Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
RSC Med Chem ; 12(11): 1935-1943, 2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-34820624

RESUMEN

Ubiquitin carboxy-terminal hydrolase L1 (UCHL1), a deubiquitinating enzyme (DUB), is a potential drug target in various cancers, and liver and lung fibrosis. However, bona fide functions and substrates of UCHL1 remain poorly understood. Herein, we report the characterization of UCHL1 covalent inhibitor MT16-001 based on a thiazole cyanopyrrolidine scaffold. In combination with chemical proteomics, a closely related activity-based probe (MT16-205) was used to generate a comprehensive quantitative profile for on- and off-targets at endogenous cellular abundance. Both compounds are selective for UCHL1 over other DUBs in intact cells but also engage a range of other targets with good selectivity over the wider proteome, including aldehyde dehydrogenases, redox-sensitive Parkinson's disease related protein PARK7, and glutamine amidotransferase. Taken together, these results underline the importance of robust profiling of activity-based probes as chemical tools and highlight the cyanopyrrolidine warhead as a versatile platform for liganding diverse classes of protein with reactive cysteine residues which can be used for further inhibitor screening, and as a starting point for inhibitor development.

3.
J Am Chem Soc ; 142(28): 12020-12026, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32579346

RESUMEN

Ubiquitin carboxy-terminal hydrolase L1 (UCHL1) is a deubiquitylating enzyme that is proposed as a potential therapeutic target in neurodegeneration, cancer, and liver and lung fibrosis. Herein we report the discovery of the most potent and selective UCHL1 probe (IMP-1710) to date based on a covalent inhibitor scaffold and apply this probe to identify and quantify target proteins in intact human cells. IMP-1710 stereoselectively labels the catalytic cysteine of UCHL1 at low nanomolar concentration in cells. We further demonstrate that potent and selective UCHL1 inhibitors block pro-fibrotic responses in a cellular model of idiopathic pulmonary fibrosis, supporting the potential of UCHL1 as a potential therapeutic target in fibrotic diseases.


Asunto(s)
Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Células HeLa , Humanos , Estructura Molecular , Ubiquitina Tiolesterasa/metabolismo
4.
Nat Rev Drug Discov ; 17(1): 57-78, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28959952

RESUMEN

More than a decade after a Nobel Prize was awarded for the discovery of the ubiquitin-proteasome system and clinical approval of proteasome and ubiquitin E3 ligase inhibitors, first-generation deubiquitylating enzyme (DUB) inhibitors are now approaching clinical trials. However, although our knowledge of the physiological and pathophysiological roles of DUBs has evolved tremendously, the clinical development of selective DUB inhibitors has been challenging. In this Review, we discuss these issues and highlight recent advances in our understanding of DUB enzymology and biology as well as technological improvements that have contributed to the current interest in DUBs as therapeutic targets in diseases ranging from oncology to neurodegeneration.


Asunto(s)
Enzimas Desubicuitinizantes/antagonistas & inhibidores , Descubrimiento de Drogas/métodos , Ubiquitina/metabolismo , Descubrimiento de Drogas/tendencias , Industria Farmacéutica , Drogas en Investigación/uso terapéutico , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo
5.
ACS Chem Biol ; 11(12): 3268-3272, 2016 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-27779380

RESUMEN

Deubiquitinating enzymes play an important role in a plethora of therapeutically relevant processes and are emerging as pioneering drug targets. Herein, we present a novel probe, Ubiquitin Specific Protease (USP) inhibitor, alongside an alkyne-tagged activity-based probe analogue. Activity-based proteome profiling identified 12 USPs, including USP4, USP16, and USP33, as inhibitor targets using submicromolar probe concentrations. This represents the first intact cell activity-based profiling of deubiquitinating enzymes. Further analysis demonstrated functional inhibition of USP33 and identified a synergistic relationship in combination with ATR inhibition, consistent with USP4 inhibition.


