Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Cell ; 142(4): 519-30, 2010 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-20723754

RESUMO

Entangling and twisting of cellular DNA (i.e., supercoiling) are problems inherent to the helical structure of double-stranded DNA. Supercoiling affects transcription, DNA replication, and chromosomal segregation. Consequently the cell must fine-tune supercoiling to optimize these key processes. Here, we summarize how supercoiling is generated and review experimental and theoretical insights into supercoil relaxation. We distinguish between the passive dissipation of supercoils by diffusion and the active removal of supercoils by topoisomerase enzymes. We also review single-molecule studies that elucidate the timescales and mechanisms of supercoil removal.


Assuntos
DNA Topoisomerases Tipo I/metabolismo , DNA Super-Helicoidal/química , Animais , Fenômenos Fisiológicos Celulares , DNA/química , DNA/metabolismo , DNA Super-Helicoidal/metabolismo , Humanos
2.
Biochemistry ; 58(6): 621-632, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30574775

RESUMO

SUMO, a conserved ubiquitin-like protein, is conjugated to a multitude of cellular proteins to maintain genomic integrity and resist genotoxic stress. Studies of the SUMO E2 conjugating enzyme mutant, UBC9P123L, suggested that altered substrate specificity enhances cell sensitivity to DNA damaging agents. Using nuclear magnetic resonance chemical shift studies, we confirm that the mutation does not alter the core globular fold of UBC9, while 15N relaxation measurements demonstrate mutant-induced stabilization of distinct chemical states in residues near the active site cysteine and substrate recognition motifs. We further demonstrate that the P123L substitution induces a switch from the preferential addition of SUMO to lysine residues in unstructured sites to acceptor lysines embedded in secondary structures, thereby also inducing alterations in SUMO chain linkages. Our results provide new insights regarding the impact that structural dynamics of UBC9 have on substrate selection and specifically SUMO chain formation. These findings highlight the potential contribution of nonconsensus SUMO targets and/or alternative SUMO chain linkages on DNA damage response and chemotherapeutic sensitivity.


Assuntos
Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Domínio Catalítico , Cisteína/química , Humanos , Leucina/química , Leucina/genética , Mutação , Prolina/química , Prolina/genética , Saccharomyces cerevisiae/química , Alinhamento de Sequência , Especificidade por Substrato , Sumoilação , Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/genética
3.
J Biol Chem ; 290(19): 12068-78, 2015 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-25795777

RESUMO

During processes such as DNA replication and transcription, DNA topoisomerase I (Top1) catalyzes the relaxation of DNA supercoils. The nuclear enzyme is also the cellular target of camptothecin (CPT) chemotherapeutics. Top1 contains four domains: the highly conserved core and C-terminal domains involved in catalysis, a coiled-coil linker domain of variable length, and a poorly conserved N-terminal domain. Yeast and human Top1 share a common reaction mechanism and domain structure. However, the human Top1 is ∼100-fold more sensitive to CPT. Moreover, substitutions of a conserved Gly(717) residue, which alter intrinsic enzyme sensitivity to CPT, induce distinct phenotypes in yeast. To address the structural basis for these differences, reciprocal swaps of yeast and human Top1 domains were engineered in chimeric enzymes. Here we report that intrinsic Top1 sensitivity to CPT is dictated by the composition of the conserved core and C-terminal domains. However, independent of CPT, biochemically similar chimeric enzymes produced strikingly distinct phenotypes in yeast. Expression of a human Top1 chimera containing the yeast linker domain proved toxic, even in the context of a catalytically inactive Y723F enzyme. Lethality was suppressed either by splicing the yeast N-terminal domain into the chimera, deleting the human N-terminal residues, or in enzymes reconstituted by polypeptide complementation. These data demonstrate a functional interaction between the N-terminal and linker domains, which, when mispaired between yeast and human enzymes, induces cell lethality. Because toxicity was independent of enzyme catalysis, the inappropriate coordination of N-terminal and linker domains may induce aberrant Top1-protein interactions to impair cell growth.


