Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 52(3): 1136-1155, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38038252

RESUMO

Maintaining chromatin integrity at the repetitive non-coding DNA sequences underlying centromeres is crucial to prevent replicative stress, DNA breaks and genomic instability. The concerted action of transcriptional repressors, chromatin remodelling complexes and epigenetic factors controls transcription and chromatin structure in these regions. The histone chaperone complex ATRX/DAXX is involved in the establishment and maintenance of centromeric chromatin through the deposition of the histone variant H3.3. ATRX and DAXX have also evolved mutually-independent functions in transcription and chromatin dynamics. Here, using paediatric glioma and pancreatic neuroendocrine tumor cell lines, we identify a novel ATRX-independent function for DAXX in promoting genome stability by preventing transcription-associated R-loop accumulation and DNA double-strand break formation at centromeres. This function of DAXX required its interaction with histone H3.3 but was independent of H3.3 deposition and did not reflect a role in the repression of centromeric transcription. DAXX depletion mobilized BRCA1 at centromeres, in line with BRCA1 role in counteracting centromeric R-loop accumulation. Our results provide novel insights into the mechanisms protecting the human genome from chromosomal instability, as well as potential perspectives in the treatment of cancers with DAXX alterations.


Assuntos
Centrômero , Quebras de DNA de Cadeia Dupla , Chaperonas Moleculares , Proteínas Nucleares , Estruturas R-Loop , Proteína Nuclear Ligada ao X , Criança , Humanos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Centrômero/metabolismo , Cromatina , Proteínas Correpressoras/metabolismo , DNA , Histonas/genética , Histonas/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteína Nuclear Ligada ao X/genética , Proteína Nuclear Ligada ao X/metabolismo
2.
J Med Chem ; 66(20): 13918-13945, 2023 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-37816126

RESUMO

A series of 25 chiral anti-cancer lipidic alkynylcarbinols (LACs) were devised by introducing an (hetero)aromatic ring between the aliphatic chain and the dialkynylcarbinol warhead. The resulting phenyl-dialkynylcarbinols (PACs) exhibit enhanced stability, while retaining cytotoxicity against HCT116 and U2OS cell lines with IC50 down to 40 nM for resolved eutomers. A clickable probe was used to confirm the PAC prodrug behavior: upon enantiospecific bio-oxidation of the carbinol by the HSD17B11 short-chain dehydrogenase/reductase (SDR), the resulting ynones covalently modify cellular proteins, leading to endoplasmic reticulum stress, ubiquitin-proteasome system inhibition, and apoptosis. Insights into the design of LAC prodrugs specifically bioactivated by HSD17B11 vs its paralogue HSD17B13 were obtained. The HSD17B11/HSD17B13-dependent cytotoxicity of PACs was exploited to develop a cellular assay to identify specific inhibitors of these enzymes. A docking study was performed with the HSD17B11 AlphaFold model, providing a molecular basis of the SDR substrates mimicry by PACs. The safety profile of a representative PAC was established in mice.


Assuntos
Alcinos , Antineoplásicos , Camundongos , Animais , Alcinos/farmacologia , Alcinos/química , Antineoplásicos/farmacologia , Antineoplásicos/química , Acetileno , Estrutura Molecular , Lipídeos/química , Linhagem Celular Tumoral
3.
Eur J Med Chem ; 259: 115646, 2023 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-37482022

RESUMO

Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) affects 10 million people each year and the emergence of resistant TB augurs for a growing incidence. In the last 60 years, only three new drugs were approved for TB treatment, for which resistances are already emerging. Therefore, there is a crucial need for new chemotherapeutic agents capable of eradicating TB. Enzymes belonging to the type II fatty acid synthase system (FAS-II) are involved in the biosynthesis of mycolic acids, cell envelope components essential for mycobacterial survival. Among them, InhA is the primary target of isoniazid (INH), one of the most effective compounds to treat TB. INH acts as a prodrug requiring activation by the catalase-peroxidase KatG, whose mutations are the major cause for INH resistance. Herein, a new series of direct InhA inhibitors were designed based on a molecular hybridization approach. They exhibit potent inhibitory activities of InhA and, for some of them, good antitubercular activities. Moreover, they display a low toxicity on human cells. A study of the mechanism of action of the most effective molecules shows that they inhibit the biosynthesis of mycolic acids. The X-ray structures of two InhA/NAD+/inhibitor complexes have been obtained showing a binding mode of a part of the molecule in the minor portal, rarely seen in the InhA structures reported so far.


