CONCLUSIONS: The day-to-day routine of caring for a child with OI may be disrupted by challenging periods and improved by long-term strategies developed to ease day-to-day care. These strategies suggest future directions for clinicians and policymakers to improve health services and caregiver well-being. RELEVANCE TO CLINICAL PRACTICE: Clinical, policy and research endeavours need to incorporate new interventions to support the needs of family caregivers. These recommendations may be relevant to other clinicians and policymakers working with families living with rare and chronic physical conditions.


Assuntos
Cuidadores/psicologia , Osteogênese Imperfeita/enfermagem , Adaptação Psicológica , Adulto , Cuidadores/organização & administração , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Avaliação das Necessidades , Pesquisa Qualitativa
9.
Cell Rep ; 28(12): 3167-3181.e6, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31533039

RESUMO

Although accumulation of DNA damage and genomic instability in resting cells can cause neurodegenerative disorders, our understanding of how transcription produces DNA double-strand breaks (DSBs) is limited. Transcription-blocking topoisomerase I cleavage complexes (TOP1ccs) are frequent events that prime DSB production in non-replicating cells. Here, we report a mechanism of their formation by showing that they arise from two nearby single-strand breaks (SSBs) on opposing DNA strands: one SSB from the removal of transcription-blocking TOP1ccs by the TDP1 pathway and the other from the cleavage of R-loops by endonucleases, including XPF, XPG, and FEN1. Genetic defects in TOP1cc removal (TDP1, PNKP, and XRCC1) or in the resolution of R-loops (SETX) enhance DSB formation and prevent their repair. Such deficiencies cause neurological disorders. Owing to the high frequency of TOP1cc trapping and the widespread distribution of R-loops, these persistent transcriptional DSBs could accumulate over time in neuronal cells, contributing to the neurodegenerative diseases.


Assuntos
Quebras de DNA de Cadeia Dupla , Quebras de DNA de Cadeia Simples , DNA Topoisomerases Tipo I/metabolismo , Estruturas R-Loop , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Endonucleases/metabolismo , Endonucleases Flap/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo
10.
Proc Natl Acad Sci U S A ; 116(3): 816-825, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30591567

RESUMO

G quadruplexes (G4s) and R loops are noncanonical DNA structures that can regulate basic nuclear processes and trigger DNA damage, genome instability, and cell killing. By different technical approaches, we here establish that specific G4 ligands stabilize G4s and simultaneously increase R-loop levels within minutes in human cancer cells. Genome-wide mapping of R loops showed that the studied G4 ligands likely cause the spreading of R loops to adjacent regions containing G4 structures, preferentially at 3'-end regions of expressed genes, which are partially ligand-specific. Overexpression of an exogenous human RNaseH1 rescued DNA damage induced by G4 ligands in BRCA2-proficient and BRCA2-silenced cancer cells. Moreover, even if the studied G4 ligands increased noncanonical DNA structures at similar levels in nuclear chromatin, their cellular effects were different in relation to cell-killing activity and stimulation of micronuclei, a hallmark of genome instability. Our findings therefore establish that G4 ligands can induce DNA damage by an R loop-dependent mechanism that can eventually lead to different cellular consequences depending on the chemical nature of the ligands.


Assuntos
Dano ao DNA , Quadruplex G , Instabilidade Genômica , Neoplasias/genética , Aminoquinolinas , Linhagem Celular Tumoral , Genes BRCA2 , Humanos , Ligantes , Ácidos Picolínicos
11.
Int J Mol Sci ; 19(11)2018 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-30404148

RESUMO

Mammalian DNA topoisomerases II are targets of anticancer anthracyclines that act by stabilizing enzyme-DNA complexes wherein DNA strands are cut and covalently linked to the protein. This molecular mechanism is the molecular basis of anthracycline anticancer activity as well as the toxic effects such as cardiomyopathy and induction of secondary cancers. Even though anthracyclines have been used in the clinic for more than 50 years for solid and blood cancers, the search of breakthrough analogs has substantially failed. The recent developments of personalized medicine, availability of individual genomic information, and immune therapy are expected to change significantly human cancer therapy. Here, we discuss the knowledge of anthracyclines as Topoisomerase II poisons, their molecular and cellular effects and toxicity along with current efforts to improve the therapeutic index. Then, we discuss the contribution of the immune system in the anticancer activity of anthracyclines, and the need to increase our knowledge of molecular mechanisms connecting the drug targets to the immune stimulatory pathways in cancer cells. We propose that the complete definition of the molecular interaction of anthracyclines with the immune system may open up more effective and safer ways to treat patients with these drugs.