Asunto(s)
Sondas Moleculares/química , Neoplasias/enzimología , Proteómica/métodos , Pirroles/química , Bibliotecas de Moléculas Pequeñas/química , Proteasas Ubiquitina-Específicas/análisis , Alquinos/química , Línea Celular Tumoral , Humanos , Técnicas de Sonda Molecular , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores
6.
J Cell Biol ; 193(1): 97-108, 2011 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-21444690

RESUMEN

Chromosomal deletions and rearrangements in tumors are often associated with common fragile sites, which are specific genomic loci prone to gaps and breaks in metaphase chromosomes. Common fragile sites appear to arise through incomplete DNA replication because they are induced after partial replication inhibition by agents such as aphidicolin. Here, we show that in G1 cells, large nuclear bodies arise that contain p53 binding protein 1 (53BP1), phosphorylated H2AX (γH2AX), and mediator of DNA damage checkpoint 1 (MDC1), as well as components of previously characterized OPT (Oct-1, PTF, transcription) domains. Notably, we find that incubating cells with low aphidicolin doses increases the incidence and number of 53BP1-OPT domains in G1 cells, and by chromatin immunoprecipitation and massively parallel sequencing analysis of γH2AX, we demonstrate that OPT domains are enriched at common fragile sites. These findings invoke a model wherein incomplete DNA synthesis during S phase leads to a DNA damage response and formation of 53BP1-OPT domains in the subsequent G1.


Asunto(s)
Replicación del ADN/genética , Fase G1/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Factor 1 de Transcripción de Unión a Octámeros/genética , Factores de Transcripción/genética , Transcripción Genética/genética , Células Cultivadas , Humanos , Proteína 1 de Unión al Supresor Tumoral P53
7.
Nucleic Acids Res ; 36(15): 4975-87, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18658245

RESUMEN

Werner syndrome (WS) is a premature aging disorder caused by mutations in the WS gene (WRN). Although WRN has been suggested to play an important role in DNA metabolic pathways, such as recombination, replication and repair, its precise role still remains to be determined. WRN possesses ATPase, helicase and exonuclease activities. Previous studies have shown that the WRN exonuclease is inhibited in vitro by certain lesions induced by oxidative stress and positioned in the digested strand of the substrate. The presence of the 70/86 Ku heterodimer (Ku), participating in the repair of double-strand breaks (DSBs), alleviates WRN exonuclease blockage imposed by the oxidatively induced DNA lesions. The current study demonstrates that WRN exonuclease is inhibited by several additional oxidized bases, and that Ku stimulates the WRN exonuclease to bypass these lesions. Specific lesions present in the non-digested strand were shown also to inhibit the progression of the WRN exonuclease; however, Ku was not able to stimulate WRN exonuclease to bypass these lesions. Thus, this study considerably broadens the spectrum of lesions which block WRN exonuclease progression, shows a blocking effect of lesions in the non-digested strand, and supports a function for WRN and Ku in a DNA damage processing pathway.


Asunto(s)
Daño del ADN , Exodesoxirribonucleasas/metabolismo , Estrés Oxidativo , RecQ Helicasas/metabolismo , Antígenos Nucleares/metabolismo , Citosina/análogos & derivados , Citosina/metabolismo , ADN/química , ADN/metabolismo , Reparación del ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Proteínas de Unión al ADN/metabolismo , Exodesoxirribonucleasas/antagonistas & inhibidores , Humanos , Autoantígeno Ku , RecQ Helicasas/antagonistas & inhibidores , Uracilo/análogos & derivados , Uracilo/metabolismo , Helicasa del Síndrome de Werner
8.
J Biol Chem ; 282(50): 36403-11, 2007 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-17911100

RESUMEN

Metal-catalyzed oxidation reactions target amino acids in the metal binding pocket of proteins. Such oxidation reactions generally result in either preferential degradation of the protein or accumulation of a catalytically inactive pool of protein with age. Consistently, levels of oxidized proteins have been shown to increase with age. The segmental, progeroid disorder Werner syndrome results from loss of the Werner syndrome protein (WRN). WRN is a member of the RecQ family of DNA helicases and possesses exonuclease and ATP-dependent helicase activities. Furthermore, each of the helicase and exonuclease domains of WRN contains a metal binding pocket. In this report we examined for metal-catalyzed oxidation of WRN in the presence of iron or copper. We found that WRN was oxidized in vitro by iron but not by copper. Iron-mediated oxidation resulted in the inhibition of both WRN helicase and exonuclease activities. Oxidation of WRN also inhibited binding to several known protein partners. In addition, we did not observe degradation of oxidized WRN by the 20 S proteasome in vitro. Finally, exposure of cells to hydrogen peroxide resulted in oxidation of WRN in vivo. Therefore, our results demonstrate that WRN undergoes metal-catalyzed oxidation in the presence of iron, and iron-mediated oxidation of WRN likely results in the accumulation of a catalytically inactive form of the protein, which may contribute to age-related phenotypes.