Assuntos
Antituberculosos , Mycobacterium tuberculosis , Humanos , Antituberculosos/farmacologia , Antituberculosos/química , Proteínas de Bactérias/metabolismo , Éter , Éteres/farmacologia , Etil-Éteres/farmacologia , Isoniazida/farmacologia , Mutação , Ácidos Micólicos
4.
Elife ; 112022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35535493

RESUMO

Hundreds of cytotoxic natural or synthetic lipidic compounds contain chiral alkynylcarbinol motifs, but the mechanism of action of those potential therapeutic agents remains unknown. Using a genetic screen in haploid human cells, we discovered that the enantiospecific cytotoxicity of numerous terminal alkynylcarbinols, including the highly cytotoxic dialkynylcarbinols, involves a bioactivation by HSD17B11, a short-chain dehydrogenase/reductase (SDR) known to oxidize the C-17 carbinol center of androstan-3-alpha,17-beta-diol to the corresponding ketone. A similar oxidation of dialkynylcarbinols generates dialkynylketones, that we characterize as highly protein-reactive electrophiles. We established that, once bioactivated in cells, the dialkynylcarbinols covalently modify several proteins involved in protein-quality control mechanisms, resulting in their lipoxidation on cysteines and lysines through Michael addition. For some proteins, this triggers their association to cellular membranes and results in endoplasmic reticulum stress, unfolded protein response activation, ubiquitin-proteasome system inhibition and cell death by apoptosis. Finally, as a proof-of-concept, we show that generic lipidic alkynylcarbinols can be devised to be bioactivated by other SDRs, including human RDH11 and HPGD/15-PGDH. Given that the SDR superfamily is one of the largest and most ubiquitous, this unique cytotoxic mechanism-of-action could be widely exploited to treat diseases, in particular cancer, through the design of tailored prodrugs.


Assuntos
Antineoplásicos , Redutases-Desidrogenases de Cadeia Curta , Antineoplásicos/farmacologia , Estresse do Retículo Endoplasmático , Humanos , Lipídeos , Resposta a Proteínas não Dobradas
5.
Cell Death Dis ; 12(10): 896, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34599155

RESUMO

Centromeres are defined by chromatin containing the histone H3 variant CENP-A assembled onto repetitive α-satellite sequences, which are actively transcribed throughout the cell cycle. Centromeres play an essential role in chromosome inheritance and genome stability through coordinating kinetochores assembly during mitosis. Structural and functional alterations of the centromeres cause aneuploidy and chromosome aberrations which can induce cell death. In human cells, the tumor suppressor BRCA1 associates with centromeric chromatin in the absence of exogenous damage. While we previously reported that BRCA1 contributes to proper centromere homeostasis, the mechanism underlying its centromeric function and recruitment was not fully understood. Here, we show that BRCA1 association with centromeric chromatin depends on the presence of R-loops, which are non-canonical three-stranded structures harboring a DNA:RNA hybrid and are frequently formed during transcription. Subsequently, BRCA1 counteracts the accumulation of R-loops at centromeric α-satellite repeats. Strikingly, BRCA1-deficient cells show impaired localization of CENP-A, higher transcription of centromeric RNA, increased breakage at centromeres and formation of acentric micronuclei, all these features being R-loop-dependent. Finally, BRCA1 depletion reveals a Rad52-dependent hyper-recombination process between centromeric satellite repeats, associated with centromere instability and missegregation. Altogether, our findings provide molecular insights into the key function of BRCA1 in maintaining centromere stability and identity.


Assuntos
Proteína BRCA1/metabolismo , Centrômero/metabolismo , Estruturas R-Loop , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , DNA Satélite/genética , Humanos , Modelos Biológicos , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Recombinação Genética/genética
6.
Nucleic Acids Res ; 49(18): 10275-10288, 2021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34551430