Assuntos
Camptotecina/química , DNA Topoisomerases Tipo I/química , Saccharomyces cerevisiae/enzimologia , Inibidores da Topoisomerase I/química , Sequência de Aminoácidos , Catálise , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
4.
J Biol Chem ; 290(10): 6203-14, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25609251

RESUMO

Tyrosyl-DNA phosphodiesterase I (Tdp1) catalyzes the repair of 3'-DNA adducts, such as the 3'-phosphotyrosyl linkage of DNA topoisomerase I to DNA. Tdp1 contains two conserved catalytic histidines: a nucleophilic His (His(nuc)) that attacks DNA adducts to form a covalent 3'-phosphohistidyl intermediate and a general acid/base His (His(gab)), which resolves the Tdp1-DNA linkage. A His(nuc) to Ala mutant protein is reportedly inactive, whereas the autosomal recessive neurodegenerative disease SCAN1 has been attributed to the enhanced stability of the Tdp1-DNA intermediate induced by mutation of His(gab) to Arg. However, here we report that expression of the yeast His(nuc)Ala (H182A) mutant actually induced topoisomerase I-dependent cytotoxicity and further enhanced the cytotoxicity of Tdp1 His(gab) mutants, including H432N and the SCAN1-related H432R. Moreover, the His(nuc)Ala mutant was catalytically active in vitro, albeit at levels 85-fold less than that observed with wild type Tdp1. In contrast, the His(nuc)Phe mutant was catalytically inactive and suppressed His(gab) mutant-induced toxicity. These data suggest that the activity of another nucleophile when His(nuc) is replaced with residues containing a small side chain (Ala, Asn, and Gln), but not with a bulky side chain. Indeed, genetic, biochemical, and mass spectrometry analyses show that a highly conserved His, immediately N-terminal to His(nuc), can act as a nucleophile to catalyze the formation of a covalent Tdp1-DNA intermediate. These findings suggest that the flexibility of Tdp1 active site residues may impair the resolution of mutant Tdp1 covalent phosphohistidyl intermediates and provide the rationale for developing chemotherapeutics that stabilize the covalent Tdp1-DNA intermediate.


Assuntos
Adutos de DNA/química , DNA/genética , Proteínas Mutantes/química , Diester Fosfórico Hidrolases/genética , Catálise , Domínio Catalítico/genética , Cristalografia por Raios X , DNA/química , Adutos de DNA/genética , Dano ao DNA/genética , Reparo do DNA/genética , DNA Topoisomerases Tipo I/química , DNA Topoisomerases Tipo I/genética , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Proteínas Mutantes/genética , Diester Fosfórico Hidrolases/química , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia
5.
Nature ; 448(7150): 213-7, 2007 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-17589503

RESUMO

Increasing the ability of chemotherapeutic drugs to kill cancer cells is often hampered by a limited understanding of their mechanism of action. Camptothecins, such as topotecan, induce cell death by poisoning DNA topoisomerase I, an enzyme capable of removing DNA supercoils. Topotecan is thought to stabilize a covalent topoisomerase-DNA complex, rendering it an obstacle to DNA replication forks. Here we use single-molecule nanomanipulation to monitor the dynamics of human topoisomerase I in the presence of topotecan. This allowed us to detect the binding and unbinding of an individual topotecan molecule in real time and to quantify the drug-induced trapping of topoisomerase on DNA. Unexpectedly, our findings also show that topotecan significantly hinders topoisomerase-mediated DNA uncoiling, with a more pronounced effect on the removal of positive (overwound) versus negative supercoils. In vivo experiments in the budding yeast verified the resulting prediction that positive supercoils would accumulate during transcription and replication as a consequence of camptothecin poisoning of topoisomerase I. Positive supercoils, however, were not induced by drug treatment of cells expressing a catalytically active, camptothecin-resistant topoisomerase I mutant. This combination of single-molecule and in vivo data suggests a cytotoxic mechanism for camptothecins, in which the accumulation of positive supercoils ahead of the replication machinery induces potentially lethal DNA lesions.


