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










Base de dados
Intervalo de ano de publicação
1.
Virus Res ; 286: 198076, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32603670

RESUMO

Ubiquitin Specific Protease 7 (USP7) is a deubiquitinating enzyme (DUB) that plays critical roles in the regulation of many cellular processes including epigenetics, tumour suppression, oncogenesis, DNA damage response, immunity and viral infection. USP7 was the first DUB associated with viral infection. Since then other DUB:viral protein interactions have been discovered, however, USP7 continues to be the most targeted DUB interacting with many proteins from various viruses. The selective pressures of evolution have allowed viruses to develop mechanisms that subvert host cellular machinery, promoting survival of the viral niche. Numerous viral proteins have been identified to target and usurp the function of USP7 to their advantage. This review explores novel developments in research focusing on the mechanisms underlying the manipulation of USP7 by viruses.


Assuntos
Peptidase 7 Específica de Ubiquitina/genética , Peptidase 7 Específica de Ubiquitina/metabolismo , Proteínas Virais/genética , Vírus/genética , Animais , Instabilidade Genômica , Humanos , Proteínas Imediatamente Precoces , Camundongos , Ligação Proteica , Ubiquitina/metabolismo , Proteínas Virais/metabolismo , Vírus/metabolismo
2.
J Mol Biol ; 432(4): 897-912, 2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-31866294

RESUMO

USP7 is a deubiquitinase that regulates many diverse cellular processes, including tumor suppression, epigenetics, and genome stability. Several substrates, including GMPS, UHRF1, and ICP0, were shown to bear a specific KxxxK motif that interacts within the C-terminal region of USP7. We identified a similar motif in Enhancer of Zeste 2 (EZH2), the histone methyltransferase found within Polycomb Repressive Complex 2 (PRC2). PRC2 is responsible for the methylation of Histone 3 Lys27 (H3K27) leading to gene silencing. GST pull-down and coimmunoprecipitation experiments showed that USP7 interacts with EZH2. We determined the structural basis of interaction between USP7 and EZH2 and identified residues mediating the interaction. Mutations in these critical residues disrupted the interaction between USP7 and EZH2. Furthermore, USP7 silencing and knockout experiments showed decreased EZH2 levels in HCT116 carcinoma cells. Finally, we demonstrated decreased H3K27Me3 levels in HCT116 USP7 knockout cells. These results indicate that USP7 interacts with EZH2 and regulates both its stability and function.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Peptidase 7 Específica de Ubiquitina/metabolismo , Linhagem Celular Tumoral , Eletroforese em Gel de Poliacrilamida , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Inativação Gênica/fisiologia , Células HCT116 , Humanos , Imunoprecipitação , Complexo Repressor Polycomb 2/genética , Estabilidade Proteica , Peptidase 7 Específica de Ubiquitina/genética , Ubiquitinação/genética , Ubiquitinação/fisiologia
3.
J Biol Chem ; 292(7): 2893-2902, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28073915

RESUMO

UbE2E1/UbcH6 is an E2 ubiquitin-conjugating enzyme that is regulated by USP7. We identified UbE2E1 as a novel component of Polycomb repressive complex 1 (PRC1), the E3 ligase complex responsible for histone H2A ubiquitination and gene silencing. We demonstrate that UbE2E1 is critical for the monoubiquitination of H2A at residue Lys-119 (uH2AK119) through its association with the PRC1 complex. UbE2E1 interacts with PRC1 subunits including Ring1A and Ring1B. Overexpression of UbE2E1 results in increased levels of uH2AK119, whereas overexpression of catalytically inactive UbE2E1_C131A or UbE2E1 knockdown results in decreased levels of uH2AK119. The down-regulation of H2A ubiquitination by loss of function of UbE2E1 is correlated with alleviated p16INK4a promoter repression and induced growth inhibition in HCT116 cells. These results are specific to UbE2E1 as knockdown of UbE2D E2s does not show any effect on uH2AK119. We extended the UbE2E1 regulation of uH2AK119 to USP7 and showed that USP7 is also a key regulator for monoubiquitination at H2A Lys-119 as both knockdown and deletion of USP7 results in decreased levels of uH2AK119. This study reveals that UbE2E1 is an in vivo E2 for the PRC1 ligase complex and thus plays an important role in the regulation of H2A Lys-119 monoubiquitination.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Catálise , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inativação Gênica , Histonas/química , Humanos , Ligação Proteica , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , Enzimas de Conjugação de Ubiquitina/genética , Peptidase 7 Específica de Ubiquitina , Ubiquitinação
4.
Cell Commun Signal ; 14(1): 17, 2016 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-27549312