RESUMO

DNA is intrinsically dynamic and folds transiently into alternative higher-order structures such as G-quadruplexes (G4s) and three-way DNA junctions (TWJs). G4s and TWJs can be stabilised by small molecules (ligands) that have high chemotherapeutic potential, either as standalone DNA damaging agents or combined in synthetic lethality strategies. While previous approaches have claimed to use ligands that specifically target either G4s or TWJs, we report here on a new approach in which ligands targeting both TWJs and G4s in vitro demonstrate cellular effects distinct from that of G4 ligands, and attributable to TWJ targeting. The DNA binding modes of these new, dual TWJ-/G4-ligands were studied by a panel of in vitro methods and theoretical simulations, and their cellular properties by extensive cell-based assays. We show here that cytotoxic activity of TWJ-/G4-ligands is mitigated by the DNA damage response (DDR) and DNA topoisomerase 2 (TOP2), making them different from typical G4-ligands, and implying a pivotal role of TWJs in cells. We designed and used a clickable ligand, TrisNP-α, to provide unique insights into the TWJ landscape in cells and its modulation upon co-treatments. This wealth of data was exploited to design an efficient synthetic lethality strategy combining dual ligands with clinically relevant DDR inhibitors.


Assuntos
Antineoplásicos/farmacologia , Compostos Azabicíclicos/farmacologia , Dano ao DNA/efeitos dos fármacos , DNA , Quadruplex G/efeitos dos fármacos , Neoplasias/genética , DNA/química , DNA/metabolismo , Humanos , Células MCF-7 , Relação Estrutura-Atividade
7.
Nucleic Acids Res ; 49(17): 9906-9925, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34500463

RESUMO

Replication-associated single-ended DNA double-strand breaks (seDSBs) are repaired predominantly through RAD51-mediated homologous recombination (HR). Removal of the non-homologous end-joining (NHEJ) factor Ku from resected seDSB ends is crucial for HR. The coordinated actions of MRE11-CtIP nuclease activities orchestrated by ATM define one pathway for Ku eviction. Here, we identify the pre-mRNA splicing protein XAB2 as a factor required for resistance to seDSBs induced by the chemotherapeutic alkylator temozolomide. Moreover, we show that XAB2 prevents Ku retention and abortive HR at seDSBs induced by temozolomide and camptothecin, via a pathway that operates in parallel to the ATM-CtIP-MRE11 axis. Although XAB2 depletion preserved RAD51 focus formation, the resulting RAD51-ssDNA associations were unproductive, leading to increased NHEJ engagement in S/G2 and genetic instability. Overexpression of RAD51 or RAD52 rescued the XAB2 defects and XAB2 loss was synthetically lethal with RAD52 inhibition, providing potential perspectives in cancer therapy.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/genética , Autoantígeno Ku/metabolismo , Fatores de Processamento de RNA/metabolismo , Alquilantes/efeitos adversos , Alquilantes/farmacologia , Camptotecina/efeitos adversos , Camptotecina/farmacologia , Linhagem Celular Tumoral , Endodesoxirribonucleases/metabolismo , Glioblastoma/tratamento farmacológico , Recombinação Homóloga/genética , Humanos , Proteína Homóloga a MRE11/metabolismo , Interferência de RNA , Fatores de Processamento de RNA/genética , RNA Interferente Pequeno/genética , Rad51 Recombinase/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Temozolomida/efeitos adversos , Temozolomida/farmacologia
8.
Elife ; 102021 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-34180392

RESUMO

G-quadruplexes (G4) are non-canonical DNA structures found in the genome of most species including human. Small molecules stabilizing these structures, called G4 ligands, have been identified and, for some of them, shown to induce cytotoxic DNA double-strand breaks. Through the use of an unbiased genetic approach, we identify here topoisomerase 2α (TOP2A) as a major effector of cytotoxicity induced by two clastogenic G4 ligands, pyridostatin and CX-5461, the latter molecule currently undergoing phase I/II clinical trials in oncology. We show that both TOP2 activity and transcription account for DNA break production following G4 ligand treatments. In contrast, clastogenic activity of these G4 ligands is countered by topoisomerase 1 (TOP1), which limits co-transcriptional G4 formation, and by factors promoting transcriptional elongation. Altogether our results support that clastogenic G4 ligands act as DNA structure-driven TOP2 poisons at transcribed regions bearing G4 structures.