Assuntos
DNA Topoisomerases Tipo I/metabolismo , DNA Super-Helicoidal/metabolismo , Inibidores Enzimáticos/farmacologia , Inibidores da Topoisomerase I , Topotecan/farmacologia , Humanos , Magnetismo , Nanotecnologia , Saccharomyces cerevisiae/genética
6.
Cancer Cell ; 2(4): 267-73, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12398890

RESUMO

The budding yeast Saccharomyces cerevisiae is a genetically tractable model system with which to establish the cellular target of a given agent and investigate mechanisms of drug action.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Saccharomyces cerevisiae/efeitos dos fármacos , Humanos , Modelos Biológicos , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética
7.
Cancer Cell ; 5(6): 519-23, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15193254

RESUMO

Activation of the phosphatidylinositol 3' kinase-Akt pathway has long been associated with malignant transformation and antiapoptotic signaling. Mutations downstream of Akt that activate the TOR kinase are found in tumor-prone syndromes, while overexpression of translation initiation complex components, such as eIF4E, occurs frequently in human cancer. However, direct roles for TOR signaling or eIF4E overexpression, in the genesis of cancer, have been lacking. Recent papers, including one by in this issue of Cancer Cell, clearly establish that dysregulation of cap-dependent translation confers malignant characteristics and induces cancer by suppressing apoptosis, underscoring the potential of therapeutics that selectively target the Akt-TOR-eIF4E pathway.


Assuntos
Apoptose , Neoplasias/patologia , Biossíntese de Proteínas , Animais , Transformação Celular Neoplásica , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Modelos Biológicos , Mutação , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais
8.
Mol Cell Biol ; 27(20): 7007-17, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17698581

RESUMO

The conserved TOR (target of rapamycin) kinase is part of a TORC1 complex that regulates cellular responses to environmental stress, such as amino acid starvation and hypoxia. Dysregulation of Akt-TOR signaling has also been linked to the genesis of cancer, and thus, this pathway presents potential targets for cancer chemotherapeutics. Here we report that rapamycin-sensitive TORC1 signaling is required for the S-phase progression and viability of yeast cells in response to genotoxic stress. In the presence of the DNA-damaging agent methyl methanesulfonate (MMS), TOR-dependent cell survival required a functional S-phase checkpoint. Rapamycin inhibition of TORC1 signaling suppressed the Rad53 checkpoint-mediated induction of ribonucleotide reductase subunits Rnr1 and Rnr3, thereby abrogating MMS-induced mutagenesis and enhancing cell lethality. Moreover, cells deleted for RNR3 were hypersensitive to rapamycin plus MMS, providing the first demonstration that Rnr3 contributes to the survival of cells exposed to DNA damage. Our findings support a model whereby TORC1 acts as a survival pathway in response to genotoxic stress by maintaining the deoxynucleoside triphosphate pools necessary for error-prone translesion DNA polymerases. Thus, TOR-dependent cell survival in response to DNA-damaging agents coincides with increased mutation rates, which may contribute to the acquisition of chemotherapeutic drug resistance.


Assuntos
Sobrevivência Celular , Dano ao DNA , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais/fisiologia , Adulto , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinase do Ponto de Checagem 2 , Cicloeximida/metabolismo , Humanos , Metanossulfonato de Metila/metabolismo , Complexos Multiproteicos , Mutagênicos/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ribonucleosídeo Difosfato Redutase/genética , Ribonucleosídeo Difosfato Redutase/metabolismo , Fase S/fisiologia , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Sirolimo/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
9.
Diabetes ; 69(9): 2003-2016, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32611548