RESUMO

BACKGROUND: CASKIN2 is a homolog of CASKIN1, a scaffolding protein that participates in a signaling network with CASK (calcium/calmodulin-dependent serine kinase). Despite a high level of homology between CASKIN2 and CASKIN1, CASKIN2 cannot bind CASK due to the absence of a CASK Interaction Domain and consequently, may have evolved undiscovered structural and functional distinctions. RESULTS: We demonstrate that the crystal structure of the Sterile Alpha Motif (SAM) domain tandem (SAM1-SAM2) oligomer from CASKIN2 is different than CASKIN1, with the minimal repeating unit being a dimer, rather than a monomer. Analytical ultracentrifugation sedimentation velocity methods revealed differences in monomer/dimer equilibria across a range of concentrations and ionic strengths for the wild type CASKIN2 SAM tandem and a structure-directed double mutant that could not oligomerize. Further distinguishing CASKIN2 from CASKIN1, EGFP-tagged SAM tandem proteins expressed in Neuro2a cells produced punctae that were distinct both in shape and size. CONCLUSIONS: This study illustrates a new way in which neuronal SAM domains can assemble into large macromolecular assemblies that might concentrate and amplify synaptic responses.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas do Tecido Nervoso/química , Multimerização Proteica , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Humanos , Simulação de Dinâmica Molecular , Proteínas do Tecido Nervoso/metabolismo , Domínios Proteicos
5.
J Biol Chem ; 291(12): 6281-91, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26786098

RESUMO

Viral interferon regulatory factor 1 (vIRF1), a Kaposi sarcoma herpesvirus protein, destabilizes p53 by inhibiting p53 acetylation and Hdm2 phosphorylation. This leads to increased ubiquitination and degradation of p53 by Hdm2, which cripples the cellular p53-mediated antiviral response. Ubiquitin-specific protease 7 (USP7) deubiquitinates p53 and Hdm2 and regulates their stability. We identified an EGPS consensus sequence in vIRF1, which is identical to that found in Epstein-Barr virus nuclear antigen 1 (EBNA1) that interacts with the N-terminal domain of USP7 (USP7-NTD). GST pulldown assays demonstrated that vIRF1 interacts with USP7-NTD via its EGPS motif. NMR heteronuclear single quantum correlation (HSQC) analysis revealed chemical perturbations after titration of USP7-NTD with vIRF1 (44)SPGEGPSGTG(53) peptide. In contrast, these perturbations were reduced with a mutant vIRF1 peptide, (44)SPGEGPAGTG(53). Fluorescence polarization analysis indicated that the vIRF1 peptide interacted with USP7-NTD with a Kd of 2.0 µm. The crystal structure of the USP7-NTD·vIRF1 peptide complex revealed an identical mode of binding as that of the EBNA1 peptide to USP7-NTD. We also showed that USP7 interacts with vIRF1 in U2OS cells. Decreased levels of p53, but not Hdm2 or ataxia telangiectasia-mutated (ATM), were seen after expression of vIRF1, but not with a vIRF1 mutant protein. Our results support a new role for vIRF1 through deregulation of the deubiquitinating enzyme USP7 to inhibit p53-mediated antiviral responses.


Assuntos
Herpesvirus Humano 8 , Fatores Reguladores de Interferon/química , Ubiquitina Tiolesterase/química , Proteínas Virais/química , Motivos de Aminoácidos , Domínio Catalítico , Linhagem Celular Tumoral , Cristalografia por Raios X , Humanos , Fatores Reguladores de Interferon/fisiologia , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina Tiolesterase/fisiologia , Peptidase 7 Específica de Ubiquitina , Proteínas Virais/fisiologia
6.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 10): 2032-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26457427

