Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 11(24)2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36552844

RESUMO

Cancer is one of the leading causes of death worldwide, thus the search for new cancer therapies is of utmost importance. Ursolic acid is a naturally occurring pentacyclic triterpene with a wide range of pharmacological activities including anti-inflammatory and anti-neoplastic effects. The latter has been assigned to its ability to promote apoptosis and inhibit cancer cell proliferation by poorly defined mechanisms. In this report, we identify lysosomes as the essential targets of the anti-cancer activity of ursolic acid. The treatment of MCF7 breast cancer cells with ursolic acid elevates lysosomal pH, alters the cellular lipid profile, and causes lysosomal membrane permeabilization and leakage of lysosomal enzymes into the cytosol. Lysosomal membrane permeabilization precedes the essential hallmarks of apoptosis placing it as an initial event in the cascade of effects induced by ursolic acid. The disruption of the lysosomal function impairs the autophagic pathway and likely partakes in the mechanism by which ursolic acid kills cancer cells. Furthermore, we find that combining treatment with ursolic acid and cationic amphiphilic drugs can significantly enhance the degree of lysosomal membrane permeabilization and cell death in breast cancer cells.


Assuntos
Neoplasias da Mama , Lisossomos , Humanos , Feminino , Lisossomos/metabolismo , Homeostase , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Lipídeos/farmacologia , Ácido Ursólico
2.
Nat Commun ; 13(1): 5303, 2022 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-36085345

RESUMO

The RNA world is changing our views about sensing and resolution of DNA damage. Here, we develop single-molecule DNA/RNA analysis approaches to visualize how nascent RNA facilitates the repair of DNA double-strand breaks (DSBs). RNA polymerase II (RNAPII) is crucial for DSB resolution in human cells. DSB-flanking, RNAPII-generated nascent RNA forms RNA:DNA hybrids, guiding the upstream DNA repair steps towards favouring the error-free Homologous Recombination (HR) pathway over Non-Homologous End Joining. Specific RNAPII inhibitor, THZ1, impairs recruitment of essential HR proteins to DSBs, implicating nascent RNA in DNA end resection, initiation and execution of HR repair. We further propose that resection factor CtIP interacts with and helps re-activate RNAPII when paused by the RNA:DNA hybrids, collectively promoting faithful repair of chromosome breaks to maintain genomic integrity.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Reparo do DNA por Junção de Extremidades , Humanos , RNA/genética , RNA Polimerase II , Reparo de DNA por Recombinação
3.
Cell Oncol (Dordr) ; 44(4): 805-820, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33939112

RESUMO

PURPOSE: Most HER2 positive invasive cancers are either intrinsic non-responsive or develop resistance when treated with 1st line HER2 targeting drugs. Both 1st and 2nd line treatments of HER2 positive cancers are aimed at targeting the HER2 receptor directly, thereby strongly limiting the treatment options of HER2/ErbB2 inhibition resistant invasive cancers. METHODS: We used phenotypic high throughput microscopy screening to identify efficient inhibitors of ErbB2-induced invasion using 1st line HER2 inhibitor trastuzumab- and pertuzumab-resistant, p95-ErbB2 expressing breast cancer cells in conjunction with the Prestwick Chemical Library®. The screening entailed a drug's ability to inhibit ErbB2-induced, invasion-promoting positioning of lysosomes at the cellular periphery, a phenotype that defines their invasiveness. In addition, we used high throughput microscopy and biochemical assays to assess the effects of the drugs on lysosomal membrane permeabilization (LMP) and autophagy, two features connected to cancer treatment. Using 2nd line HER2 inhibitor lapatinib resistant 3-dimensional model systems, we assessed the effects of the drugs on ErbB2 positive breast cancer spheroids and developed a high-throughput invasion assay for HER2 positive ovarian cancer organoids for further evaluation. RESULTS: We identified Auranofin, Colchicine, Monensin, Niclosamide, Podophyllotoxin, Quinacrine and Thiostrepton as efficient inhibitors of invasive growth of 2nd line HER2 inhibitor lapatinib resistant breast cancer spheroids and ovarian cancer organoids. We classified these drugs into four groups based on their ability to target lysosomes by inducing autophagy and/or LMP, i.e., drugs inducing early LMP, early autophagy with late LMP, late LMP, or neither. CONCLUSIONS: Our results indicate that targetable lysosome-engaging cellular pathways downstream of ErbB2 contribute to invasion. They support lysosomal trafficking as an attractive target for therapy aiming at preventing the spreading of cancer cells. Since these drugs additionally possess anti-inflammatory activities, they could serve as multipurpose drugs simultaneously targeting infection/inflammation and cancer spreading.