Assuntos
Antraciclinas/farmacologia , Inibidores da Topoisomerase II/farmacologia , Animais , Antraciclinas/efeitos adversos , Antraciclinas/química , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Antineoplásicos/farmacologia , Cardiotoxicidade/etiologia , Morte Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Segunda Neoplasia Primária/etiologia , Inibidores da Topoisomerase II/efeitos adversos , Inibidores da Topoisomerase II/química
12.
Genome Biol ; 19(1): 100, 2018 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-30060749

RESUMO

BACKGROUND: Co-transcriptional R-loops are abundant non-B DNA structures in mammalian genomes. DNA Topoisomerase I (Top1) is often thought to regulate R-loop formation owing to its ability to resolve both positive and negative supercoils. How Top1 regulates R-loop structures at a global level is unknown. RESULTS: Here, we perform high-resolution strand-specific R-loop mapping in human cells depleted for Top1 and find that Top1 depletion results in both R-loop gains and losses at thousands of transcribed loci, delineating two distinct gene classes. R-loop gains are characteristic for long, highly transcribed, genes located in gene-poor regions anchored to Lamin B1 domains and in proximity to H3K9me3-marked heterochromatic patches. R-loop losses, by contrast, occur in gene-rich regions overlapping H3K27me3-marked active replication initiation regions. Interestingly, Top1 depletion coincides with a block of the cell cycle in G0/G1 phase and a trend towards replication delay. CONCLUSIONS: Our findings reveal new properties of Top1 in regulating R-loop homeostasis in a context-dependent manner and suggest a potential role for Top1 in modulating the replication process via R-loop formation.


Assuntos
DNA Topoisomerases Tipo I/genética , DNA/química , Genoma Humano , Heterocromatina/química , Transcrição Gênica , Imunoprecipitação da Cromatina , DNA/genética , DNA/metabolismo , Replicação do DNA , DNA Topoisomerases Tipo I/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular , Inativação Gênica , Células HEK293 , Heterocromatina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Conformação de Ácido Nucleico , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Fase de Repouso do Ciclo Celular
13.
J Med Chem ; 60(23): 9448-9461, 2017 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-29140695

RESUMO

The human KRAS transcript contains a G-rich 5'-UTR sequence (77% GC) harboring several G4 motifs capable to form stable RNA G-quadruplex (RG4) structures that can serve as targets for small molecules. A biotin-streptavidin pull-down assay showed that 4,11-bis(2-aminoethylamino)anthra[2,3-b]furan-5,10-dione (2a) binds to RG4s in the KRAS transcript under low-abundance cellular conditions. Dual-luciferase assays demonstrated that 2a and its analogue 4,11-bis(2-aminoethylamino)anthra[2,3-b]thiophene-5,10-dione (2b) repress translation in a dose-dependent manner. The effect of the G4-ligands on Panc-1 cancer cells has also been examined. Both 2a and 2b efficiently penetrate the cells, suppressing protein p21KRAS to <10% of the control. The KRAS down-regulation induces apoptosis together with a dramatic reduction of cell growth and colony formation. In summary, we report a strategy to suppress the KRAS oncogene in pancreatic cancer cells by means of small molecules binding to RG4s in the 5'-UTR of mRNA.


Assuntos
Regiões 5' não Traduzidas/efeitos dos fármacos , Quadruplex G/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas p21(ras)/genética , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Descoberta de Drogas , Humanos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Tiofenos/química , Tiofenos/farmacologia
14.
J Med Chem ; 60(6): 2169-2192, 2017 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-28072526

RESUMO

DNA topoisomerases constitute a large family of enzymes that are essential for all domains of life. Although they share general reaction chemistry and the capacity to govern DNA topology and resolve strand entanglements during fundamental molecular processes, they are characterized by differences in their structural organization, modes of enzymatic catalysis, and biological functions. Moreover, hundreds of compounds interfere with bacterial and/or eukaryotic enzymes, some of which are effective drugs for the treatment of infectious diseases and cancers. Research over the past decade has focused on the biological functions of DNA topoisomerases, and several findings have revealed unexpected roles of type I DNA topoisomerases, a subclass of these enzymes, in regulating gene expression and DNA and chromatin conformations. These new findings highlight that type I topoisomerases are still interesting targets for drug discovery for the treatment of several human diseases, including multidrug-resistant infections and genetic disorders.