Asunto(s)
Hierro/metabolismo , RecQ Helicasas/metabolismo , Síndrome de Werner/enzimología , Sitios de Unión/fisiología , Catálisis , Cobre/química , Cobre/metabolismo , Exodesoxirribonucleasas , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , Hierro/química , Oxidantes/farmacología , Oxidación-Reducción/efectos de los fármacos , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo , Estructura Terciaria de Proteína/fisiología , RecQ Helicasas/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Helicasa del Síndrome de Werner
9.
J Biol Chem ; 282(36): 26591-602, 2007 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-17611195

RESUMEN

The mammalian DNA glycosylase, NEIL1, specific for repair of oxidatively damaged bases in the genome via the base excision repair pathway, is activated by reactive oxygen species and prevents toxicity due to radiation. We show here that the Werner syndrome protein (WRN), a member of the RecQ family of DNA helicases, associates with NEIL1 in the early damage-sensing step of base excision repair. WRN stimulates NEIL1 in excision of oxidative lesions from bubble DNA substrates. The binary interaction between NEIL1 and WRN (K(D) = 60 nM) involves C-terminal residues 288-349 of NEIL1 and the RecQ C-terminal (RQC) region of WRN, and is independent of the helicase activity WRN. Exposure to oxidative stress enhances the NEIL-WRN association concomitant with their strong nuclear co-localization. WRN-depleted cells accumulate some prototypical oxidized bases (e.g. 8-oxoguanine, FapyG, and FapyA) indicating a physiological function of WRN in oxidative damage repair in mammalian genomes. Interestingly, WRN deficiency does not have an additive effect on in vivo damage accumulation in NEIL1 knockdown cells suggesting that WRN participates in the same repair pathway as NEIL1.


Asunto(s)
Daño del ADN/fisiología , ADN Glicosilasas/metabolismo , Reparación del ADN/fisiología , Genoma Humano/fisiología , Estrés Oxidativo/fisiología , RecQ Helicasas/metabolismo , Línea Celular Tumoral , Daño del ADN/efectos de la radiación , ADN Glicosilasas/química , ADN Glicosilasas/genética , Reparación del ADN/efectos de la radiación , Exodesoxirribonucleasas , Humanos , Oxidación-Reducción/efectos de la radiación , Estrés Oxidativo/efectos de la radiación , Unión Proteica/fisiología , Unión Proteica/efectos de la radiación , Estructura Terciaria de Proteína/fisiología , Pirimidinas/química , Pirimidinas/metabolismo , Especies Reactivas de Oxígeno/química , Especies Reactivas de Oxígeno/metabolismo , RecQ Helicasas/química , RecQ Helicasas/genética , Especificidad por Sustrato/fisiología , Especificidad por Sustrato/efectos de la radiación , Helicasa del Síndrome de Werner
10.
Mech Ageing Dev ; 128(3): 259-66, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17224176

RESUMEN

Reactive oxygen species, generated either by cellular respiration or upon exposure to environmental agents such as ionizing radiation (IR), attack DNA to form a variety of oxidized base and sugar modifications. Accumulation of oxidative DNA damage has been associated with age-related disease as well as the aging process. Single-strand breaks harboring oxidative 3' obstructive termini, e.g. 3' phosphates and 3' phosphoglycolates, must be removed prior to DNA repair synthesis or ligation. In addition, 3' tyrosyl-linked protein damage, resulting from therapeutic agents such as camptothecin (CPT), must be processed to initiate repair. Several nucleases participate in DNA repair and the excision of 3' obstructive ends. As the protein defective in the segmental progeroid Werner syndrome (WRN) possesses 3'-5' exonuclease activity, and Werner syndrome cells are hypersensitive to IR and CPT, we examined for WRN exonuclease activity on 3' blocking lesions. Moreover, we compared side-by-side the activity of four prominent human 3'-5' exonucleases (WRN, APE1, TREX1, and p53) on substrates containing 3' phosphates, phosphoglycolates, and tyrosyl residues. Our studies reveal that while WRN degrades 3' hydroxyl containing substrates in a non-processive manner, it does not excise 3' phosphate, phosphoglycolate, or tyrosyl groups. In addition, we found that APE1 was most active at excising 3' blocking termini in comparison to the disease-related exonucleases TREX1, WRN, and p53 under identical physiological reaction conditions, and that TREX1 was the most powerful 3'-5' exonuclease on undamaged oligonucleotide substrates.