Assuntos
Aminoquinolinas/farmacologia , Antineoplásicos/farmacologia , Benzotiazóis/farmacologia , DNA Topoisomerases Tipo II/metabolismo , Naftiridinas/farmacologia , Ácidos Picolínicos/farmacologia , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Linhagem Celular , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Quebras de DNA de Cadeia Dupla , DNA Topoisomerases Tipo I/genética , DNA Topoisomerases Tipo I/metabolismo , DNA Topoisomerases Tipo II/genética , Quadruplex G , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas de Ligação a Poli-ADP-Ribose/genética , Polimorfismo de Nucleotídeo Único , Interferência de RNA , RNA-Seq
9.
RSC Chem Biol ; 2(1): 47-76, 2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-35340894

RESUMO

Damaging DNA is a current and efficient strategy to fight against cancer cell proliferation. Numerous mechanisms exist to counteract DNA damage, collectively referred to as the DNA damage response (DDR) and which are commonly dysregulated in cancer cells. Precise knowledge of these mechanisms is necessary to optimise chemotherapeutic DNA targeting. New research on DDR has uncovered a series of promising therapeutic targets, proteins and nucleic acids, with application notably via an approach referred to as combination therapy or combinatorial synthetic lethality. In this review, we summarise the cornerstone discoveries which gave way to the DNA being considered as an anticancer target, and the manipulation of DDR pathways as a valuable anticancer strategy. We describe in detail the DDR signalling and repair pathways activated in response to DNA damage. We then summarise the current understanding of non-B DNA folds, such as G-quadruplexes and DNA junctions, when they are formed and why they can offer a more specific therapeutic target compared to that of canonical B-DNA. Finally, we merge these subjects to depict the new and highly promising chemotherapeutic strategy which combines enhanced-specificity DNA damaging and DDR targeting agents. This review thus highlights how chemical biology has given rise to significant scientific advances thanks to resolutely multidisciplinary research efforts combining molecular and cell biology, chemistry and biophysics. We aim to provide the non-specialist reader a gateway into this exciting field and the specialist reader with a new perspective on the latest results achieved and strategies devised.

10.
J Am Chem Soc ; 142(1): 424-435, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31833764

RESUMO

Translocation of DNA and RNA polymerases along their duplex substrates results in DNA supercoiling. This torsional stress promotes the formation of plectonemic structures, including three-way DNA junction (TWJ), which can block DNA transactions and lead to DNA damage. While cells have evolved multiple mechanisms to prevent the accumulation of such structures, stabilizing TWJ through ad hoc ligands offer an opportunity to trigger DNA damage in cells with high levels of transcription and replication, such as cancer cells. Here, we develop a series of azacryptand-based TWJ ligands, we thoroughly characterize their TWJ-interacting properties in vitro and demonstrate their capacity to trigger DNA damage in rapidly dividing human cancer cells. We also demonstrate that TWJ ligands are amenable to chemically induced synthetic lethality strategies upon association with inhibitors of DNA repair, thus paving the way toward innovative drug combinations to fight cancers.


Assuntos
Dano ao DNA , Reparo do DNA/efeitos dos fármacos , DNA/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Ligantes , Células MCF-7 , Conformação de Ácido Nucleico
11.
J Med Chem ; 62(9): 4456-4466, 2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-30942581

RESUMO

The human genome is replete with repetitive DNA sequences that can fold into thermodynamically stable secondary structures such as hairpins and quadruplexes. Cellular enzymes exist to cope with these structures whose stable accumulation would result in DNA damage through interference with DNA transactions such as transcription and replication. Therefore, the chemical stabilization of secondary DNA structures offers an attractive way to foster DNA transaction-associated damages to trigger cell death in proliferating cancer cells. While much emphasis has been recently given to DNA quadruplexes, we focused here on three-way DNA junctions (TWJ) and report on a strategy to identify TWJ-targeting agents through a combination of in vitro techniques (TWJ-screen, polyacrylamide gel electrophoresis, fluorescence resonance energy transfer-melting, electrospray ionization mass spectrometry, dialysis equilibrium, and sulforhodamine B assays). We designed a complete workflow and screened 1200 compounds to identify promising TWJ ligands selected on stringent criteria in terms of TWJ-folding ability, affinity, and selectivity.


Assuntos
DNA/metabolismo , Bibliotecas de Moléculas Pequenas/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA/genética , Eletroforese em Gel de Poliacrilamida , Transferência Ressonante de Energia de Fluorescência , Humanos , Ligantes , Conformação de Ácido Nucleico , Bibliotecas de Moléculas Pequenas/farmacologia , Espectrometria de Massas por Ionização por Electrospray
12.
Chembiochem ; 19(23): 2438-2442, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30303294

RESUMO

The first biologically relevant clickable probe related to the antitumor marine lipid jaspine B is reported. The concise synthetic route to both enantiomers relied on the supercritical fluid chromatography (SFC) enantiomeric resolution of racemic materials. The eutomeric dextrogyre derivative represents the first jaspine B analogue with enhanced cytotoxicity with IC50 down to 30 nm. These enantiomeric probes revealed a chiralitydependent cytoplasmic imaging of U2OS cancer cells by in situ click labeling.