Assuntos
DNA Topoisomerases Tipo I/metabolismo , Descoberta de Drogas , Inibidores da Topoisomerase I/química , Inibidores da Topoisomerase I/farmacologia , Animais , DNA/genética , DNA/metabolismo , DNA Topoisomerases Tipo I/química , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Modelos Moleculares
15.
Biochim Biophys Acta Gen Subj ; 1861(5 Pt B): 1271-1280, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27836755

RESUMO

BACKGROUND: Guanine-rich DNA motifs can form non-canonical structures known as G-quadruplexes, whose role in tumorigenic processes makes them attractive drug-target candidates for cancer therapy. Recent studies revealed that the folding and unfolding pathways of G-quadruplexes proceed through a quite stable intermediate named G-triplex. METHODS: Virtual screening was employed to identify a small set of putative G-triplex ligands. The G-triplex stabilizing properties of these compounds were analyzed by CD melting assay. DSC, non-denaturing gel electrophoresis, NMR and molecular modeling studies were performed to investigate the interaction between the selected compound 1 and G-rich DNA structures. Cytotoxic activity of 1 was evaluated by MTT cell proliferation assay. RESULTS: The experiments led to the identification of a promising hit that was shown to bind preferentially to G-triplex and parallel-stranded G-quadruplexes over duplex and antiparallel G-quadruplexes. Molecular modeling results suggested a partial end-stacking of 1 to the external G-triad/G-tetrads as a binding mode. Biological assays showed that 1 is endowed with cytotoxic effect on human osteosarcoma cells. CONCLUSIONS: A tandem application of virtual screening along with the experimental investigation was employed to discover a G-triplex-targeting ligand. Experiments revealed that the selected compound actually acts as a dual G-triplex/G-quadruplex stabilizer, thus stimulating further studies aimed at its optimization. GENERAL SIGNIFICANCE: The discovery of molecules able to bind and stabilize G-triplex structures is highly appealing, but their transient state makes challenging their recognition. These findings suggest that the identification of ligands with dual G-triplex/G-quadruplex stabilizing properties may represent a new route for the design of anticancer agents targeting the G-rich DNA structures. This article is part of a Special Issue entitled "G-quadruplex" Guest Editor: Dr. Concetta Giancola and Dr. Daniela Montesarchio.


Assuntos
Antineoplásicos/farmacologia , DNA de Neoplasias/efeitos dos fármacos , Desenho de Fármacos , Quadruplex G/efeitos dos fármacos , Guanosina/química , Osteossarcoma/tratamento farmacológico , Antineoplásicos/química , Antineoplásicos/metabolismo , Sítios de Ligação , Varredura Diferencial de Calorimetria , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dicroísmo Circular , DNA de Neoplasias/química , DNA de Neoplasias/metabolismo , Relação Dose-Resposta a Droga , Guanosina/metabolismo , Humanos , Concentração Inibidora 50 , Ligantes , Espectroscopia de Ressonância Magnética , Simulação de Acoplamento Molecular , Eletroforese em Gel de Poliacrilamida Nativa , Osteossarcoma/genética , Osteossarcoma/patologia , Relação Estrutura-Atividade , Fatores de Tempo
16.
PLoS One ; 11(1): e0147053, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26784695

RESUMO

Topoisomerase I-DNA-cleavage complexes (Top1cc) stabilized by camptothecin (CPT) have specific effects at transcriptional levels. We recently reported that Top1cc increase antisense transcript (aRNAs) levels at divergent CpG-island promoters and, transiently, DNA/RNA hybrids (R-loop) in nuclear and mitochondrial genomes of colon cancer HCT116 cells. However, the relationship between R-loops and aRNAs was not established. Here, we show that aRNAs can form R-loops in N-TERA-2 cells under physiological conditions, and that promoter-associated R-loops are somewhat increased and extended in length immediately upon cell exposure to CPT. In contrast, persistent Top1ccs reduce the majority of R-loops suggesting that CPT-accumulated aRNAs are not commonly involved in R-loops. The enhancement of aRNAs by Top1ccs is present both in human colon cancer HCT116 cells and WI38 fibroblasts suggesting a common response of cancer and normal cells. Although Top1ccs lead to DSB and DDR kinases activation, we do not detect a dependence of aRNA accumulation on ATM or DNA-PK activation. However, we showed that the cell response to persistent Top1ccs can involve an impairment of aRNA turnover rather than a higher synthesis rate. Finally, a genome-wide analysis shows that persistent Top1ccs also determine an accumulation of sense transcripts at 5'-end gene regions suggesting an increased occurrence of truncated transcripts. Taken together, the results indicate that Top1 may regulate transcription initiation by modulating RNA polymerase-generated negative supercoils, which can in turn favor R-loop formation at promoters, and that transcript accumulation at TSS is a response to persistent transcriptional stress by Top1 poisoning.