Asunto(s)
ADN/química , ADN/farmacología , RecQ Helicasas/antagonistas & inhibidores , Células Cultivadas , Reparación del ADN/fisiología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/antagonistas & inhibidores , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Exodesoxirribonucleasas , Genes p53/genética , Glicolatos/farmacología , Humanos , Oligonucleótidos/farmacología , Oxidación-Reducción , Proteínas/antagonistas & inhibidores , Proteínas/genética , Especies Reactivas de Oxígeno , RecQ Helicasas/genética , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Tirosina/química , Helicasa del Síndrome de Werner
11.
Nucleic Acids Res ; 34(2): 745-54, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16449207

RESUMEN

Genome instability is a characteristic of cancer and aging, and is a hallmark of the premature aging disorder Werner syndrome (WS). Evidence suggests that the Werner syndrome protein (WRN) contributes to the maintenance of genome integrity through its involvement in DNA repair. In particular, biochemical evidence indicates a role for WRN in base excision repair (BER). We have previously reported that WRN helicase activity stimulates DNA polymerase beta (pol beta) strand displacement synthesis in vitro. In this report we demonstrate that WRN exonuclease activity can act cooperatively with pol beta, a polymerase lacking 3'-5' proofreading activity. Furthermore, using small interference RNA technology, we demonstrate that WRN knockdown cells are hypersensitive to the alkylating agent methyl methanesulfonate, which creates DNA damage that is primarily repaired by the BER pathway. In addition, repair assays using whole cell extracts from WRN knockdown cells indicate a defect in long patch (LP) BER. These findings demonstrate that WRN plays a direct role in the repair of methylation-induced DNA damage, and suggest a role for both WRN helicase and exonuclease activities together with pol beta during LP BER.


Asunto(s)
ADN Helicasas/fisiología , ADN Polimerasa beta/metabolismo , Reparación del ADN , Exodesoxirribonucleasas/fisiología , Alquilantes/toxicidad , Disparidad de Par Base , Línea Celular , Daño del ADN , ADN Helicasas/antagonistas & inhibidores , Exodesoxirribonucleasas/antagonistas & inhibidores , Humanos , Metilmetanosulfonato/toxicidad , Interferencia de ARN , RecQ Helicasas , Helicasa del Síndrome de Werner
12.
Toxicol In Vitro ; 20(4): 426-38, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16198082