Assuntos
Anti-Inflamatórios/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Receptor ErbB-2/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Antineoplásicos/uso terapêutico , Autofagia/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Lapatinib/uso terapêutico , Lisossomos/metabolismo , Células MCF-7 , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Invasividade Neoplásica
4.
Nat Commun ; 11(1): 4709, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32948765

RESUMO

Glioblastoma cancer-stem like cells (GSCs) display marked resistance to ionizing radiation (IR), a standard of care for glioblastoma patients. Mechanisms underpinning radio-resistance of GSCs remain largely unknown. Chromatin state and the accessibility of DNA lesions to DNA repair machineries are crucial for the maintenance of genomic stability. Understanding the functional impact of chromatin remodeling on DNA repair in GSCs may lay the foundation for advancing the efficacy of radio-sensitizing therapies. Here, we present the results of a high-content siRNA microscopy screen, revealing the transcriptional elongation factor SPT6 to be critical for the genomic stability and self-renewal of GSCs. Mechanistically, SPT6 transcriptionally up-regulates BRCA1 and thereby drives an error-free DNA repair in GSCs. SPT6 loss impairs the self-renewal, genomic stability and tumor initiating capacity of GSCs. Collectively, our results provide mechanistic insights into how SPT6 regulates DNA repair and identify SPT6 as a putative therapeutic target in glioblastoma.


Assuntos
Reparo do DNA , Instabilidade Genômica , Glioblastoma/genética , Células-Tronco Neoplásicas , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Apoptose , Proteína BRCA1 , Neoplasias Encefálicas/genética , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Inativação Gênica , Glioblastoma/patologia , Células HEK293 , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Células-Tronco Neoplásicas/patologia , RNA Interferente Pequeno/genética , Tolerância a Radiação , Radiação Ionizante , Transcriptoma
5.
Nat Commun ; 7: 12364, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27503537

RESUMO

There are two major and alternative pathways to repair DNA double-strand breaks: non-homologous end-joining and homologous recombination. Here we identify and characterize novel factors involved in choosing between these pathways; in this study we took advantage of the SeeSaw Reporter, in which the repair of double-strand breaks by homology-independent or -dependent mechanisms is distinguished by the accumulation of green or red fluorescence, respectively. Using a genome-wide human esiRNA (endoribonuclease-prepared siRNA) library, we isolate genes that control the recombination/end-joining ratio. Here we report that two distinct sets of genes are involved in the control of the balance between NHEJ and HR: those that are required to facilitate recombination and those that favour NHEJ. This last category includes CCAR2/DBC1, which we show inhibits recombination by limiting the initiation and the extent of DNA end resection, thereby acting as an antagonist of CtIP.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Reparo do DNA por Junção de Extremidades , Genoma Humano , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Cromatina/metabolismo , Dano ao DNA , Endodesoxirribonucleases , Redes Reguladoras de Genes , Humanos , Modelos Biológicos , Proteínas Nucleares/metabolismo , Ligação Proteica , Reparo de DNA por Recombinação
6.
Mol Cell ; 51(4): 469-79, 2013 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-23973375

RESUMO

Chromatin remodeling is tightly linked to all DNA-transacting activities. To study chromatin remodeling during DNA repair, we established quantitative fluorescence imaging methods to measure the exchange of histones in chromatin in living cells. We show that particularly H2A and H2B are evicted and replaced at an accelerated pace at sites of UV-induced DNA damage. This accelerated exchange of H2A/H2B is facilitated by SPT16, one of the two subunits of the histone chaperone FACT (facilitates chromatin transcription) but largely independent of its partner SSRP1. Interestingly, SPT16 is targeted to sites of UV light-induced DNA damage-arrested transcription and is required for efficient restart of RNA synthesis upon damage removal. Together, our data uncover an important role for chromatin dynamics at the crossroads of transcription and the UV-induced DNA damage response.