RESUMO

Targeting retinoid X receptor (RXR) has been proposed as one of the therapeutic strategies to treat individuals with metabolic syndrome, as RXR heterodimerizes with multiple nuclear receptors that regulate genes involved in metabolism. Despite numerous efforts, RXR ligands (rexinoids) have not been approved for clinical trials to treat metabolic syndrome due to the serious side effects such as hypertriglyceridemia and altered thyroid hormone axis. In this study, we demonstrate a novel rexinoid-like small molecule, UAB126, which has positive effects on metabolic syndrome without the known side effects of potent rexinoids. Oral administration of UAB126 ameliorated obesity, insulin resistance, hepatic steatosis, and hyperlipidemia without changes in food intake, physical activity, and thyroid hormone levels. RNA-sequencing analysis revealed that UAB126 regulates the expression of genes in the liver that are modulated by several nuclear receptors, including peroxisome proliferator-activated receptor α and/or liver X receptor in conjunction with RXR. Furthermore, UAB126 not only prevented but also reversed obesity-associated metabolic disorders. The results suggest that optimized modulation of RXR may be a promising strategy to treat metabolic disorders without side effects. Thus, the current study reveals that UAB126 could be an attractive therapy to treat individuals with obesity and its comorbidities.


Assuntos
Dieta Hiperlipídica , Fígado Gorduroso/tratamento farmacológico , Hiperlipidemias/tratamento farmacológico , Resistência à Insulina/fisiologia , Fígado/efeitos dos fármacos , Obesidade/tratamento farmacológico , Receptores X de Retinoides/agonistas , Animais , Fígado Gorduroso/sangue , Hiperlipidemias/sangue , Lipídeos/sangue , Masculino , Camundongos , Obesidade/sangue
10.
Mol Cell Biol ; 26(13): 4958-69, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16782883

RESUMO

Covalent modification with SUMO alters protein function, intracellular localization, or protein-protein interactions. Target recognition is determined, in part, by the SUMO E2 enzyme, Ubc9, while Siz/Pias E3 ligases may facilitate select interactions by acting as substrate adaptors. A yeast conditional Ubc9P(123)L mutant was viable at 36 degrees C yet exhibited enhanced sensitivity to DNA damage. To define functional domains in Ubc9 that dictate cellular responses to genotoxic stress versus those necessary for cell viability, a 1.75-A structure of yeast Ubc9 that demonstrated considerable conservation of backbone architecture with human Ubc9 was solved. Nevertheless, differences in side chain geometry/charge guided the design of human/yeast chimeras, where swapping domains implicated in (i) binding residues within substrates that flank canonical SUMOylation sites, (ii) interactions with the RanBP2 E3 ligase, and (iii) binding of the heterodimeric E1 and SUMO had distinct effects on cell growth and resistance to DNA-damaging agents. Our findings establish a functional interaction between N-terminal and substrate-binding domains of Ubc9 and distinguish the activities of E3 ligases Siz1 and Siz2 in regulating cellular responses to genotoxic stress.


Assuntos
Dano ao DNA , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimologia , Enzimas de Conjugação de Ubiquitina/química , Sequência de Aminoácidos , Dano ao DNA/genética , Humanos , Dados de Sequência Molecular , Estrutura Terciária de Proteína/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidade por Substrato , Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Difração de Raios X
11.
F1000Res ; 82019.
Artigo em Inglês | MEDLINE | ID: mdl-31602296

RESUMO

DNA topoisomerases are enzymes that catalyze changes in the torsional and flexural strain of DNA molecules. Earlier studies implicated these enzymes in a variety of processes in both prokaryotes and eukaryotes, including DNA replication, transcription, recombination, and chromosome segregation. Studies performed over the past 3 years have provided new insight into the roles of various topoisomerases in maintaining eukaryotic chromosome structure and facilitating the decatenation of daughter chromosomes at cell division. In addition, recent studies have demonstrated that the incorporation of ribonucleotides into DNA results in trapping of topoisomerase I (TOP1)-DNA covalent complexes during aborted ribonucleotide removal. Importantly, such trapped TOP1-DNA covalent complexes, formed either during ribonucleotide removal or as a consequence of drug action, activate several repair processes, including processes involving the recently described nuclear proteases SPARTAN and GCNA-1. A variety of new TOP1 inhibitors and formulations, including antibody-drug conjugates and PEGylated complexes, exert their anticancer effects by also trapping these TOP1-DNA covalent complexes. Here we review recent developments and identify further questions raised by these new findings.