Assuntos
Alcinos/química , Antineoplásicos/química , Corantes Fluorescentes/química , Sondas Moleculares/química , Esfingosina/análogos & derivados , Alcinos/síntese química , Alcinos/toxicidade , Antineoplásicos/síntese química , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Química Click , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/toxicidade , Humanos , Sondas Moleculares/síntese química , Sondas Moleculares/toxicidade , Esfingosina/síntese química , Esfingosina/toxicidade , Estereoisomerismo
13.
Nat Commun ; 7: 12889, 2016 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-27641979

RESUMO

Repair of single-ended DNA double-strand breaks (seDSBs) by homologous recombination (HR) requires the generation of a 3' single-strand DNA overhang by exonuclease activities in a process called DNA resection. However, it is anticipated that the highly abundant DNA end-binding protein Ku sequesters seDSBs and shields them from exonuclease activities. Despite pioneering works in yeast, it is unclear how mammalian cells counteract Ku at seDSBs to allow HR to proceed. Here we show that in human cells, ATM-dependent phosphorylation of CtIP and the epistatic and coordinated actions of MRE11 and CtIP nuclease activities are required to limit the stable loading of Ku on seDSBs. We also provide evidence for a hitherto unsuspected additional mechanism that contributes to prevent Ku accumulation at seDSBs, acting downstream of MRE11 endonuclease activity and in parallel with MRE11 exonuclease activity. Finally, we show that Ku persistence at seDSBs compromises Rad51 focus assembly but not DNA resection.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas de Transporte/metabolismo , Quebras de DNA de Cadeia Dupla , Autoantígeno Ku/metabolismo , Proteína Homóloga a MRE11/metabolismo , Proteínas Nucleares/metabolismo , Linhagem Celular Tumoral , Reparo do DNA , Endodesoxirribonucleases , Humanos , Fosforilação , Rad51 Recombinase/metabolismo
14.
EMBO Rep ; 17(4): 508-18, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26964895

RESUMO

Ku heterodimer is a DNA binding protein with a prominent role in DNA repair. Here, we investigate whether and how Ku impacts the DNA damage response by acting as a post-transcriptional regulator of gene expression. We show that Ku represses p53 protein synthesis and p53-mediated apoptosis by binding to a bulged stem-loop structure within the p53 5' UTR However, Ku-mediated translational repression of the p53 mRNA is relieved after genotoxic stress. The underlying mechanism involves Ku acetylation which disrupts Ku-p53 mRNA interactions. These results suggest that Ku-mediated repression of p53 mRNA translation constitutes a novel mechanism linking DNA repair and mRNA translation.


Assuntos
Dano ao DNA/fisiologia , Reparo do DNA , Autoantígeno Ku/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/genética , Proteína Supressora de Tumor p53/genética , Regiões 5' não Traduzidas , Acetilação , Apoptose , Dano ao DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Humanos , Autoantígeno Ku/genética , Ligação Proteica , RNA Mensageiro/metabolismo , Proteína Supressora de Tumor p53/metabolismo
15.
Mol Cell Biol ; 35(15): 2699-713, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25986610