Assuntos
Montagem e Desmontagem da Cromatina/fisiologia , Dano ao DNA/efeitos da radiação , Proteínas de Ligação a DNA/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Histonas/metabolismo , Transcrição Gênica , Fatores de Elongação da Transcrição/metabolismo , Raios Ultravioleta , Western Blotting , Proteínas de Ciclo Celular , Imunoprecipitação da Cromatina , Reagentes de Ligações Cruzadas/farmacologia , Dano ao DNA/genética , Reparo do DNA/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Células HeLa , Proteínas de Grupo de Alta Mobilidade/genética , Histonas/genética , Humanos , Nucleossomos/genética , RNA/genética , RNA/metabolismo , Fatores de Transcrição , Fatores de Elongação da Transcrição/genética
7.
Cell Cycle ; 12(11): 1688-95, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23656789

RESUMO

Genotoxic insults, such as ionizing radiation (IR), cause DNA damage that evokes a multifaceted cellular DNA damage response (DDR). DNA damage signaling events that control protein activity, subcellular localization, DNA binding, protein-protein interactions, etc. rely heavily on time-dependent posttranslational modifications (PTMs). To complement our previous analysis of IR-induced temporal dynamics of nuclear phosphoproteome, we now identify a range of human nuclear proteins that are dynamically regulated by acetylation, and predominantly deacetylation, during IR-induced DDR by using mass spectrometry-based proteomic approaches. Apart from cataloging acetylation sites through SILAC proteomic analyses before IR and at 5 and 60 min after IR exposure of U2OS cells, we report that: (1) key components of the transcriptional machinery, such as EP300 and CREBBP, are dynamically acetylated; (2) that nuclear acetyltransferases themselves are regulated, not on the protein abundance level, but by (de)acetylation; and (3) that the recently reported p53 co-activator and methyltransferase MLL3 is acetylated on five lysines during the DDR. For selected examples, protein immunoprecipitation and immunoblotting were used to assess lysine acetylation status and thereby validate the mass spectrometry data. We thus present evidence that nuclear proteins, including those known to regulate cellular functions via epigenetic modifications of histones, are regulated by (de)acetylation in a timely manner upon cell's exposure to genotoxic insults. Overall, these results present a resource of temporal profiles of a spectrum of protein acetylation sites during DDR and provide further insights into the highly dynamic nature of regulatory PTMs that help orchestrate the maintenance of genome integrity.


Assuntos
Dano ao DNA/efeitos da radiação , Reparo do DNA , Proteínas Nucleares/metabolismo , Radiação Ionizante , Acetilação/efeitos da radiação , Acetiltransferases/metabolismo , Proteína de Ligação a CREB/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Proteína p300 Associada a E1A/metabolismo , Histonas/metabolismo , Humanos , Lisina/metabolismo , Processamento de Proteína Pós-Traducional , Proteômica , Proteína Supressora de Tumor p53/metabolismo
8.
Int J Mol Sci ; 13(10): 13322-37, 2012 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-23202955

RESUMO

Nucleotide excision repair (NER) is an important DNA repair mechanism required for cellular resistance against UV light and toxic chemicals such as those found in tobacco smoke. In living cells, NER efficiently detects and removes DNA lesions within the large nuclear macromolecular complex called chromatin. The condensed nature of chromatin inhibits many DNA metabolizing activities, including NER. In order to promote efficient repair, detection of a lesion not only has to activate the NER pathway but also chromatin remodeling. In general, such remodeling is thought on the one hand to precede NER, thus allowing repair proteins to efficiently access DNA. On the other hand, after completion of the repair, the chromatin must be returned to its previous undamaged state. Chromatin remodeling can refer to three separate but interconnected processes, histone post-translational modifications, insertion of histone variants and histone displacement (including nucleosome sliding). Here we review current knowledge, and speculate about current unknowns, regarding those chromatin remodeling activities that physically displace histones before, during and after NER.