RESUMO

Nicotinamide mononucleotide adenylyltransferase (NMNAT) catalyzes the biosynthesis of NAD(+) and NaAD(+). The crystal structure of NMNAT from Methanobacterium thermoautotrophicum complexed with NAD(+) and SO4(2-) revealed the active-site residues involved in binding and catalysis. Site-directed mutagenesis was used to further characterize the roles played by several of these residues. Arg11 and Arg136 were implicated in binding the phosphate groups of the ATP substrate. Both of these residues were mutated to lysine individually. Arg47 does not interact with either NMN or ATP substrates directly, but was deemed to play a role in binding as it is proximal to Arg11 and Arg136. Arg47 was mutated to lysine and glutamic acid. Surprisingly, when expressed in Escherichia coli all of these NMNAT mutants trapped a molecule of NADP(+) in their active sites. This NADP(+) was bound in a conformation that was quite different from that displayed by NAD(+) in the native enzyme complex. When NADP(+) was co-crystallized with wild-type NMNAT, the same structural arrangement was observed. These studies revealed a different conformation of NADP(+) in the active site of NMNAT, indicating plasticity of the active site.


Assuntos
Methanobacterium/enzimologia , NADP/metabolismo , NAD/metabolismo , Nicotinamida-Nucleotídeo Adenililtransferase/química , Nicotinamida-Nucleotídeo Adenililtransferase/metabolismo , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Methanobacterium/química , Methanobacterium/metabolismo , Modelos Moleculares , Conformação Proteica
7.
PLoS Pathog ; 11(6): e1004950, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26046769

RESUMO

Herpes simplex virus-1 immediate-early protein ICP0 activates viral genes during early stages of infection, affects cellular levels of multiple host proteins and is crucial for effective lytic infection. Being a RING-type E3 ligase prone to auto-ubiquitination, ICP0 relies on human deubiquitinating enzyme USP7 for protection against 26S proteasomal mediated degradation. USP7 is involved in apoptosis, epigenetics, cell proliferation and is targeted by several herpesviruses. Several USP7 partners, including ICP0, GMPS, and UHRF1, interact through its C-terminal domain (CTD), which contains five ubiquitin-like (Ubl) structures. Despite the fact that USP7 has emerged as a drug target for cancer therapy, structural details of USP7 regulation and the molecular mechanism of interaction at its CTD have remained elusive. Here, we mapped the binding site between an ICP0 peptide and USP7 and determined the crystal structure of the first three Ubl domains bound to the ICP0 peptide, which showed that ICP0 binds to a loop on Ubl2. Sequences similar to the USP7-binding site in ICP0 were identified in GMPS and UHRF1 and shown to bind USP7-CTD through Ubl2. In addition, co-immunoprecipitation assays in human cells comparing binding to USP7 with and without a Ubl2 mutation, confirmed the importance of the Ubl2 binding pocket for binding ICP0, GMPS and UHRF1. Therefore we have identified a novel mechanism of USP7 recognition that is used by both viral and cellular proteins. Our structural information was used to generate a model of near full-length USP7, showing the relative position of the ICP0/GMPS/UHRF1 binding pocket and the structural basis by which it could regulate enzymatic activity.


Assuntos
Infecções por Herpesviridae/metabolismo , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/metabolismo , Ubiquitina Tiolesterase/química , Ubiquitina Tiolesterase/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Sítios de Ligação/fisiologia , Western Blotting , Cristalização , Células HEK293 , Humanos , Imunoprecipitação , Ligação Proteica/fisiologia , Conformação Proteica , Transfecção , Peptidase 7 Específica de Ubiquitina
8.
Endocr Relat Cancer ; 22(1): T35-54, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25605410

RESUMO

Deubiquitinases (DUBs) play important roles and therefore are potential drug targets in various diseases including cancer and neurodegeneration. In this review, we recapitulate structure-function studies of the most studied DUBs including USP7, USP22, CYLD, UCHL1, BAP1, A20, as well as ataxin 3 and connect them to regulatory mechanisms and their growing protein interaction networks. We then describe DUBs that have been associated with endocrine carcinogenesis with a focus on prostate, ovarian, and thyroid cancer, pheochromocytoma, and adrenocortical carcinoma. The goal is enhancing our understanding of the connection between dysregulated DUBs and cancer to permit the design of therapeutics and to establish biomarkers that could be used in diagnosis and prognosis.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Proteases Específicas de Ubiquitina/antagonistas & inibidores , Proteases Específicas de Ubiquitina/metabolismo , Ubiquitina/metabolismo , Animais , Humanos , Terapia de Alvo Molecular , Transdução de Sinais , Ubiquitina Tiolesterase/antagonistas & inibidores , Ubiquitina Tiolesterase/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/metabolismo
9.
Mol Cell Biol ; 34(1): 132-45, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24190967