Assuntos
DNA Topoisomerases/fisiologia , Neoplasias , DNA , Dano ao DNA , Replicação do DNA , Humanos
12.
J Mol Biol ; 372(4): 1070-1081, 2007 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-17707402

RESUMO

Tyrosyl-DNA phosphodiesterase 1 (Tdp1) catalyzes the resolution of 3' and 5' phospho-DNA adducts. A defective mutant, associated with the recessive neurodegenerative disease SCAN1, accumulates Tdp1-DNA complexes in vitro. To assess the conservation of enzyme architecture, a 2.0 A crystal structure of yeast Tdp1 was determined that is very similar to human Tdp1. Poorly conserved regions of primary structure are peripheral to an essentially identical catalytic core. Enzyme mechanism was also conserved, because the yeast SCAN1 mutant (H(432)R) enhanced cell sensitivity to the DNA topoisomerase I (Top1) poison camptothecin. A more severe Top1-dependent lethality of Tdp1H(432)N was drug-independent, coinciding with increased covalent Top1-DNA and Tdp1-DNA complex formation in vivo. However, both H(432) mutants were recessive to wild-type Tdp1. Thus, yeast H(432) acts in the general acid/base catalytic mechanism of Tdp1 to resolve 3' phosphotyrosyl and 3' phosphoamide linkages. However, the distinct pattern of mutant Tdp1 activity evident in yeast cells, suggests a more severe defect in Tdp1H(432)N-catalyzed resolution of 3' phospho-adducts.


Assuntos
Sítios de Ligação , DNA Topoisomerases Tipo I/metabolismo , Mutação , Diester Fosfórico Hidrolases , Estrutura Terciária de Proteína , Proteínas de Saccharomyces cerevisiae , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Adutos de DNA , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Diester Fosfórico Hidrolases/química , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/metabolismo , Diester Fosfórico Hidrolases/toxicidade , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/toxicidade , Alinhamento de Sequência , Especificidade por Substrato
13.
J Mol Biol ; 369(3): 619-30, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17475278

RESUMO

The SUMO ubiquitin-like proteins play regulatory roles in cell division, transcription, DNA repair, and protein subcellular localization. Paralleling other ubiquitin-like proteins, SUMO proteins are proteolytically processed to maturity, conjugated to targets by E1-E2-E3 cascades, and subsequently recognized by specific downstream effectors containing a SUMO-binding motif (SBM). SUMO and its E2 from the budding yeast Saccharomyces cerevisiae, Smt3p and Ubc9p, are encoded by essential genes. Here we describe the 1.9 A resolution crystal structure of a non-covalent Smt3p-Ubc9p complex. Unexpectedly, a heterologous portion of the crystallized complex derived from the expression construct mimics an SBM, and binds Smt3p in a manner resembling SBM binding to human SUMO family members. In the complex, Smt3p binds a surface distal from Ubc9's catalytic cysteine. The structure implies that a single molecule of Smt3p cannot bind concurrently to both the non-covalent binding site and the catalytic cysteine of a single Ubc9p molecule. However, formation of higher-order complexes can occur, where a single Smt3p covalently linked to one Ubc9p's catalytic cysteine also binds non-covalently to another molecule of Ubc9p. Comparison with other structures from the SUMO pathway suggests that formation of the non-covalent Smt3p-Ubc9p complex occurs mutually exclusively with many other Smt3p and Ubc9p interactions in the conjugation cascade. By contrast, high-resolution insights into how Smt3p-Ubc9p can also interact with downstream recognition machineries come from contacts with the SBM mimic. Interestingly, the overall architecture of the Smt3p-Ubc9p complex is strikingly similar to recent structures from the ubiquitin pathway. The results imply that non-covalent ubiquitin-like protein-E2 complexes are conserved platforms, which function as parts of larger assemblies involved in many protein post-translational regulatory pathways.