RESUMEN

Cytochrome P-450s (CYPs) detoxify a wide variety of xenobiotics and environmental contaminants, but can also bioactivate carcinogenic polycyclic aromatic hydrocarbons, such as benzo(a)pyrene (BaP), to DNA-reactive species. The primary CYPs involved in the metabolism and bioactivation of BaP are CYP1A1 and CYP1B1. Furthermore, BaP can induce expression of CYP1A1 and CYP1B1 via the aryl hydrocarbon receptor. Induction of CYP1A1 and CYP1B1 by BaP in target (lung) and non-target (liver) tissues was investigated utilizing precision-cut rat liver and lung slices exposed to BaP in vitro. Tissue slices were also prepared from rats pretreated in vivo with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) to induce expression of CYP1A1 and CYP1B1. In addition, in vivo exposure studies were performed with BaP to characterize and validate the use of the in vitro tissue slice model. In vitro exposure of liver and lung slices to BaP resulted in a concentration-dependent increase in CYP1A1 and CYP1B1 mRNA and protein levels, which correlated directly with the exposure-related increase in BaP-DNA adduct levels observed previously in the tissue slices [Harrigan, J.A., Vezina, C.M., McGarrigle, B.P., Ersing, N., Box, H.C., Maccubbin, A.E., Olson, J.R., 2004. DNA adduct formation in precision-cut rat liver and lung slices exposed to benzo(a)pyrene. Toxicological Sciences 77, 307-314]. Pretreatment of animals in vivo with TCDD produced a marked induction of CYP1A1 and CYP1B1 expression in the tissue slices, which was similar to the levels of CYP1A1 and CYP1B1 mRNA achieved in liver and lung following in vivo treatment with BaP. Following in vitro exposure to BaP, the levels of CYP1A1 were greater in the lung than the liver, while following all exposures (in vitro and in vivo), the levels of CYP1B1 mRNA were greater in lung tissue compared to liver. The higher expression of CYP1A1 and CYP1B1 in the lung was associated with higher levels of BaP-DNA adducts in the lung slices (Harrigan et al.'s work) and together, these results may contribute to the tissue specificity of BaP-mediated carcinogenesis.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/biosíntesis , Benzo(a)pireno/toxicidad , Carcinógenos/toxicidad , Citocromo P-450 CYP1A1/biosíntesis , Hígado/efectos de los fármacos , Pulmón/efectos de los fármacos , Animales , Hidrocarburo de Aril Hidroxilasas/genética , Biotransformación , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1B1 , Relación Dosis-Respuesta a Droga , Inducción Enzimática , Expresión Génica/efectos de los fármacos , Técnicas In Vitro , Inyecciones Intraperitoneales , Hígado/enzimología , Hígado/patología , Pulmón/enzimología , Pulmón/patología , Masculino , Dibenzodioxinas Policloradas/toxicidad , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
13.
Mol Cell Biol ; 25(17): 7625-36, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16107709

RESUMEN

Cockayne syndrome (CS) is a rare genetic disorder characterized as a segmental premature-aging syndrome. The CS group B (CSB) protein has previously been implicated in transcription-coupled repair, transcriptional elongation, and restoration of RNA synthesis after DNA damage. Recently, evidence for a role of CSB in base excision repair of oxidative DNA lesions has accumulated. In our search to understand the molecular function of CSB in this process, we identify a physical and functional interaction between CSB and poly(ADP-ribose) polymerase-1 (PARP-1). PARP-1 is a nuclear enzyme that protects the integrity of the genome by responding to oxidative DNA damage and facilitating DNA repair. PARP-1 binds to single-strand DNA breaks which activate the catalytic ability of PARP-1 to add polymers of ADP-ribose to various proteins. We find that CSB is present at sites of activated PARP-1 after oxidative stress, identify CSB as a new substrate of PARP-1, and demonstrate that poly(ADP-ribosyl)ation of CSB inhibits its DNA-dependent ATPase activity. Furthermore, we find that CSB-deficient cell lines are hypersensitive to inhibition of PARP. Our results implicate CSB in the PARP-1 poly(ADP-ribosyl)ation response after oxidative stress and thus suggest a novel role of CSB in the cellular response to oxidative damage.


Asunto(s)
Síndrome de Cockayne , ADN Helicasas/metabolismo , Estrés Oxidativo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Transporte Activo de Núcleo Celular , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , ADN Helicasas/deficiencia , ADN Helicasas/genética , Enzimas Reparadoras del ADN , Humanos , Mutación/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/genética , Proteínas de Unión a Poli-ADP-Ribosa , Unión Proteica , Especificidad por Sustrato
14.
J Biol Chem ; 279(51): 53465-74, 2004 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-15385537

RESUMEN

Werner syndrome patients are deficient in the Werner protein (WRN), which is a multifunctional nuclear protein possessing 3'-5' exonuclease and ATP-dependent helicase activities. Studies of Werner syndrome cells and biochemical studies of WRN suggest that WRN plays a role in several DNA metabolic pathways. WRN interacts with DNA polymerase beta (pol beta) and stimulates pol beta strand displacement synthesis on a base excision repair (BER) intermediate in a helicase-dependent manner. In this report, we examined the effect of the major human apurinic/apyrimidinic endonuclease (APE1) and of pol beta on WRN helicase activity. The results show that WRN alone is able to unwind several single strand break BER intermediates. However, APE1 inhibits WRN helicase activity on these intermediates. This inhibition is likely due to the binding of APE1 to nicked apurinic/apyrimidinic sites, suggesting that APE1 prevents the promiscuous unwinding of BER intermediates. This inhibitory effect was relieved by the presence of pol beta. A model involving the pol beta-mediated hand-off of WRN protein is proposed based on these results.