RESUMO

The minichromosome maintenance (MCM) complex, which plays multiple important roles in DNA replication, is loaded onto chromatin following mitosis, remains on chromatin until the completion of DNA synthesis, and then is unloaded by a poorly defined mechanism that involves the MCM binding protein (MCM-BP). Here we show that MCM-BP directly interacts with the ubiquitin-specific protease USP7, that this interaction occurs predominantly on chromatin, and that MCM-BP can tether USP7 to MCM proteins. Detailed biochemical and structure analyses of the USP7-MCM-BP interaction showed that the (155)PSTS(158) MCM-BP sequence mediates critical interactions with the TRAF domain binding pocket of USP7. Analysis of the effects of USP7 knockout on DNA replication revealed that lack of USP7 results in slowed progression through late S phase without globally affecting the fork rate or origin usage. Lack of USP7 also resulted in increased levels of MCM proteins on chromatin, and investigation of the cause of this increase revealed a defect in the dissociation of MCM proteins from chromatin in mid- to late S phase. This role of USP7 mirrors the previously described role for MCM-BP in MCM complex unloading and suggests that USP7 works with MCM-BP to unload MCM complexes from chromatin at the end of S phase.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cromatina/metabolismo , Replicação do DNA , Proteínas Nucleares/metabolismo , Ubiquitina Tiolesterase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Western Blotting , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Cromatina/genética , Cristalografia por Raios X , Citometria de Fluxo , Fase G1/genética , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Componente 7 do Complexo de Manutenção de Minicromossomo/genética , Componente 7 do Complexo de Manutenção de Minicromossomo/metabolismo , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica , Estrutura Terciária de Proteína , Interferência de RNA , Fase S/genética , Ubiquitina Tiolesterase/química , Ubiquitina Tiolesterase/genética , Peptidase 7 Específica de Ubiquitina
10.
J Biol Chem ; 288(23): 16975-16985, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23603909

RESUMO

Ubiquitin-specific protease 7 (USP7) is a deubiquitinating enzyme found in all eukaryotes that catalyzes the removal of ubiquitin from specific target proteins. Here, we report that UbE2E1, an E2 ubiquitin conjugation enzyme with a unique N-terminal extension, is a novel USP7-interacting protein. USP7 forms a complex with UbE2E1 in vitro and in vivo through the ASTS USP7 binding motif within its N-terminal extension in an identical manner with other known USP7 binding proteins. We show that USP7 attenuates UbE2E1-mediated ubiquitination, an effect that requires the N-terminal ASTS sequence of UbE2E1 as well as the catalytic activity of USP7. Additionally, USP7 is critical in maintaining the steady state levels of UbE2E1 in cells. This study reveals a new cellular mechanism that couples the opposing activities of the ubiquitination machinery and a deubiquitinating enzyme to maintain and modulate the dynamic balance of the ubiquitin-proteasome system.


Assuntos
Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina Tiolesterase/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinação/fisiologia , Motivos de Aminoácidos , Células HeLa , Humanos , Complexo de Endopeptidases do Proteassoma/genética , Ligação Proteica , Ubiquitina Tiolesterase/genética , Enzimas de Conjugação de Ubiquitina/genética , Peptidase 7 Específica de Ubiquitina
11.
J Biol Chem ; 286(6): 4796-808, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21084285

RESUMO

The tumor suppressor p53 maintains genome stability and prevents malignant transformation by promoting cell cycle arrest and apoptosis. Both Mdm2 and Pirh2 have been shown to ubiquitylate p53 through their RING domains, thereby targeting p53 for proteasomal degradation. Using structural and functional analyses, here we show that the Pirh2 RING domain differs from the Mdm2 RING domain in its oligomeric state, surface charge distribution, and zinc coordination scheme. Pirh2 also possesses weaker E3 ligase activity toward p53 and directs ubiquitin to different residues on p53. NMR and mutagenesis studies suggest that whereas Pirh2 and Mdm2 share a conserved E2 binding site, the seven C-terminal residues of the Mdm2 RING directly contribute to Mdm2 E3 ligase activity, a feature unique to Mdm2 and absent in the Pirh2 RING domain. This comprehensive analysis of the Pirh2 and Mdm2 RING domains provides structural and mechanistic insight into p53 regulation by its E3 ligases.