Assuntos
Reparo do DNA , Histonas/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , Dano ao DNA , Chaperonas de Histonas/metabolismo , Humanos
9.
Cell ; 150(4): 697-709, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22884692

RESUMO

Histone ubiquitylation is a prominent response to DNA double-strand breaks (DSBs), but how these modifications are confined to DNA lesions is not understood. Here, we show that TRIP12 and UBR5, two HECT domain ubiquitin E3 ligases, control accumulation of RNF168, a rate-limiting component of a pathway that ubiquitylates histones after DNA breakage. We find that RNF168 can be saturated by increasing amounts of DSBs. Depletion of TRIP12 and UBR5 allows accumulation of RNF168 to supraphysiological levels, followed by massive spreading of ubiquitin conjugates and hyperaccumulation of ubiquitin-regulated genome caretakers such as 53BP1 and BRCA1. Thus, regulatory and proteolytic ubiquitylations are wired in a self-limiting circuit that promotes histone ubiquitylation near the DNA lesions but at the same time counteracts its excessive spreading to undamaged chromosomes. We provide evidence that this mechanism is vital for the homeostasis of ubiquitin-controlled events after DNA breakage and can be subverted during tumorigenesis.


Assuntos
Proteínas de Transporte/metabolismo , Cromatina/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Ubiquitina-Proteína Ligases/metabolismo , Alphapapillomavirus , Linhagem Celular , Linhagem Celular Tumoral , Inativação Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/virologia , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/patologia , Transcrição Gênica , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ubiquitinação
10.
J Cell Biol ; 196(6): 681-8, 2012 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-22431748

RESUMO

The recognition of helix-distorting deoxyribonucleic acid (DNA) lesions by the global genome nucleotide excision repair subpathway is performed by the XPC-RAD23-CEN2 complex. Although it has been established that Rad23 homologs are essential to protect XPC from proteasomal degradation, it is unclear whether RAD23 proteins have a direct role in the recognition of DNA damage. In this paper, we show that the association of XPC with ultraviolet-induced lesions was impaired in the absence of RAD23 proteins. Furthermore, we show that RAD23 proteins rapidly dissociated from XPC upon binding to damaged DNA. Our data suggest that RAD23 proteins facilitate lesion recognition by XPC but do not participate in the downstream DNA repair process.


Assuntos
Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , DNA/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/genética , Humanos , Camundongos
11.
J Cell Biol ; 190(5): 731-40, 2010 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-20805324

RESUMO

In response to ionizing radiation (IR), cells delay cell cycle progression and activate DNA repair. Both processes are vital for genome integrity, but the mechanisms involved in their coordination are not fully understood. In a mass spectrometry screen, we identified the adenosine triphosphate-dependent chromatin-remodeling protein CHD4 (chromodomain helicase DNA-binding protein 4) as a factor that becomes transiently immobilized on chromatin after IR. Knockdown of CHD4 triggers enhanced Cdc25A degradation and p21(Cip1) accumulation, which lead to more pronounced cyclin-dependent kinase inhibition and extended cell cycle delay. At DNA double-strand breaks, depletion of CHD4 disrupts the chromatin response at the level of the RNF168 ubiquitin ligase, which in turn impairs local ubiquitylation and BRCA1 assembly. These cell cycle and chromatin defects are accompanied by elevated spontaneous and IR-induced DNA breakage, reduced efficiency of DNA repair, and decreased clonogenic survival. Thus, CHD4 emerges as a novel genome caretaker and a factor that facilitates both checkpoint signaling and repair events after DNA damage.


Assuntos
Cromatina/metabolismo , Dano ao DNA/fisiologia , Reparo do DNA , Transdução de Sinais/genética , Autoantígenos/genética , Autoantígenos/metabolismo , Ciclo Celular/genética , Linhagem Celular Tumoral , Cromatina/genética , Cromossomos/metabolismo , DNA/genética , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Genes cdc , Humanos , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/farmacologia , Radiação Ionizante , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitinação , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo
12.
DNA Repair (Amst) ; 9(7): 848-55, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20439168

RESUMO

In the last decade, live cell fluorescence microscopy experiments have revolutionized cellular and molecular biology, enabling the localization of proteins within cellular compartments to be analysed and to determine kinetic parameters of enzymatic reactions in living nuclei to be measured. Recently, in vivo DNA labelling by DNA-stains such as DRAQ5, has provided the opportunity to measure kinetic reactions of GFP-fused proteins in targeted areas of the nucleus with different chromatin compaction levels. To verify the suitability of combining DRAQ5-staining with protein dynamic measurements, we have tested the cellular consequences of DRAQ5 DNA intercalation. We show that DRAQ5 intercalation rapidly modifies both the localization and the mobility properties of several DNA-binding proteins such as histones, DNA repair, replication and transcription factors, by stimulating a release of these proteins from their substrate. Most importantly, the effect of DRAQ5 on the mobility of essential cellular enzymes results in a potent inhibition of the corresponding cellular functions. From these observations, we suggest that great caution must be used when interpreting live cell data obtained using DRAQ5.


Assuntos
Antraquinonas/farmacologia , Cromatina/química , Reparo do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/química , DNA/química , Substâncias Intercalantes/química , Antraquinonas/química , Linhagem Celular , Cromatina/metabolismo , DNA/biossíntese , DNA/genética , Replicação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Substâncias Intercalantes/farmacologia , Coloração e Rotulagem/métodos , Estresse Fisiológico , Transcrição Gênica/efeitos dos fármacos
13.
Mol Cell Biol ; 29(24): 6335-40, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19805510

RESUMO

Heterochromatin protein 1 (HP1) family members are versatile proteins involved in transcription, chromatin organization, and replication. Recent findings now have implicated HP1 proteins in the DNA damage response as well. Cell-biological approaches showed that reducing the levels of all three HP1 isoforms enhances DNA repair, possibly due to heterochromatin relaxation. Additionally, HP1 is phosphorylated in response to DNA damage, which was suggested to initiate the DNA damage response. These findings have led to the conclusion that heterochromatic proteins are inhibitory to repair and that their dissociation from heterochromatin may facilitate repair. In contrast with an inhibitory role, a more active role for HP1 in DNA repair also was proposed based on the finding that all HP1 isoforms are recruited to UV-induced lesions, oxidative lesions, and DNA breaks. The loss of HP1 renders nematodes highly sensitive to DNA damage, and mice lacking HP1beta suffer from genomic instability, suggesting that the loss of HP1 is not necessarily beneficial for repair. These findings raise the possibility that HP1 facilitates DNA repair by reorganizing chromatin, which may involve interactions between phosphorylated HP1 and other DNA damage response proteins. Taken together, these studies illustrate an emerging role of HP1 proteins in the response to genotoxic stress.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Dano ao DNA , Heterocromatina/metabolismo , Isoformas de Proteínas/metabolismo , Animais , Linhagem Celular , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/genética , DNA/genética , DNA/metabolismo , DNA/efeitos da radiação , Reparo do DNA , Heterocromatina/genética , Histonas/metabolismo , Humanos , Modelos Genéticos , Estresse Oxidativo , Isoformas de Proteínas/genética , Raios Ultravioleta
14.
J Cell Biol ; 186(5): 655-63, 2009 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-19736316

RESUMO

Homologous recombination (HR) is essential for faithful repair of DNA lesions yet must be kept in check, as unrestrained HR may compromise genome integrity and lead to premature aging or cancer. To limit unscheduled HR, cells possess DNA helicases capable of preventing excessive recombination. In this study, we show that the human Fbh1 (hFbh1) helicase accumulates at sites of DNA damage or replication stress in a manner dependent fully on its helicase activity and partially on its conserved F box. hFbh1 interacted with single-stranded DNA (ssDNA), the formation of which was required for hFbh1 recruitment to DNA lesions. Conversely, depletion of endogenous Fbh1 or ectopic expression of helicase-deficient hFbh1 attenuated ssDNA production after replication block. Although elevated levels of hFbh1 impaired Rad51 recruitment to ssDNA and suppressed HR, its small interfering RNA-mediated depletion increased the levels of chromatin-associated Rad51 and caused unscheduled sister chromatid exchange. Thus, by possessing both pro- and anti-recombinogenic potential, hFbh1 may cooperate with other DNA helicases in tightly controlling cellular HR activity.


Assuntos
DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Genoma Humano , Recombinases/metabolismo , Animais , Linhagem Celular , Cromatina/metabolismo , Dano ao DNA , DNA Helicases/genética , Reparo do DNA , Replicação do DNA , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/genética , Humanos , Interferência de RNA , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Recombinases/genética , Recombinação Genética
15.
J Cell Biol ; 185(4): 577-86, 2009 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-19451271

RESUMO

Heterochromatin protein 1 (HP1) family members are chromatin-associated proteins involved in transcription, replication, and chromatin organization. We show that HP1 isoforms HP1-alpha, HP1-beta, and HP1-gamma are recruited to ultraviolet (UV)-induced DNA damage and double-strand breaks (DSBs) in human cells. This response to DNA damage requires the chromo shadow domain of HP1 and is independent of H3K9 trimethylation and proteins that detect UV damage and DSBs. Loss of HP1 results in high sensitivity to UV light and ionizing radiation in the nematode Caenorhabditis elegans, indicating that HP1 proteins are essential components of DNA damage response (DDR) systems. Analysis of single and double HP1 mutants in nematodes suggests that HP1 homologues have both unique and overlapping functions in the DDR. Our results show that HP1 proteins are important for DNA repair and may function to reorganize chromatin in response to damage.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Dano ao DNA , Animais , Caenorhabditis elegans , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/fisiologia , Quebras de DNA de Cadeia Dupla , Dano ao DNA/efeitos da radiação , Reparo do DNA , Histonas/metabolismo , Mutação , Isoformas de Proteínas , Radiação Ionizante , Raios Ultravioleta/efeitos adversos
16.
J Cell Biol ; 185(1): 21-6, 2009 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-19332890

RESUMO

Live-cell imaging studies aided by mathematical modeling have provided unprecedented insight into assembly mechanisms of multiprotein complexes that control genome function. Such studies have unveiled emerging properties of chromatin-associated systems involved in DNA repair and transcription.


Assuntos
Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Genoma , Proteínas Nucleares/metabolismo , Cromatina/química , Reparo do DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/fisiologia , Cinética , Modelos Biológicos , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/fisiologia , Transcrição Gênica
17.
DNA Repair (Amst) ; 8(6): 767-76, 2009 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-19332393

RESUMO

Although the basic principle of nucleotide excision repair (NER), which can eliminate various DNA lesions, have been dissected at the genetic, biochemical and cellular levels, the important in vivo regulation of the critical damage recognition step is poorly understood. Here we analyze the in vivo dynamics of the essential NER damage recognition factor XPC fused to the green fluorescence protein (GFP). Fluorescence recovery after photobleaching analysis revealed that the UV-induced transient immobilization of XPC, reflecting its actual engagement in NER, is regulated in a biphasic manner depending on the number of (6-4) photoproducts and titrated by the number of functional UV-DDB molecules. A similar biphasic UV-induced immobilization of TFIIH was observed using XPB-GFP. Surprisingly, subsequent integration of XPA into the NER complex appears to follow only the low UV dose immobilization of XPC. Our results indicate that when only a small number of (6-4) photoproducts are generated, the UV-DDB-dependent damage recognition pathway predominates over direct recognition by XPC, and they also suggest the presence of rate-limiting regulatory steps in NER prior to the assembly of XPA.


Assuntos
Dano ao DNA/efeitos da radiação , Reparo do DNA/genética , Proteínas de Ligação a DNA/genética , Fibroblastos/efeitos da radiação , Raios Ultravioleta , Células Cultivadas , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Técnicas Imunoenzimáticas , Cinética , RNA Interferente Pequeno/farmacologia , Fator de Transcrição TFIIH/metabolismo , Proteína de Xeroderma Pigmentoso Grupo A/genética
18.
Methods Mol Biol ; 505: 69-96, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19117140

RESUMO

Quantitative imaging techniques of fluorescently-tagged proteins have been instrumental in the study of the behavior of nuclear receptors (NRs) and coregulators in living cells. Ligand-activated NRs exert their function in transcription regulation by binding to specific response elements in promotor and enhancer sequences of genes. Fluorescence recovery after photobleaching (FRAP) has proven to be a powerful tool to study the mobility of fluorescently-labeled molecules in living cells. Since binding to DNA leads to the immobilization of DNA-interacting proteins like NRs, FRAP is especially useful for determining DNA-binding kinetics of these proteins. The coordinated interaction of NRs with promoters/enhancers and subsequent transcription activation is not only regulated by ligand but also by interactions with sets of cofactors and, at least in the case of the androgen receptor (AR), by dimerization and interdomain interactions. In living cells, these interactions can be studied by fluorescence resonance energy transfer (FRET). Here we provide and discuss detailed protocols for FRAP and FRET procedures to study the behavior of nuclear receptors in living cells. On the basis of our studies of the AR, we provide protocols for two different FRAP methods (strip-FRAP and FLIP-FRAP) to quantitatively investigate DNA-interactions and for two different FRET approaches, ratio imaging, and acceptor photobleaching FRET to study AR domain interactions and interactions with cofactor motifs. Finally, we provide a protocol of a technique where FRAP and acceptor photobleaching FRET are combined to study the dynamics of interacting ARs.


Assuntos
Recuperação de Fluorescência Após Fotodegradação/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , Receptores Citoplasmáticos e Nucleares/metabolismo , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Recuperação de Fluorescência Após Fotodegradação/instrumentação , Transferência Ressonante de Energia de Fluorescência/instrumentação , Humanos , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Modelos Moleculares , Conformação Proteica , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
19.
Epigenetics Chromatin ; 1(1): 9, 2008 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-19014481

RESUMO

The integrity of the genome is continuously challenged by both endogenous and exogenous DNA damaging agents. These damaging agents can induce a wide variety of lesions in the DNA, such as double strand breaks, single strand breaks, oxidative lesions and pyrimidine dimers. The cell has evolved intricate DNA damage response mechanisms to counteract the genotoxic effects of these lesions. The two main features of the DNA damage response mechanisms are cell-cycle checkpoint activation and, at the heart of the response, DNA repair. For both damage signalling and repair, chromatin remodelling is most likely a prerequisite. Here, we discuss current knowledge on chromatin remodelling with respect to the cellular response to DNA damage, with emphasis on the response to lesions resolved by nucleotide excision repair. We will discuss the role of histone modifications as well as their displacement or exchange in nucleotide excision repair and make a comparison with their requirement in transcription and double strand break repair.

20.
J Cell Sci ; 121(Pt 17): 2850-9, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18682493

RESUMO

To investigate how the nucleotide excision repair initiator XPC locates DNA damage in mammalian cell nuclei we analyzed the dynamics of GFP-tagged XPC. Photobleaching experiments showed that XPC constantly associates with and dissociates from chromatin in the absence of DNA damage. DNA-damaging agents retard the mobility of XPC, and UV damage has the most pronounced effect on the mobility of XPC-GFP. XPC exhibited a surprising distinct dynamic behavior and subnuclear distribution compared with other NER factors. Moreover, we uncovered a novel regulatory mechanism for XPC. Under unchallenged conditions, XPC is continuously exported from and imported into the nucleus, which is impeded when NER lesions are present. XPC is omnipresent in the nucleus, allowing a quick response to genotoxic stress. To avoid excessive DNA probing by the low specificity of the protein, the steady-state level in the nucleus is controlled by nucleus-cytoplasm shuttling, allowing temporally higher concentrations of XPC in the nucleus under genotoxic stress conditions.


Assuntos
Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Genoma/genética , Sequência de Aminoácidos , Linhagem Celular , Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Reparo do DNA/efeitos da radiação , Proteínas de Ligação a DNA/química , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Recuperação de Fluorescência Após Fotodegradação , Genoma/efeitos da radiação , Proteínas de Fluorescência Verde/metabolismo , Humanos , Cinética , Modelos Biológicos , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Ligação Proteica/efeitos da radiação , Transporte Proteico/efeitos da radiação , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição TFIIH/metabolismo , Raios Ultravioleta , Proteína de Xeroderma Pigmentoso Grupo A/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...