Asunto(s)
Daño del ADN , ADN Helicasas/metabolismo , Reparación del ADN , Regulación Enzimológica de la Expresión Génica , Adenosina Trifosfato/metabolismo , ADN/metabolismo , ADN Polimerasa beta/química , ADN Polimerasa beta/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Exodesoxirribonucleasas , Glutatión Transferasa/metabolismo , Células HeLa , Humanos , Immunoblotting , Microscopía Fluorescente , Modelos Genéticos , Mutación , Desnaturalización de Ácido Nucleico , Unión Proteica , RecQ Helicasas , Síndrome de Werner/metabolismo , Helicasa del Síndrome de Werner
15.
Nucleic Acids Res ; 32(13): 4003-14, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15292449

RESUMEN

Werner syndrome (WS) is a genetic premature aging disorder in which patients appear much older than their chronological age. The gene mutated in WS encodes a nuclear protein (WRN) which possesses 3'-5' exonuclease and ATPase-dependent 3'-5' helicase activities. The genomic instability associated with WS cells and the biochemical characteristics of WRN suggest that WRN plays a role in DNA metabolic pathways such as transcription, replication, recombination and repair. Recently we have identified poly(ADP-ribose) polymerase-1 (PARP-1) as a new WRN interacting protein. In this paper, we further mapped the interacting domains. We found that PARP-1 bound to the N-terminus of WRN and to the C-terminus containing the RecQ-conserved (RQC) domain. WRN bound to the N-terminus of PARP-1 containing DNA binding and BRCA1 C-terminal (BRCT) domains. We show that unmodified PARP-1 inhibited both WRN exonuclease and helicase activities, and to our knowledge is the only known WRN protein partner that inactivates both of the WRN's catalytic activities suggesting a biologically significant regulation. Moreover, this dual inhibition seems to be specific for PARP-1, as PARP-2 did not affect WRN helicase activity and only slightly inhibited WRN exonuclease activity. The differential effect of PARP-1 and PARP-2 on WRN catalytic activity was not due to differences in affinity for WRN or the DNA substrate. Finally, we demonstrate that the inhibition of WRN by PARP-1 was influenced by the poly(ADP-ribosyl)ation state of PARP-1. The biological relevance of the specific modulation of WRN catalytic activities by PARP-1 are discussed in the context of pathways in which these proteins may function together, namely in the repair of DNA strand breaks.


Asunto(s)
ADN Helicasas/metabolismo , Exonucleasas/metabolismo , Poli(ADP-Ribosa) Polimerasas/fisiología , Sitios de Unión , ADN/química , ADN/metabolismo , ADN Helicasas/química , Exodesoxirribonucleasas , Exonucleasas/química , Células HeLa , Humanos , Modelos Biológicos , Poli(ADP-Ribosa) Polimerasas/química , Poli(ADP-Ribosa) Polimerasas/metabolismo , Estructura Terciaria de Proteína , RecQ Helicasas , Helicasa del Síndrome de Werner
16.
Mech Ageing Dev ; 125(7): 491-6, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15246744

RESUMEN

The leading causes of death for individuals with Werner syndrome (WS) are myocardial infarction (MI) and stroke. The WS gene encodes a nuclear protein with both helicase and exonuclease activities. While individuals with WS have mutations that result in truncated, inactive proteins, several sequence variants have been described in apparently unaffected individuals. Some of these gene polymorphisms encode non-conservative amino acid substitutions, and it is expected that the changes would affect enzyme activity, although this has not been determined. Two research groups have studied the Cys/Arg 1367 polymorphism (located near the nuclear localization signal) in healthy and MI patients. Their results suggest that the Arg allele is protective against MI. We have characterized the Cys (C) and Arg (R) forms of the protein and find no notable difference in helicase and nuclease activities, or in nuclear/cytoplasmic distribution. The frequency of the C/R alleles in healthy individuals and subjects with coronary artery disease (CAD) drawn from the Baltimore Longitudinal Study of Aging (BLSA) was also examined. There was no indication that the R allele was protective against CAD. We conclude that the C/R polymorphism does not affect enzyme function or localization and does not influence CAD incidence in the BLSA cohort.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , ADN Helicasas/genética , Predisposición Genética a la Enfermedad , Polimorfismo Genético , Población Blanca/genética , Anciano , Anciano de 80 o más Años , Alelos , Sustitución de Aminoácidos , Animales , Núcleo Celular/metabolismo , Células Cultivadas , Estudios de Cohortes , Citoplasma/metabolismo , ADN Helicasas/metabolismo , Exodesoxirribonucleasas , Exonucleasas/genética , Exonucleasas/metabolismo , Frecuencia de los Genes , Humanos , Masculino , Persona de Mediana Edad , RecQ Helicasas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Helicasa del Síndrome de Werner
17.
Toxicol Sci ; 77(2): 307-14, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14691214