RESUMO

Scaffold attachment factor A (SAF-A), also called heterogenous nuclear ribonuclear protein U (hnRNP-U), is phosphorylated on serine 59 by the DNA-dependent protein kinase (DNA-PK) in response to DNA damage. Since SAF-A, DNA-PK catalytic subunit (DNA-PKcs), and protein phosphatase 6 (PP6), which interacts with DNA-PKcs, have all been shown to have roles in mitosis, we asked whether DNA-PKcs phosphorylates SAF-A in mitosis. We show that SAF-A is phosphorylated on serine 59 in mitosis, that phosphorylation requires polo-like kinase 1 (PLK1) rather than DNA-PKcs, that SAF-A interacts with PLK1 in nocodazole-treated cells, and that serine 59 is dephosphorylated by protein phosphatase 2A (PP2A) in mitosis. Moreover, cells expressing SAF-A in which serine 59 is mutated to alanine have multiple characteristics of aberrant mitoses, including misaligned chromosomes, lagging chromosomes, polylobed nuclei, and delayed passage through mitosis. Our findings identify serine 59 of SAF-A as a new target of both PLK1 and PP2A in mitosis and reveal that both phosphorylation and dephosphorylation of SAF-A serine 59 by PLK1 and PP2A, respectively, are required for accurate and timely exit from mitosis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo U/metabolismo , Mitose/genética , Proteína Fosfatase 2/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteína Quinase CDC2 , Linhagem Celular Tumoral , Ciclina B1/metabolismo , Dano ao DNA/genética , Células HeLa , Humanos , Nocodazol/farmacologia , Paclitaxel/farmacologia , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Interferência de RNA , RNA Interferente Pequeno , Securina/metabolismo , Moduladores de Tubulina/farmacologia , Quinase 1 Polo-Like
16.
Cell Rep ; 11(5): 704-14, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25921528

RESUMO

The activities of many DNA-repair proteins are controlled through reversible covalent modification by ubiquitin and ubiquitin-like molecules. Nonhomologous end-joining (NHEJ) is the predominant DNA double-strand break (DSB) repair pathway in mammalian cells and is initiated by DSB ends being recognized by the Ku70/Ku80 (Ku) heterodimer. By using MLN4924, an anti-cancer drug in clinical trials that specifically inhibits conjugation of the ubiquitin-like protein, NEDD8, to target proteins, we demonstrate that NEDD8 accumulation at DNA-damage sites is a highly dynamic process. In addition, we show that depleting cells of the NEDD8 E2-conjugating enzyme, UBE2M, yields ionizing radiation hypersensitivity and reduced cell survival following NHEJ. Finally, we demonstrate that neddylation promotes Ku ubiquitylation after DNA damage and release of Ku and Ku-associated proteins from damage sites following repair. These studies provide insights into how the NHEJ core complex dissociates from repair sites and highlight its importance for cell survival following DSB induction.


Assuntos
Antígenos Nucleares/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Ubiquitinas/metabolismo , Antígenos Nucleares/química , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Ciclopentanos/toxicidade , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/química , Histonas/metabolismo , Humanos , Autoantígeno Ku , Proteína NEDD8 , Ligação Proteica , Estrutura Terciária de Proteína , Proteômica , Pirimidinas/toxicidade , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Radiação Ionizante , Enzimas de Conjugação de Ubiquitina/antagonistas & inibidores , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinação/efeitos dos fármacos , Ubiquitinas/antagonistas & inibidores
17.
Nucleic Acids Res ; 42(14): 9047-62, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25030905

RESUMO

We previously identified the heterogeneous ribonucleoprotein SAF-A/hnRNP U as a substrate for DNA-PK, a protein kinase involved in DNA damage response (DDR). Using laser micro-irradiation in human cells, we report here that SAF-A exhibits a two-phase dynamics at sites of DNA damage, with a rapid and transient recruitment followed by a prolonged exclusion. SAF-A recruitment corresponds to its binding to Poly(ADP-ribose) while its exclusion is dependent on the activity of ATM, ATR and DNA-PK and reflects the dissociation from chromatin of SAF-A associated with ongoing transcription. Having established that SAF-A RNA-binding domain recapitulates SAF-A dynamics, we show that this domain is part of a complex comprising several mRNA biogenesis proteins of which at least two, FUS/TLS and TAFII68/TAF15, exhibit similar biphasic dynamics at sites of damage. Using an original reporter for live imaging of DNA:RNA hybrids (R-loops), we show a transient transcription-dependent accumulation of R-loops at sites of DNA damage that is prolonged upon inhibition of RNA biogenesis factors exclusion. We propose that a new component of the DDR is an active anti-R-loop mechanism operating at damaged transcribed sites which includes the exclusion of mRNA biogenesis factors such as SAF-A, FUS and TAF15.


Assuntos
Dano ao DNA , Ribonucleoproteínas Nucleares Heterogêneas Grupo U/metabolismo , Proteínas de Ligação a RNA/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo U/química , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Estrutura Terciária de Proteína , Proteína FUS de Ligação a RNA/metabolismo , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Transcrição Gênica
18.
Science ; 344(6183): 527-32, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24786082

RESUMO

Down-regulation and mutations of the nuclear-architecture proteins lamin A and C cause misshapen nuclei and altered chromatin organization associated with cancer and laminopathies, including the premature-aging disease Hutchinson-Gilford progeria syndrome (HGPS). Here, we identified the small molecule "Remodelin" that improved nuclear architecture, chromatin organization, and fitness of both human lamin A/C-depleted cells and HGPS-derived patient cells and decreased markers of DNA damage in these cells. Using a combination of chemical, cellular, and genetic approaches, we identified the acetyl-transferase protein NAT10 as the target of Remodelin that mediated nuclear shape rescue in laminopathic cells via microtubule reorganization. These findings provide insights into how NAT10 affects nuclear architecture and suggest alternative strategies for treating laminopathies and aging.


Assuntos
Núcleo Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Hidrazonas/farmacologia , Acetiltransferase N-Terminal E/antagonistas & inibidores , Progéria/enzimologia , Tiazóis/farmacologia , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/ultraestrutura , Cromatina/metabolismo , Inibidores Enzimáticos/química , Humanos , Hidrazonas/química , Lamina Tipo A/genética , Microtúbulos/metabolismo , Acetiltransferase N-Terminal E/química , Acetiltransferase N-Terminal E/genética , Acetiltransferases N-Terminal , Nocodazol/farmacologia , Progéria/genética , Estrutura Terciária de Proteína , RNA Interferente Pequeno/genética , Tiazóis/química
19.
J Cell Biol ; 202(3): 579-95, 2013 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-23897892

RESUMO

DNA double-strand breaks (DSBs) are the most toxic of all genomic insults, and pathways dealing with their signaling and repair are crucial to prevent cancer and for immune system development. Despite intense investigations, our knowledge of these pathways has been technically limited by our inability to detect the main repair factors at DSBs in cells. In this paper, we present an original method that involves a combination of ribonuclease- and detergent-based preextraction with high-resolution microscopy. This method allows direct visualization of previously hidden repair complexes, including the main DSB sensor Ku, at virtually any type of DSB, including those induced by anticancer agents. We demonstrate its broad range of applications by coupling it to laser microirradiation, super-resolution microscopy, and single-molecule counting to investigate the spatial organization and composition of repair factories. Furthermore, we use our method to monitor DNA repair and identify mechanisms of repair pathway choice, and we show its utility in defining cellular sensitivities and resistance mechanisms to anticancer agents.


Assuntos
Antígenos Nucleares/química , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas de Ligação a DNA/química , Microscopia/métodos , Animais , Antígenos Nucleares/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Humanos , Autoantígeno Ku , Camundongos
20.
Nat Chem Biol ; 8(3): 301-10, 2012 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-22306580

RESUMO

Guanine-rich DNA sequences that can adopt non-Watson-Crick structures in vitro are prevalent in the human genome. Whether such structures normally exist in mammalian cells has, however, been the subject of active research for decades. Here we show that the G-quadruplex-interacting drug pyridostatin promotes growth arrest in human cancer cells by inducing replication- and transcription-dependent DNA damage. A chromatin immunoprecipitation sequencing analysis of the DNA damage marker γH2AX provided the genome-wide distribution of pyridostatin-induced sites of damage and revealed that pyridostatin targets gene bodies containing clusters of sequences with a propensity for G-quadruplex formation. As a result, pyridostatin modulated the expression of these genes, including the proto-oncogene SRC. We observed that pyridostatin reduced SRC protein abundance and SRC-dependent cellular motility in human breast cancer cells, validating SRC as a target of this drug. Our unbiased approach to define genomic sites of action for a drug establishes a framework for discovering functional DNA-drug interactions.


Assuntos
Aminoquinolinas/farmacologia , Antineoplásicos/farmacologia , Dano ao DNA , DNA/química , DNA/efeitos dos fármacos , Ácidos Picolínicos/farmacologia , Aminoquinolinas/síntese química , Aminoquinolinas/química , Antineoplásicos/síntese química , Antineoplásicos/química , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , DNA/genética , Ensaios de Seleção de Medicamentos Antitumorais , Quadruplex G/efeitos dos fármacos , Humanos , Peso Molecular , Ácidos Picolínicos/síntese química , Ácidos Picolínicos/química , Proto-Oncogene Mas , Relação Estrutura-Atividade , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA