Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 11(11): e1005248, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26544981

RESUMEN

Anaplasma phagocytophilum, the causative agent of Human Granulocytic Anaplasmosis (HGA), is an obligately intracellular α-proteobacterium that is transmitted by Ixodes spp ticks. However, the pathogen is not transovarially transmitted between tick generations and therefore needs to survive in both a mammalian host and the arthropod vector to complete its life cycle. To adapt to different environments, pathogens rely on differential gene expression as well as the modification of proteins and other molecules. Random transposon mutagenesis of A. phagocytophilum resulted in an insertion within the coding region of an o-methyltransferase (omt) family 3 gene. In wild-type bacteria, expression of omt was up-regulated during binding to tick cells (ISE6) at 2 hr post-inoculation, but nearly absent by 4 hr p.i. Gene disruption reduced bacterial binding to ISE6 cells, and the mutant bacteria that were able to enter the cells were arrested in their replication and development. Analyses of the proteomes of wild-type versus mutant bacteria during binding to ISE6 cells identified Major Surface Protein 4 (Msp4), but also hypothetical protein APH_0406, as the most differentially methylated. Importantly, two glutamic acid residues (the targets of the OMT) were methyl-modified in wild-type Msp4, whereas a single asparagine (not a target of the OMT) was methylated in APH_0406. In vitro methylation assays demonstrated that recombinant OMT specifically methylated Msp4. Towards a greater understanding of the overall structure and catalytic activity of the OMT, we solved the apo (PDB_ID:4OA8), the S-adenosine homocystein-bound (PDB_ID:4OA5), the SAH-Mn2+ bound (PDB_ID:4PCA), and SAM- Mn2+ bound (PDB_ID:4PCL) X-ray crystal structures of the enzyme. Here, we characterized a mutation in A. phagocytophilum that affected the ability of the bacteria to productively infect cells from its natural vector. Nevertheless, due to the lack of complementation, we cannot rule out secondary mutations.


Asunto(s)
Anaplasma phagocytophilum/enzimología , Ehrlichiosis/microbiología , Ixodes/microbiología , Metiltransferasas/metabolismo , Garrapatas/microbiología , Animales , Ehrlichiosis/genética , Ixodes/inmunología , Metiltransferasas/genética , Activación Transcripcional , Regulación hacia Arriba
2.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 5): 615-21, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25961325

RESUMEN

Prior studies have highlighted the potential of superoxide dismutases as drug targets in eukaryotic pathogens. This report presents the structures of three iron-dependent superoxide dismutases (FeSODs) from Trypanosoma cruzi, Leishmania major and Babesia bovis. Comparison with existing structures from Plasmodium and other trypanosome isoforms shows a very conserved overall fold with subtle differences. In particular, structural data suggest that B. bovis FeSOD may display similar resistance to peroxynitrite-mediated inactivation via an intramolecular electron-transfer pathway as previously described in T. cruzi FeSOD isoform B, thus providing valuable information for structure-based drug design. Furthermore, lysine-acetylation results in T. cruzi indicate that acetylation occurs at a position close to that responsible for the regulation of acetylation-mediated activity in the human enzyme.


Asunto(s)
Babesia bovis/enzimología , Eucariontes/enzimología , Leishmania major/enzimología , Superóxido Dismutasa/química , Trypanosoma cruzi/enzimología , Secuencia de Aminoácidos , Apicomplexa/química , Apicomplexa/enzimología , Apicomplexa/genética , Babesia bovis/química , Babesia bovis/genética , Cristalización , Cristalografía por Rayos X , Eucariontes/química , Eucariontes/genética , Humanos , Leishmania major/química , Leishmania major/genética , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Superóxido Dismutasa/genética , Trypanosoma cruzi/química , Trypanosoma cruzi/genética
3.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 5): 609-14, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25945716

RESUMEN

The crystal structures of prostaglandin F synthase (PGF) from both Leishmania major and Trypanosoma cruzi with and without their cofactor NADP have been determined to resolutions of 2.6 Å for T. cruzi PGF, 1.25 Å for T. cruzi PGF with NADP, 1.6 Å for L. major PGF and 1.8 Å for L. major PGF with NADP. These structures were determined by molecular replacement to a final R factor of less than 18.6% (Rfree of less than 22.9%). PGF in the infectious protozoa L. major and T. cruzi is a potential therapeutic target.