RESUMEN

Chemical-DNA adducts provide an integrated measure of exposure, absorption, bioactivation, detoxification, and DNA repair following exposure to a genotoxic agent. Benzo[a]pyrene (BaP), a prototypical polycyclic aromatic hydrocarbon (PAH), can be bioactivated by cytochrome P-450s (CYPs) and epoxide hydrolase to genotoxic metabolites which form covalent adducts with DNA. In this study, we utilized precision-cut rat liver and lung slices exposed to BaP to investigate tissue-specific differences in chemical absorption and formation of DNA adducts. To investigate the contribution of bioactivating CYPs (such as CYP1A1 and CYP1B1) on the formation of BaP-DNA adducts, animals were also pretreated in vivo with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin) prior to in vitro incubation of tissue slices with BaP. Furthermore, the tissue distribution of BaP and BaP-DNA adduct levels from in vivo studies were compared with those from the in vitro tissue slice experiments. The results indicate a time- and concentration-dependent increase in tissue-associated BaP following exposure of rat liver and lung tissue slices to BaP in vitro, with generally higher levels of BaP retained in lung tissue. Furthermore, rat liver and lung slices metabolized BaP to reactive intermediates that formed covalent adducts with DNA. Total BaP-DNA adducts increased with concentration and incubation time. Adduct levels (fmol adduct/microg DNA) in lung slices were greater than liver at all doses. Liver slices contained one major and two minor adducts, while lung slices contained two major and 3 minor adducts. The tissue-specific qualitative profile of these adducts in tissue slices was similar to that observed from in vivo studies, further validating the use of this model. Pretreatment of animals with TCDD prior to in vitro incubation with BaP potentiated the levels of DNA adduct formation. TCDD pretreatment altered the adduct distribution in lung but not in liver slices. Together, the results suggest that tissue-specific qualitative and quantitative differences in BaP-DNA adducts could contribute to the lung being a target tissue for BaP carcinogenesis. Furthermore, the results validate the use of precision-cut tissue slices incubated in dynamic organ culture as a useful model for the study of chemical-DNA adduct formation.


Asunto(s)
Benzo(a)pireno/metabolismo , Benzo(a)pireno/farmacología , Carcinógenos Ambientales/farmacología , Aductos de ADN/metabolismo , Hígado/efectos de los fármacos , Pulmón/efectos de los fármacos , Animales , Hidrocarburo de Aril Hidroxilasas/biosíntesis , Benzo(a)pireno/análisis , Citocromo P-450 CYP1A1/biosíntesis , Citocromo P-450 CYP1B1 , Aductos de ADN/análisis , Aductos de ADN/biosíntesis , Hígado/metabolismo , Pulmón/metabolismo , Masculino , Modelos Animales , Técnicas de Cultivo de Órganos/métodos , Radioisótopos de Fósforo , Dibenzodioxinas Policloradas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Tritio
18.
Mol Cell Biol ; 23(23): 8601-13, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14612404