Assuntos
Camptotecina/farmacologia , Replicação do DNA , DNA Topoisomerases Tipo I/química , Regiões Promotoras Genéticas/genética , Inibidores da Topoisomerase I/farmacologia , DNA Topoisomerases Tipo I/genética , Células HCT116 , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Transcrição Gênica
17.
PLoS One ; 9(12): e114904, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25489853

RESUMO

DNA topoisomerases are enzymes responsible for the relaxation of DNA torsional strain, as well as for the untangling of DNA duplexes after replication, and are important cancer drug targets. One class of topoisomerase inhibitors, "poisons", binds to the transient enzyme-DNA complex which occurs during the mechanism of action, and inhibits the religation of DNA. This ultimately leads to the accumulation of DNA double strand breaks and cell death. Different types of topoisomerases occur in human cells and several poisons of topoisomerase I and II are widely used clinically. However, their use is compromised by a variety of side effects. Recent studies confirm that the inhibition of the α-isoform of topoisomerase II is responsible for the cytotoxic effect, whereas the inhibition of the ß-isoform leads to development of adverse drug reactions. Thus, the discovery of agents selective for topoisomerase IIα is an important strategy for the development of topoisomerase II poisons with improved clinical profiles. Here, we present a computer-aided drug design study leading to the identification of structurally novel topoisomerase IIα poisons. The study combines ligand- and structure-based drug design methods including pharmacophore models, homology modelling, docking, and virtual screening of the National Cancer Institute compound database. From the 8 compounds identified from the computational work, 6 were tested for their capacity to poison topoisomerase II in vitro: 4 showed selective inhibitory activity for the α- over the ß-isoform and 3 of these exhibited cytotoxic activity. Thus, our study confirms the applicability of computer-aided methods for the discovery of novel topoisomerase II poisons, and presents compounds which could be investigated further as selective topoisomerase IIα inhibitors.


Assuntos
Antígenos de Neoplasias/química , Clivagem do DNA/efeitos dos fármacos , DNA Topoisomerases Tipo II/química , Proteínas de Ligação a DNA/química , Bases de Dados de Produtos Farmacêuticos , Simulação de Acoplamento Molecular , Conformação Proteica , Inibidores da Topoisomerase II/farmacologia , Antígenos de Neoplasias/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Desenho de Fármacos , Humanos , Modelos Moleculares
18.
Mol Cancer Ther ; 13(1): 239-48, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24252850

RESUMO

DNA topoisomerase I (Top1) inhibition by camptothecin derivatives can impair the hypoxia-induced cell transcriptional response. In the present work, we determined molecular aspects of the mechanism of camptothecin's effects on hypoxia-inducible factor-1α (HIF-1α) activity in human cancer cells. In particular, we provide evidence that low concentrations of camptothecin, without interfering with HIF-1α mRNA levels, can reduce HIF-1α protein expression and activity. As luciferase assays demonstrated the involvement of the HIF-1α mRNA 3' untranslated region in camptothecin-induced impairment of HIF-1α protein regulation, we performed microarray analysis to identify camptothecin-induced modification of microRNAs (miRNA) targeting HIF-1α mRNA under hypoxic-mimetic conditions. The selected miRNAs were then further analyzed, demonstrating a role for miR-17-5p and miR-155 in HIF-1α protein expression after camptothecin treatments. The present findings establish miRNAs as key factors in a molecular pathway connecting Top1 inhibition and human HIF-1α protein regulation and activity, widening the biologic and molecular activity of camptothecin derivatives and the perspective for novel clinical interventions.


Assuntos
DNA Topoisomerases Tipo I/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , MicroRNAs/biossíntese , Neoplasias/genética , Camptotecina/administração & dosagem , Hipóxia Celular/efeitos dos fármacos , DNA Topoisomerases Tipo I/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , MicroRNAs/genética , Neoplasias/patologia , RNA Mensageiro/genética , Inibidores da Topoisomerase I/administração & dosagem
19.
Nucleic Acids Res ; 41(22): 10110-23, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23999093

RESUMO

DNA Topoisomerase I (Top1) is required to relax DNA supercoils generated by RNA polymerases (RNAPs). Top1 is inhibited with high specificity by camptothecin (CPT), an effective anticancer agent, and by oxidative base damage and ribonucleotides in DNA strands, resulting into Top1-DNA cleavage complexes (Top1ccs). To understand how Top1ccs affect genome stability, we have investigated the global transcriptional response to CPT-induced Top1ccs. Top1ccs trigger an accumulation of antisense RNAPII transcripts specifically at active divergent CpG-island promoters in a replication-independent and Top1-dependent manner. As CPT increases antisense transcript levels in the presence of 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole, a transcription inhibitor, Top1ccs likely impair antisense RNA degradation. Time-course data showed a burst of Top1ccs increased by CPT at promoter sites and along transcribed regions, causing a transient block of RNAPII at the promoter. Moreover, cell immunofluorescence analyses showed that Top1ccs induce a transient increase of R-loops specifically at highly transcribed regions such as nucleoli in a Top1-dependent manner. Thus, a specific and highly dynamic transcriptional response to Top1ccs occurs at divergent active CpG-island promoters, which may include a transient stabilization of R-loops. The results clarify molecular features of a response pathway leading to transcription-dependent genome instability and altered transcription regulation.


Assuntos
Camptotecina/farmacologia , Ilhas de CpG , Clivagem do DNA , DNA Topoisomerases Tipo I/metabolismo , Regiões Promotoras Genéticas , RNA Antissenso/biossíntese , Inibidores da Topoisomerase I/farmacologia , Linhagem Celular Tumoral , Quinase 9 Dependente de Ciclina/metabolismo , DNA/química , Replicação do DNA , Humanos , RNA Polimerase II/metabolismo
20.
J Exp Clin Cancer Res ; 32: 24, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23631805

RESUMO

BACKGROUND: Tumors are diseases characterized by uncontrolled cell growth and, in spite of the progress of medicine over the years, continue to represent a major threat to the health, requiring new therapies. Several synthetic compounds, such as those derived from natural sources, have been identified as anticancer drugs; among these compounds quinone represent the second largest class of anticancer agents in use. Several studies have shown that these act on tumor cells through several mechanisms. An important objective of this work is to develop quinoidscompounds showing antitumor activity, but with fewer side effects. The parachinone cannabinol HU-331, is a small molecule that with its core 4-hydroxy-1,4-benzoquinone, exhibits a potent and selective cytotoxic activity on different tumor cell lines. A series of derivatives 3-hydroxy-1,4-benzochinoni were thus developed through HU-331 chemical modifications. The purpose of the work is to test the ability of the compounds to induce proliferative inhibition and study the mechanisms of cell death. METHODS: The antitumor activities were evaluated in vitro by examining their cytotoxic effects against different human cancer cell lines. All cell lines tested were plated in 96-multiwell and treated with HU-100-V at different concentrations and cell viability was evaluated byMTT assay. Subsequently via flow cytometry (FACS) it was possible to assess apoptosis by the system of double labeling with PI and Annexin-V, and the effect of the compounds on ROS formation by measuring the dichlorofluorescein fluorescence. RESULTS: The substitution by n-hexyl chain considerably enhanced the bioactivity of the compounds. In details, 2-hexyl-5-hydroxycyclohexa-2,5-diene-1,4-dione (V), 2,5-Dimethoxy-3-hexyl-2,5-cyclohexadiene-1,4-dione (XII) and 2-hydroxy-5-methoxy-3-hexyl-cyclohexa-2,5-diene-1,4-dione (XIII) showed most prominent cytotoxicity against almost human tumour cell lines. Compound V was further subjected to downstream apoptotic analysis, demostrating a time-dependent pro-apoptotic activity on human melanoma M14 cell line mediated by caspases activation and poly-(ADP-ribose)-polymerase (PARP) protein cleavage. CONCLUSIONS: These findings indicate that 2-hexyl-5-idrossicicloesa-2,5-diene-1,4-dione can be a promising compound for the design of a new class of antineoplastic derivatives.Carmen Petronzi, Michela Festa, Antonella Peduto and Maria Castellano: equally contributed equally to this work.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Benzoquinonas/química , Benzoquinonas/farmacologia , Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Benzoquinonas/toxicidade , Canabidiol/análogos & derivados , Canabidiol/farmacologia , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Desenho de Fármacos , Humanos , Concentração Inibidora 50 , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Inibidores da Topoisomerase/química , Inibidores da Topoisomerase/farmacologia , Inibidores da Topoisomerase/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...