Assuntos
Proteínas Proto-Oncogênicas c-mdm2/química , Proteína Supressora de Tumor p53/química , Ubiquitina-Proteína Ligases/química , Sítios de Ligação , Humanos , Mutagênese , Ressonância Magnética Nuclear Biomolecular , Multimerização Proteica , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Domínios RING Finger , Relação Estrutura-Atividade , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
12.
J Mol Biol ; 402(5): 825-37, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20713061

RESUMO

Ubiquitin-specific protease 7 (USP7) catalyzes the deubiquitination of several substrate proteins including p53 and Hdm2. We have previously shown that USP7, and more specifically its amino-terminal domain (USP7-NTD), interacts with distinct regions on p53 and Hdm2 containing P/AxxS motifs. The ability of USP7 to also deubiquitinate and control the turnover of HdmX was recently demonstrated. We utilized a combination of biochemistry and structural biology to identify which domain of USP7 interacts with HdmX as well as to identify regions of HdmX that interact with USP7. We showed that USP7-NTD recognized two of six P/AxxS motifs of HdmX ((8)AQCS(11) and (398)AHSS(401)). The crystal structure of the USP7-NTD:HdmX(AHSS) complex was determined providing the molecular basis of complex formation between USP7-NTD and the HdmX(AHSS) peptide. The HdmX peptide interacted within the same residues of USP7-NTD as previously demonstrated with p53, Hdm2, and EBNA1 peptides. We also identified an additional site on Hdm2 ((397)PSTS(400)) that interacts with USP7-NTD and determined the crystal structure of this complex. Finally, analysis of USP7-interacting peptides on filter arrays confirmed the importance of the serine residue at the fourth position for the USP7-NTD interaction and showed that phosphorylation of serines within the binding sequence prevents this interaction. These results lead to a better understanding of the mechanism of substrate recognition by USP7-NTD.


Assuntos
Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Mapeamento de Interação de Proteínas , Proteínas Proto-Oncogênicas c-mdm2/química , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Ubiquitina Tiolesterase/química , Ubiquitina Tiolesterase/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Proteínas de Ciclo Celular , Cristalografia por Raios X , Modelos Moleculares , Dados de Sequência Molecular , Fosforilação , Ligação Proteica , Estrutura Quaternária de Proteína , Alinhamento de Sequência , Serina/metabolismo , Peptidase 7 Específica de Ubiquitina
13.
Biochem J ; 419(1): 141-8, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19099536

RESUMO

ROCK (Rho-associated coiled-coil kinase) 2 is a member of the AGC kinase family that plays an essential role downstream of Rho in actin cytoskeleton assembly and contractility. The process of ROCK2 activation is complex and requires suppression of an autoinhibitory mechanism that is facilitated by Rho binding. ROCK2 harbours a C-terminal extension within the kinase domain that contains a hydrophobic cluster of phenylalanine and tyrosine residues surrounding a key threonine residue. In growth-factor-stimulated AGC kinases, the hydrophobic motif is important for the transition of the kinase from inactive to active complex and requires phosphorylation of the conserved serine/threonine residue. Less is understood about the contribution that the hydrophobic motif plays in the activation of ROCK, and the role of the hydrophobic motif threonine at position 405. In the present study, we show that this residue of ROCK is essential for substrate phosphorylation and kinase domain dimerization. However, in contrast with the growth-factor-activated AGC kinases, a phosphomimetic residue at position 405 was inhibitory for ROCK2 activity and dimerization. A soluble hydrophobic motif peptide allosterically activated ROCK2 In vitro, but not the equivalent peptide with Asp(405) substitution. Mechanistically, both ROCK2 activity and dimerization were dependent upon the interaction between Thr(405) of the hydrophobic motif and Asp(39) of the N-terminal extension. The reciprocal exchange of these residues was permissive for kinase activity, but dimerization was lost. These results support the rationale for development of small-molecule inhibitors designed to block ROCK activation by selectively interfering with hydrophobic motif-mediated activation-state transition and dimer formation.