RESUMEN

A defect in the Werner syndrome protein (WRN) leads to the premature aging disease Werner syndrome (WS). Hallmark features of cells derived from WS patients include genomic instability and hypersensitivity to certain DNA-damaging agents. WRN contains a highly conserved region, the RecQ conserved domain, that plays a central role in protein interactions. We searched for proteins that bound to this region, and the most prominent direct interaction was with poly(ADP-ribose) polymerase 1 (PARP-1), a nuclear enzyme that protects the genome by responding to DNA damage and facilitating DNA repair. In pursuit of a functional interaction between WRN and PARP-1, we found that WS cells are deficient in the poly(ADP-ribosyl)ation pathway after they are treated with the DNA-damaging agents H2O2 and methyl methanesulfonate. After cellular stress, PARP-1 itself becomes activated, but the poly(ADP-ribosyl)ation of other cellular proteins is severely impaired in WS cells. Overexpression of the PARP-1 binding domain of WRN strongly inhibits the poly(ADP-ribosyl)ation activity in H2O2-treated control cell lines. These results indicate that the WRN/PARP-1 complex plays a key role in the cellular response to oxidative stress and alkylating agents, suggesting a role for these proteins in the base excision DNA repair pathway.


Asunto(s)
Daño del ADN , ADN Helicasas/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Síndrome de Werner/metabolismo , Sitios de Unión , Línea Celular , ADN Helicasas/química , ADN Helicasas/genética , Reparación del ADN , Exodesoxirribonucleasas , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Peróxido de Hidrógeno/toxicidad , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Sustancias Macromoleculares , Metilmetanosulfonato/toxicidad , Mutación , Estrés Oxidativo , Poli(ADP-Ribosa) Polimerasas/química , Estructura Terciaria de Proteína , RecQ Helicasas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Síndrome de Werner/genética , Helicasa del Síndrome de Werner
19.
Cancer Res ; 63(21): 7136-46, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14612507

RESUMEN

Werner syndrome (WS) is a human premature aging disorder characterized by the early onset of age-related clinical features and an elevated incidence of cancer. The Werner protein (WRN) belongs to the RecQ family of DNA helicases and is required for the maintenance of genomic stability in human cells. Potential cooperation between RecQ helicases and topoisomerases in many aspects of DNA metabolism, such as the progression of replication forks, transcription, recombination, and repair, has been reported. Here, we show a physical and functional interaction between WRN and topoisomerase I (topo I). WRN colocalizes and interacts directly with topo I. WRN stimulates the ability of topo I to relax negatively supercoiled DNA and specifically stimulates the religation step of the relaxation reaction. Moreover, cell extracts from WS fibroblasts exhibit a decrease in the relaxation activity of negatively supercoiled DNA. We have identified two regions of WRN that mediate functional interaction with topo I, and they are located at the NH(2) and COOH termini of the WRN protein. In a reciprocal functional interaction, topo I inhibits the ATPase activity of WRN. Our data provide new insight into the interrelationship between RecQ helicases and topoisomerases in the maintenance of genomic integrity and prevention of tumorigenesis.


Asunto(s)
ADN Helicasas/metabolismo , ADN-Topoisomerasas de Tipo I/metabolismo , Adenosina Trifosfatasas/antagonistas & inhibidores , Camptotecina/farmacología , Células Cultivadas , ADN Helicasas/antagonistas & inhibidores , ADN Superhelicoidal/metabolismo , Exodesoxirribonucleasas , Fibroblastos/enzimología , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Mapeo Peptídico , Estructura Terciaria de Proteína , RecQ Helicasas , Síndrome de Werner/enzimología , Síndrome de Werner/patología , Helicasa del Síndrome de Werner
20.
Carcinogenesis ; 24(5): 791-802, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12771022

RESUMEN

Werner syndrome (WS) is a hallmark premature aging disease, in which the patients appear much older than their chronological age, and exhibit many of the clinical signs and symptoms of normal aging at an early stage in life. They develop many age-associated diseases early in life including atherosclerosis, osteoporosis, cataracts and display a high incidence of cancer. WS is also marked by increased genomic instability, manifested as chromosomal alterations. Characterization and study of the Werner protein (WRN) suggests that it participates in several important DNA metabolic pathways, and that its primary function may be in DNA repair processes. Thus, the WRN protein represents an important link between defective DNA repair and the processes related to aging and cancer.


Asunto(s)
Envejecimiento/fisiología , ADN Helicasas/fisiología , Reparación del ADN , Síndrome de Werner/genética , Exodesoxirribonucleasas , Humanos , RecQ Helicasas , Helicasa del Síndrome de Werner
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...