Assuntos
Proteínas/química , Proteínas Repressoras/química , Proteínas de Saccharomyces cerevisiae/química , Enzimas de Conjugação de Ubiquitina/química , Ubiquitina/química , Sequência de Aminoácidos , Domínio Catalítico , Clonagem Molecular , Cristalografia por Raios X , Cisteína/química , Humanos , Conformação Molecular , Dados de Sequência Molecular , Ligação Proteica , Saccharomyces cerevisiae/metabolismo , Homologia de Sequência de Aminoácidos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina
15.
DNA Repair (Amst) ; 5(7): 799-809, 2006 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-16713756

RESUMO

The aim of the present study was to identify proteins that bind nicked DNA intermediates formed in the course of base excision repair (BER) in cell free extracts of Saccharomyces cerevisiae. In mammalian cells, nicks in DNA are targets of proteins such as PARP-1 or XRCC1 that have no homologues in yeast. One of the most promising methodologies to trap proteins that interact with damaged DNA lies in using a photocrosslinking technique with photoactivable dNTP analogues such as exo-N-{2-[N-(4-azido-2,5-difluoro-3-chloropyridine-6-yl)-3-aminopropionyl]-aminoethyl}-2'-deoxycytidine-5'-triphosphate (FAP-dCTP) for enzymatic synthesis of DNA probes with a photoreactive dNMP residue at the 3'-margin of a nick. Using this approach, we identified a major covalent DNA-protein adduct between a nick-containing 34-mer DNA duplex and a protein of a molecular mass of around 100-kDa. Unexpectedly, the formation of the 100-kDa adduct did not require the incorporation of the photoreactive dNMP residue at the 3'-margin of the nick nor exposure to near UV-light. However, the formation of the 100-kDa adduct strictly required a nick or a short gap in the DNA probe. Furthermore, the 100-kDa adduct was not detected in yeast extracts lacking DNA topoisomerase I (Top1). To further establish the nature of crosslinked protein, yeast Top1 was tagged with a Myc-epitope. In this case, the mobility of the Top1-DNA adduct increased by 7- kDa. Therefore, our data speak in favor of Top1 trapping by nicked DNA. In support of this hypothesis, purified yeast Top1 was also crosslinked to nicked DNA structures. Undamaged, uracil- and abasic (AP) site-containing DNAs were unable to trap Top1 under the same assay conditions. Since nicked DNA structures are frequently formed in the course of BER, their covalent linkage to Top1 has the potential to interfere with BER in vivo.


Assuntos
DNA Topoisomerases Tipo I/metabolismo , DNA Fúngico/metabolismo , Saccharomyces cerevisiae/metabolismo , Marcadores de Afinidade , Sequência de Bases , Sistema Livre de Células , Reagentes de Ligações Cruzadas , Dano ao DNA , DNA Polimerase beta/metabolismo , Reparo do DNA , DNA Fúngico/química , DNA Fúngico/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidade por Substrato
16.
Cancer Chemother Pharmacol ; 80(1): 1-14, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28528358

RESUMO

To resolve the topological problems that threaten the function and structural integrity of nuclear and mitochondrial genomes and RNA molecules, human cells encode six different DNA topoisomerases including type IB enzymes (TOP1 and TOP1mt), type IIA enzymes (TOP2α and TOP2ß) and type IA enzymes (TOP3α and TOP3ß). DNA entanglements and the supercoiling of DNA molecules are regulated by topoisomerases through the introduction of transient enzyme-linked DNA breaks. The covalent topoisomerase-DNA complexes are the cellular targets of a diverse group of cancer chemotherapeutics, which reversibly stabilize these reaction intermediates. Here we review the structure-function and catalytic mechanisms of each family of eukaryotic DNA topoisomerases and the topoisomerase-targeting agents currently approved for patient therapy or in clinical trials, and highlight novel developments and challenges in the clinical development of these agents.


Assuntos
Antineoplásicos/farmacologia , DNA Topoisomerases/metabolismo , Neoplasias/tratamento farmacológico , Animais , Quebras de DNA , Desenho de Fármacos , Humanos , Mitocôndrias/enzimologia , Mitocôndrias/genética , Terapia de Alvo Molecular , Neoplasias/enzimologia
17.
J Mol Biol ; 428(24 Pt B): 4905-4916, 2016 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-27825925

RESUMO

Saccharomyces cerevisiae sir2Δ or top1Δ mutants exhibit similar phenotypes involving ribosomal DNA, including (i) loss of transcriptional silencing, resulting in non-coding RNA hyperproduction from cryptic RNA polymerase II promoters; (ii) alterations in recombination; and (iii) a general increase in histone acetylation. Given the distinct enzymatic activities of Sir2 and Top1 proteins, a histone deacetylase and a DNA topoisomerase, respectively, we investigated whether genetic and/or physical interactions between the two proteins could explain the shared ribosomal RNA genes (rDNA) phenotypes. We employed an approach of complementing top1Δ cells with yeast, human, truncated, and chimeric yeast/human TOP1 constructs and of assessing the extent of non-coding RNA silencing and histone H4K16 deacetylation. Our findings demonstrate that residues 115-125 within the yeast Top1p N-terminal domain are required for the complementation of the top1∆ rDNA phenotypes. In chromatin immunoprecipitation and co-immunoprecipitation experiments, we further demonstrate the physical interaction between Top1p and Sir2p. Our genetic and biochemical studies support a model whereby Top1p recruits Sir2p to the rDNA and clarifies a structural role of DNA topoisomerase I in the epigenetic regulation of rDNA, independent of its known catalytic activity.


Assuntos
DNA Topoisomerases Tipo I/metabolismo , DNA Ribossômico/metabolismo , Regulação Fúngica da Expressão Gênica , RNA Ribossômico/biossíntese , Saccharomyces cerevisiae/metabolismo , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/metabolismo , Sirtuína 2/metabolismo , Imunoprecipitação da Cromatina , DNA Topoisomerases Tipo I/genética , Deleção de Genes , Teste de Complementação Genética , Ligação Proteica , Saccharomyces cerevisiae/genética , Transcrição Gênica
18.
J Lab Autom ; 21(1): 198-203, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26663785

RESUMO

The development of acoustic droplet ejection (ADE) technology has resulted in many positive changes associated with the operations in a high-throughput screening (HTS) laboratory. Originally, this liquid transfer technology was used to simply transfer DMSO solutions of primarily compounds. With the introduction of Labcyte's Echo 555, which has aqueous dispense capability, the application of this technology has been expanded beyond its original use. This includes the transfer of many biological reagents solubilized in aqueous buffers, including siRNAs. The Echo 555 is ideal for siRNA dispensing because it is accurate at low volumes and a step-down dilution is not necessary. The potential for liquid carryover and cross-contamination is eliminated, as no tips are needed. Herein, we describe the siRNA screening platform at Southern Research's HTS Center using the ADE technology. With this technology, an siRNA library can be dispensed weeks or even months in advance of the assay itself. The protocol has been optimized to achieve assay parameters comparable to small-molecule screening parameters, and exceeding the norm reported for genomewide siRNA screens.