Assuntos
Quinases Associadas a rho/química , Quinases Associadas a rho/metabolismo , Motivos de Aminoácidos , Animais , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Linhagem Celular , Dimerização , Células HeLa , Humanos , Interações Hidrofóbicas e Hidrofílicas , Immunoblotting , Imunoprecipitação , Camundongos , Mutagênese Sítio-Dirigida , Células NIH 3T3 , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Treonina/genética , Treonina/metabolismo , Quinases Associadas a rho/genética
14.
J Mol Biol ; 377(3): 655-67, 2008 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-18272181

RESUMO

Transcriptional regulators belonging to the MarR family are characterized by a winged-helix DNA binding domain. These transcriptional regulators regulate the efflux and influx of phenolic agents in bacteria and archaea. In Escherichia coli, MarR regulates the multiple antibiotic resistance operon and its inactivation produces a multiple antibiotic resistance phenotype. In some organisms, active efflux of drug compounds will produce a drug resistance phenotype, whereas in other organisms, active influx of chlorinated hydrocarbons results in their rapid degradation. Although proteins in the MarR family are regulators of important biological processes, their mechanism of action is not well understood and structural information about how phenolic agents regulate the activity of these proteins is lacking. This article presents the three-dimensional structure of a protein of the MarR family, MTH313, in its apo form and in complex with salicylate, a known inactivator. A comparison of these two structures indicates that the mechanism of regulation involves a large conformational change in the DNA binding lobe. Electrophoretic mobility shift assay and biophysical analyses further suggest that salicylate inactivates MTH313 and prevents it from binding to its promoter region.


Assuntos
Proteínas de Bactérias/química , Proteínas de Ligação a DNA/química , Methanobacterium/química , Modelos Moleculares , Proteínas Repressoras/química , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Resistência Microbiana a Medicamentos , Dados de Sequência Molecular , Conformação Proteica , Salicilatos/farmacologia
15.
Nat Struct Mol Biol ; 13(3): 285-91, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16474402

RESUMO

The ubiquitin-specific protease, USP7, has key roles in the p53 pathway whereby it stabilizes both p53 and MDM2. We show that the N-terminal domain of USP7 binds two closely spaced 4-residue sites in both p53 and MDM2, falling between p53 residues 359-367 and MDM2 residues 147-159. Cocrystal structures with USP7 were determined for both p53 peptides and for one MDM2 peptide. These peptides bind the same surface of USP7 as Epstein-Barr nuclear antigen-1, explaining the competitive nature of the interactions. The structures and mutagenesis data indicate a preference for a P/AXXS motif in peptides that bind USP7. Contacts made by serine are identical and crucial for all peptides, and Trp165 in the peptide-binding pocket of USP7 is also crucial. These results help to elucidate the mechanism of substrate recognition by USP7 and the regulation of the p53 pathway.


Assuntos
Endopeptidases/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Sítios de Ligação/genética , Cristalografia por Raios X , Endopeptidases/química , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-mdm2/química , Proteína Supressora de Tumor p53/química , Ubiquitina Tiolesterase , Peptidase 7 Específica de Ubiquitina
16.
Mol Cell ; 18(1): 25-36, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15808506

RESUMO

USP7/HAUSP is a key regulator of p53 and Mdm2 and is targeted by the Epstein-Barr nuclear antigen 1 (EBNA1) protein of Epstein-Barr virus (EBV). We have determined the crystal structure of the p53 binding domain of USP7 alone and bound to an EBNA1 peptide. This domain is an eight-stranded beta sandwich similar to the TRAF-C domains of TNF-receptor associated factors, although the mode of peptide binding differs significantly from previously observed TRAF-peptide interactions in the sequence (DPGEGPS) and the conformation of the bound peptide. NMR chemical shift analyses of USP7 bound by EBNA1 and p53 indicated that p53 binds the same pocket as EBNA1 but makes less extensive contacts with USP7. Functional studies indicated that EBNA1 binding to USP7 can protect cells from apoptotic challenge by lowering p53 levels. The data provide a structural and conceptual framework for understanding how EBNA1 might contribute to the survival of Epstein-Barr virus-infected cells.


Assuntos
Endopeptidases/química , Endopeptidases/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/química , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/metabolismo , Sequência de Aminoácidos , Apoptose , Sítios de Ligação , Cristalografia por Raios X , Escherichia coli/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Ubiquitina Tiolesterase , Peptidase 7 Específica de Ubiquitina
17.
J Biol Chem ; 279(22): 23646-53, 2004 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-15044458

RESUMO

ATP:cobalamin adenosyltransferase MMAB was recently identified as the gene responsible for a disorder of cobalamin metabolism in humans (cblB complementation group). The crystal structure of the MMAB sequence homologue from Thermoplasma acidophilum (TA1434; GenBank identification number gi|16082403) was determined to a resolution of 1.5 A. TA1434 was confirmed to be an ATP:cobalamin adenosyltransferase, which depended absolutely on divalent metal ions (Mg2+ > Mn2+ > Co2+) and only used ATP or dATP as adenosyl donors. The apparent Km of TA1434 was 110 microM (kcat = 0.23 s(-1)) for ATP, 140 microM (kcat = 0.11 s(-1)) for dATP, and 3 microM (kcat = 0.18 s(-1)) for cobalamin. TA1434 is a trimer in solution and in the crystal structure, with each subunit composed of a five-helix bundle. The location of disease-related point mutations and other residues conserved among the homologues of TA1434 suggest that the active site lies at the junctions between the subunits. Mutations in TA1434 that correspond to the disease-related mutations resulted in proteins that were inactive for ATP:cobalamin adenosyltransferase activity in vitro, confirming that these mutations define the molecular basis of the human disease.


Assuntos
Alquil e Aril Transferases/química , Proteínas Arqueais/química , Thermoplasma/enzimologia , Alquil e Aril Transferases/metabolismo , Sequência de Aminoácidos , Proteínas Arqueais/metabolismo , Ácido Metilmalônico/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Alinhamento de Sequência , Vitamina B 12/metabolismo
19.
J Biol Chem ; 278(36): 34356-63, 2003 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-12810729

RESUMO

Several residues lining the ATP-binding site of Methanobacterium thermoautotrophicum nicotinamide mononucleotide adenylyltransferase (NMNATase) were mutated in an effort to better characterize their roles in substrate binding and catalysis. Residues selected were Arg-11 and Arg-136, both of which had previously been implicated as substrate binding residues, as well as His-16 and His-19, part of the HXGH active site motif and postulated to be of importance in catalysis. Kinetic studies revealed that both Arg-11 and Arg-136 contributed to the binding of the substrate, ATP. When these amino acids were replaced by lysines, the apparent Km values of the respective mutants for ATP decreased by factors of 1.3 and 2.9 and by factors of 1.9 and 8.8 when the same residues were changed to alanines. All four Arg mutants displayed unaltered Km values for NMN. The apparent kcat values of the R11K and R136K mutants were the same as those of WT NMNATase but the apparent kcat values of the alanine mutants had decreased. Crystal structures of the Arg mutants revealed NAD+ and SO42- molecules trapped at their active sites. The binding interactions of NAD+ were unchanged but the binding of SO42- was altered in these mutants compared with wild type. The alanine mutants at positions His-16 and His-19 retained approximately 6 and 1.3%, respectively, of WT NMNATase activity indicating that His-19 is a key catalytic group. Surprisingly, this H19A mutant displayed a novel and distinct mode of NAD+ binding when co-crystallized in the presence of NAD+ and SO42-.


Assuntos
Methanobacterium/enzimologia , Nicotinamida-Nucleotídeo Adenililtransferase/química , Trifosfato de Adenosina/metabolismo , Alanina/química , Motivos de Aminoácidos , Arginina/química , Sítios de Ligação , Catálise , Cristalografia por Raios X , Análise Mutacional de DNA , Histidina/química , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , NAD/metabolismo , Fosfatos/química , Conformação Proteica
20.
Protein Sci ; 11(6): 1409-14, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12021439

RESUMO

As part of our structural proteomics initiative, we have determined the crystal structure of MTH1491, a previously uncharacterized hypothetical protein from Methanobacterium thermoautotrophicum. MTH1491 is one of numerous structural genomics targets selected in a genome-wide survey of uncharacterized proteins. It belongs to a family of proteins whose biological function is not known. The crystal structure of MTH1491, the first structure for this family of proteins, consists of an overall five-stranded parallel beta-sheet with strand order 51234 and flanking helices. The oligomeric form of this molecule is a trimer as seen from both crystal contacts and gel filtration studies. Analysis revealed that the structure of MTH1491 is similar to that of dehydrogenases, amidohydrolases, and oxidoreductases. Using a combination of sequence and structural analyses, we showed that MTH1491 does not belong to either the dehydrogenase or the amidohydrolase superfamilies of proteins.


Assuntos
Proteínas de Bactérias/química , Methanobacterium/química , Proteínas de Bactérias/fisiologia , Sítios de Ligação , Cristalografia por Raios X , Estrutura Molecular , Conformação Proteica , Relação Estrutura-Atividade , Sulfatos/química , Sulfatos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...