Asunto(s)
Hidroxiprostaglandina Deshidrogenasas/química , Leishmania major/química , NADP/química , Trypanosoma cruzi/química , Secuencia de Aminoácidos , Cristalización , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Leishmania major/genética , Datos de Secuencia Molecular , NADP/genética , Estructura Secundaria de Proteína , Trypanosoma cruzi/genética
4.
PLoS One ; 10(4): e0125010, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25909780

RESUMEN

Crystallization of a maltose-binding protein MCL1 fusion has yielded a robust crystallography platform that generated the first apo MCL1 crystal structure, as well as five ligand-bound structures. The ability to obtain fragment-bound structures advances structure-based drug design efforts that, despite considerable effort, had previously been intractable by crystallography. In the ligand-independent crystal form we identify inhibitor binding modes not observed in earlier crystallographic systems. This MBP-MCL1 construct dramatically improves the structural understanding of well-validated MCL1 ligands, and will likely catalyze the structure-based optimization of high affinity MCL1 inhibitors.


Asunto(s)
Proteínas de Unión a Maltosa/química , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/química , Apoproteínas/química , Apoproteínas/genética , Cristalización , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Ligandos , Proteínas de Unión a Maltosa/genética , Modelos Moleculares , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Unión Proteica , Conformación Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética
5.
Tuberculosis (Edinb) ; 95(2): 142-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25613812

RESUMEN

High-resolution three-dimensional structures of essential Mycobacterium tuberculosis (Mtb) proteins provide templates for TB drug design, but are available for only a small fraction of the Mtb proteome. Here we evaluate an intra-genus "homolog-rescue" strategy to increase the structural information available for TB drug discovery by using mycobacterial homologs with conserved active sites. Of 179 potential TB drug targets selected for x-ray structure determination, only 16 yielded a crystal structure. By adding 1675 homologs from nine other mycobacterial species to the pipeline, structures representing an additional 52 otherwise intractable targets were solved. To determine whether these homolog structures would be useful surrogates in TB drug design, we compared the active sites of 106 pairs of Mtb and non-TB mycobacterial (NTM) enzyme homologs with experimentally determined structures, using three metrics of active site similarity, including superposition of continuous pharmacophoric property distributions. Pair-wise structural comparisons revealed that 19/22 pairs with >55% overall sequence identity had active site Cα RMSD <1 Å, >85% side chain identity, and ≥80% PSAPF (similarity based on pharmacophoric properties) indicating highly conserved active site shape and chemistry. Applying these results to the 52 NTM structures described above, 41 shared >55% sequence identity with the Mtb target, thus increasing the effective structural coverage of the 179 Mtb targets over three-fold (from 9% to 32%). The utility of these structures in TB drug design can be tested by designing inhibitors using the homolog structure and assaying the cognate Mtb enzyme; a promising test case, Mtb cytidylate kinase, is described. The homolog-rescue strategy evaluated here for TB is also generalizable to drug targets for other diseases.


Asunto(s)
Antituberculosos/farmacología , Diseño de Fármacos , Terapia Molecular Dirigida/métodos , Mycobacterium tuberculosis/efectos de los fármacos , Antituberculosos/química , Proteínas Bacterianas/química , Biología Computacional/métodos , Cristalografía por Rayos X/métodos , Bases de Datos de Proteínas , Activación Enzimática , Genómica/métodos , Humanos , Modelos Moleculares , Mycobacterium/clasificación , Mycobacterium/enzimología , Mycobacterium/genética , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/genética , Relación Estructura-Actividad Cuantitativa , Especificidad de la Especie
6.
ACS Med Chem Lett ; 5(12): 1308-12, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25516789

RESUMEN

A direct binding screen of 100 000 sp(3)-rich molecules identified a single diastereomer of a macrolactam core that binds specifically to myeloid cell leukemia 1 (MCL1). A comprehensive toolbox of biophysical methods was applied to validate the original hit and subsequent analogues and also established a binding mode competitive with NOXA BH3 peptide. X-ray crystallography of ligand bound to MCL1 reveals a remarkable ligand/protein shape complementarity that diverges from previously disclosed MCL1 inhibitor costructures.

7.
Sci Rep ; 4: 5944, 2014 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-25089892

RESUMEN

Influenza A viruses cause the respiratory illness influenza, which can be mild to fatal depending on the strain and host immune response. The flu polymerase acidic (PA), polymerase basic 1 (PB1), and polymerase basic 2 (PB2) proteins comprise the RNA-dependent RNA polymerase complex responsible for viral genome replication. The first crystal structures of the C-terminal domain of PA (PA-CTD) in the absence of PB1-derived peptides show a number of structural changes relative to the previously reported PB1-peptide bound structures. The human A/WSN/1933 (H1N1) and avian A/Anhui1/2013 (H7N9) strain PA-CTD proteins exhibit the same global topology as other strains in the absence of PB1, but differ extensively in the PB1 binding pocket including a widening of the binding groove and the unfolding of a ß-turn. Both PA-CTD proteins exhibited a significant increase in thermal stability in the presence of either a PB1-derived peptide or a previously reported inhibitor in differential scanning fluorimetry assays. These structural changes demonstrate plasticity in the PA-PB1 binding interface which may be exploited in the development of novel therapeutics.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/química , Subtipo H7N9 del Virus de la Influenza A/química , ARN Polimerasa Dependiente del ARN/química , Proteínas Virales/química , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Estabilidad de Enzimas , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Subtipo H1N1 del Virus de la Influenza A/enzimología , Subtipo H7N9 del Virus de la Influenza A/enzimología , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/fisiología
8.
J Vis Exp ; (76)2013 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-23851357

RESUMEN

Pandemic outbreaks of highly virulent influenza strains can cause widespread morbidity and mortality in human populations worldwide. In the United States alone, an average of 41,400 deaths and 1.86 million hospitalizations are caused by influenza virus infection each year (1). Point mutations in the polymerase basic protein 2 subunit (PB2) have been linked to the adaptation of the viral infection in humans (2). Findings from such studies have revealed the biological significance of PB2 as a virulence factor, thus highlighting its potential as an antiviral drug target. The structural genomics program put forth by the National Institute of Allergy and Infectious Disease (NIAID) provides funding to Emerald Bio and three other Pacific Northwest institutions that together make up the Seattle Structural Genomics Center for Infectious Disease (SSGCID). The SSGCID is dedicated to providing the scientific community with three-dimensional protein structures of NIAID category A-C pathogens. Making such structural information available to the scientific community serves to accelerate structure-based drug design. Structure-based drug design plays an important role in drug development. Pursuing multiple targets in parallel greatly increases the chance of success for new lead discovery by targeting a pathway or an entire protein family. Emerald Bio has developed a high-throughput, multi-target parallel processing pipeline (MTPP) for gene-to-structure determination to support the consortium. Here we describe the protocols used to determine the structure of the PB2 subunit from four different influenza A strains.


Asunto(s)
ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , Proteínas Virales/química , Proteínas Virales/genética , Cristalografía por Rayos X , Genómica/métodos , Subtipo H1N1 del Virus de la Influenza A/química , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H3N2 del Virus de la Influenza A/química , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Subtipo H5N1 del Virus de la Influenza A/química , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Modelos Moleculares , Estructura Secundaria de Proteína , Subunidades de Proteína
9.
PLoS One ; 8(1): e53851, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23382856

RESUMEN

BACKGROUND: The genus Burkholderia includes pathogenic gram-negative bacteria that cause melioidosis, glanders, and pulmonary infections of patients with cancer and cystic fibrosis. Drug resistance has made development of new antimicrobials critical. Many approaches to discovering new antimicrobials, such as structure-based drug design and whole cell phenotypic screens followed by lead refinement, require high-resolution structures of proteins essential to the parasite. METHODOLOGY/PRINCIPAL FINDINGS: We experimentally identified 406 putative essential genes in B. thailandensis, a low-virulence species phylogenetically similar to B. pseudomallei, the causative agent of melioidosis, using saturation-level transposon mutagenesis and next-generation sequencing (Tn-seq). We selected 315 protein products of these genes based on structure-determination criteria, such as excluding very large and/or integral membrane proteins, and entered them into the Seattle Structural Genomics Center for Infection Disease (SSGCID) structure determination pipeline. To maximize structural coverage of these targets, we applied an "ortholog rescue" strategy for those producing insoluble or difficult to crystallize proteins, resulting in the addition of 387 orthologs (or paralogs) from seven other Burkholderia species into the SSGCID pipeline. This structural genomics approach yielded structures from 31 putative essential targets from B. thailandensis, and 25 orthologs from other Burkholderia species, yielding an overall structural coverage for 49 of the 406 essential gene families, with a total of 88 depositions into the Protein Data Bank. Of these, 25 proteins have properties of a potential antimicrobial drug target i.e., no close human homolog, part of an essential metabolic pathway, and a deep binding pocket. We describe the structures of several potential drug targets in detail. CONCLUSIONS/SIGNIFICANCE: This collection of structures, solubility and experimental essentiality data provides a resource for development of drugs against infections and diseases caused by Burkholderia. All expression clones and proteins created in this study are freely available by request.


Asunto(s)
Infecciones por Burkholderia/genética , Burkholderia pseudomallei/genética , Genómica , Redes y Vías Metabólicas/genética , Infecciones por Burkholderia/tratamiento farmacológico , Burkholderia pseudomallei/patogenicidad , Biología Computacional , Bases de Datos de Proteínas , Diseño de Fármacos , Genes Esenciales , Genoma Bacteriano , Humanos , Filogenia , Conformación Proteica
10.
Proc Natl Acad Sci U S A ; 109(25): 9857-62, 2012 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-22679291

RESUMEN

Bacterial pathogens are becoming increasingly resistant to antibiotics. As an alternative therapeutic strategy, phage therapy reagents containing purified viral lysins have been developed against gram-positive organisms but not against gram-negative organisms due to the inability of these types of drugs to cross the bacterial outer membrane. We solved the crystal structures of a Yersinia pestis outer membrane transporter called FyuA and a bacterial toxin called pesticin that targets this transporter. FyuA is a ß-barrel membrane protein belonging to the family of TonB dependent transporters, whereas pesticin is a soluble protein with two domains, one that binds to FyuA and another that is structurally similar to phage T4 lysozyme. The structure of pesticin allowed us to design a phage therapy reagent comprised of the FyuA binding domain of pesticin fused to the N-terminus of T4 lysozyme. This hybrid toxin kills specific Yersinia and pathogenic E. coli strains and, importantly, can evade the pesticin immunity protein (Pim) giving it a distinct advantage over pesticin. Furthermore, because FyuA is required for virulence and is more common in pathogenic bacteria, the hybrid toxin also has the advantage of targeting primarily disease-causing bacteria rather than indiscriminately eliminating natural gut flora.


Asunto(s)
Bacteriófagos/metabolismo , Bacterias Gramnegativas/virología , Mucoproteínas/metabolismo , Proteínas Bacterianas/química , Bacteriocinas/química , Bacteriófagos/fisiología , Membrana Celular/metabolismo , Microscopía por Crioelectrón , Modelos Moleculares , Mucoproteínas/química , Conformación Proteica , Ingeniería de Proteínas , Transporte de Proteínas , Receptores de Superficie Celular/química
11.
Structure ; 20(7): 1233-43, 2012 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-22658748

RESUMEN

Intimins and invasins are virulence factors produced by pathogenic Gram-negative bacteria. They contain C-terminal extracellular passenger domains that are involved in adhesion to host cells and N-terminal ß domains that are embedded in the outer membrane. Here, we identify the domain boundaries of an E. coli intimin ß domain and use this information to solve its structure and the ß domain structure of a Y. pseudotuberculosis invasin. Both ß domain structures crystallized as monomers and reveal that the previous range of residues assigned to the ß domain also includes a protease-resistant domain that is part of the passenger. Additionally, we identify 146 nonredundant representative members of the intimin/invasin family based on the boundaries of the highly conserved intimin and invasin ß domains. We then use this set of sequences along with our structural data to find and map the evolutionarily constrained residues within the ß domain.


Asunto(s)
Adhesinas Bacterianas/química , Escherichia coli Enterohemorrágica/química , Proteínas de Escherichia coli/química , Proteínas Recombinantes de Fusión/química , Yersinia pseudotuberculosis/química , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Secuencia de Aminoácidos , Adhesión Bacteriana , Secuencia Conservada , Cristalografía por Rayos X , Escherichia coli Enterohemorrágica/metabolismo , Escherichia coli Enterohemorrágica/patogenicidad , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Evolución Molecular , Modelos Moleculares , Datos de Secuencia Molecular , Plásmidos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Virulencia/química , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/patogenicidad
12.
Curr Opin Struct Biol ; 21(4): 523-31, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21719274

RESUMEN

The outer membranes of Gram-negative bacteria, mitochondria, and chloroplasts all contain transmembrane ß-barrel proteins. These ß-barrel proteins serve essential functions in cargo transport and signaling and are also vital for membrane biogenesis. They have also been adapted to perform a diverse set of important cellular functions including acting as porins, transporters, enzymes, virulence factors and receptors. Recent structures of transmembrane ß-barrels include that of a full length autotransporter (EstA), a bacterial heme transporter complex (HasR), a bacterial porin in complex with several ligands (PorB), and the mitochondrial voltage-dependent anion channel (VDAC) from both mouse and human. These represent only a few of the interesting structures of ß-barrel membrane proteins recently elucidated. However, they demonstrate many of the advancements made within the field of transmembrane protein structure in the past few years.


Asunto(s)
Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Animales , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Cristalografía por Rayos X , Humanos , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Estructura Secundaria de Proteína
13.
J Mol Biol ; 407(2): 248-60, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21277859

RESUMEN

Escherichia coli BamB is the largest of four lipoproteins in the ß-barrel assembly machinery (BAM) complex. It interacts with the periplasmic domain of BamA, an integral outer membrane protein (OMP) essential for OMP biogenesis. Although BamB is not essential, it serves an important function in the BAM complex, significantly increasing the folding efficiency of some OMPs in vivo and in vitro. To learn more about the BAM complex, we solved structures of BamB in three different crystal forms. BamB crystallized in space groups P2(1)3, I222, and P2(1)2(1)2(1), with one molecule per asymmetric unit in each case. Crystals from the space group I222 diffracted to 1. 65-Å resolution. BamB forms an eight-bladed ß-propeller with a central pore and is shaped like a doughnut. A DALI search revealed that BamB shares structural homology to several eukaryotic proteins containing WD40 repeat domains, which commonly have ß-propeller folds and often serve as scaffolding proteins within larger multi-protein complexes that carry out signal transduction, cell division, and chemotaxis. Using mutagenesis data from previous studies, we docked BamB onto a BamA structural model and assessed known and possible interactions between these two proteins. Our data suggest that BamB serves as a scaffolding protein within the BAM complex by optimally orienting the flexible periplasmic domain of BamA for interaction with other BAM components and chaperones. This may facilitate integration of newly synthesized OMPs into the outer membrane.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Proteínas de Escherichia coli/química , Lipoproteínas/metabolismo , Secuencia de Aminoácidos , Proteínas de la Membrana Bacteriana Externa/metabolismo , Cristalografía , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Peso Molecular , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
14.
Pharmaceuticals (Basel) ; 4(10): 1328-1354, 2011 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-23115527

RESUMEN

Ribonucleotide reductase (RR) is a crucial enzyme in de novo DNA synthesis, where it catalyses the rate determining step of dNTP synthesis. RRs consist of a large subunit called RR1 (α), that contains two allosteric sites and one catalytic site, and a small subunit called RR2 (ß), which houses a tyrosyl free radical essential for initiating catalysis. The active form of mammalian RR is an α(n)ß(m) hetero oligomer. RR inhibitors are cytotoxic to proliferating cancer cells. In this brief review we will discuss the three classes of RR, the catalytic mechanism of RR, the regulation of the dNTP pool, the substrate selection, the allosteric activation, inactivation by ATP and dATP, and the nucleoside drugs that target RR. We will also discuss possible strategies for developing a new class of drugs that disrupts the RR assembly.

15.
J Med Chem ; 51(15): 4653-9, 2008 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-18610997

RESUMEN

Eukaryotic ribonucleotide reductase (RR) catalyzes nucleoside diphosphate conversion to deoxynucleoside diphosphate. Crucial for rapidly dividing cells, RR is a target for cancer therapy. RR activity requires formation of a complex between subunits R1 and R2 in which the R2 C-terminal peptide binds to R1. Here we report crystal structures of heterocomplexes containing mammalian R2 C-terminal heptapeptide, P7 (Ac-1FTLDADF7) and its peptidomimetic P6 (1Fmoc(Me)PhgLDChaDF7) bound to Saccharomyces cerevisiae R1 (ScR1). P7 and P6, both of which inhibit ScRR, each bind at two contiguous sites containing residues that are highly conserved among eukaryotes. Such binding is quite distinct from that reported for prokaryotes. The Fmoc group in P6 peptide makes several hydrophobic interactions that contribute to its enhanced potency in binding to ScR1. Combining all of our results, we observe three distinct conformations for peptide binding to ScR1. These structures provide pharmacophores for designing highly potent nonpeptide class I RR inhibitors.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Péptidos/química , Péptidos/farmacología , Ribonucleótido Reductasas/antagonistas & inhibidores , Ribonucleótido Reductasas/metabolismo , Saccharomyces cerevisiae/enzimología , Animales , Cristalografía por Rayos X , Modelos Moleculares , Estructura Molecular , Unión Proteica , Ribonucleótido Reductasas/química , Saccharomyces cerevisiae/efectos de los fármacos
16.
Proc Natl Acad Sci U S A ; 103(11): 4022-7, 2006 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-16537479

RESUMEN

Ribonucleotide reductase catalyzes a crucial step in de novo DNA synthesis and is allosterically controlled by relative levels of dNTPs to maintain a balanced pool of deoxynucleoside triphosphates in the cell. In eukaryotes, the enzyme comprises a heterooligomer of alpha(2) and beta(2) subunits. The alpha subunit, Rnr1, contains catalytic and regulatory sites. Here, we report the only x-ray structures of the eukaryotic alpha subunit of ribonucleotide reductase from Saccharomyces cerevisiae. The structures of the apo-, AMPPNP only-, AMPPNP-CDP-, AMPPNP-UDP-, dGTP-ADP- and TTP-GDP-bound complexes give insight into substrate and effector binding and specificity cross-talk. These are Class I structures with the only fully ordered catalytic sites, including loop 2, a stretch of polypeptide that spans specificity and catalytic sites, conferring specificity. Binding of specificity effector rearranges loop 2; in our structures, this rearrangement moves P294, a residue unique to eukaryotes, out of the catalytic site, accommodating substrate binding. Substrate binding further rearranges loop 2. Cross-talk, by which effector binding regulates substrate preference, occurs largely through R293 and Q288 of loop 2, which are analogous to residues in Thermotoga maritima that mediate cross-talk. However loop-2 conformations and residue-substrate interactions differ substantially between yeast and T. maritima. In most effector-substrate complexes, water molecules help mediate substrate-loop 2 interactions. Finally, the substrate ribose binds with its 3' hydroxyl closer than its 2' hydroxyl to C218 of the catalytic redox pair. We also see a conserved water molecule at the catalytic site in all our structures, near the ribose 2' hydroxyl.


Asunto(s)
Desoxirribonucleótidos/metabolismo , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Modelos Moleculares , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/enzimología , Especificidad por Sustrato , Thermotoga maritima/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...