Assuntos
Tecnologia Biomédica/métodos , Estudos de Associação Genética/métodos , Ensaios de Triagem em Larga Escala/métodos , Interferência de RNA , Acústica , Soluções
19.
Cancer Chemother Pharmacol ; 55(5): 411-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15503024

RESUMO

PURPOSE: Irofulven, a novel chemotherapeutic agent with a broad spectrum of activity, is effective against preclinical models of pediatric tumors. The cytotoxic activity of irofulven is augmented when combined with agents that interact with DNA topoisomerase I; however, none of the reported studies have used the protracted dosing schedule found to be active clinically in treatment of childhood cancers. The objective of this study was to evaluate the antitumor activity of irofulven in combination with irinotecan administered on a protracted schedule in a panel of pediatric solid tumor xenografts. METHODS: Irofulven and irinotecan were evaluated alone or in combination against eight independent xenografts, which included childhood brain tumors (n=5), neuroblastoma (n=1), and rhabdomyosarcoma (n=2). Irofulven was administered i.v. daily for 5 days with courses repeated every 21 days for a total of three cycles. Doses of irofulven ranged from 1.33 to 4.6 mg/kg. Irinotecan was given i.v. daily for 5 days each week for 2 weeks repeated every 21 days for three cycles at doses between 0.28 and 1.25 mg/kg. RESULTS: Irofulven and irinotecan, given as single agents, induced few responses in pediatric solid tumor xenografts at the selected doses. At the same doses, irofulven in combination with irinotecan demonstrated superior antitumor activity, inducing complete responses in seven of the eight xenograft lines. CONCLUSIONS: These studies show that the cytotoxic activity of irofulven is greater when combined with protracted administration of irinotecan. Although the systemic exposure of irofulven required to induce objective responses in this panel of pediatric solid tumors was in excess of that achievable in patients receiving maximally tolerated doses using this schedule of drug administration, the enhanced activity of irofulven in combination with irinotecan supports the pursuit of alternative administration strategies and combinations.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Camptotecina/análogos & derivados , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Camptotecina/administração & dosagem , Esquema de Medicação , Feminino , Irinotecano , Camundongos , Camundongos Endogâmicos ICR , Sesquiterpenos/administração & dosagem , Falha de Tratamento
20.
Mol Cancer Ther ; 3(4): 393-402, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15078982

RESUMO

Phase I and II clinical trails are currently investigating the antitumor activity of cisplatin and camptothecins (CPTs; DNA topoisomerase I poisons), based on the dramatic synergistic cytotoxicity of these agents in some preclinical models. However, the mechanistic basis for this synergism is poorly understood. By exploiting the evolutionary conservation of DNA repair pathways from genetically tractable organisms such as budding and fission yeasts to mammalian cells, we demonstrate that the synergism of CPT and cisplatin requires homologous recombination. In yeast and mammalian cell lines defective for RAD52 and XRCC2/3, respectively, the combination of these agents proved antagonistic, while greater than additive activity was evident in isogenic wild-type cells. Homologous recombination appears to mediate a similar interaction of X-rays and CPT, but antagonizes the synergism of cytarabine (Ara-C) with CPT. These findings suggest that homologous recombination comprises an evolutionarily conserved determinant of cellular sensitivity when CPTs are used in combination with other therapeutics.


Assuntos
Camptotecina/toxicidade , Cisplatino/toxicidade , Inibidores Enzimáticos/toxicidade , Recombinação Genética/efeitos dos fármacos , Inibidores da Topoisomerase I , Animais , Células CHO , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Cricetinae , Cricetulus , Citarabina/farmacologia , Dano ao DNA/efeitos dos fármacos , Reparo do DNA , DNA Topoisomerases Tipo I/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Sinergismo Farmacológico , Proteína Rad52 de Recombinação e Reparo de DNA , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efeitos da radiação , Proteínas de Saccharomyces cerevisiae , Schizosaccharomyces/efeitos dos fármacos , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Schizosaccharomyces/efeitos da radiação , Raios X
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa