Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.740
Filtrar
1.
DNA Repair (Amst) ; 139: 103693, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38776712

RESUMEN

MutT proteins belong to the Nudix hydrolase superfamily that includes a diverse group of Mg2+ requiring enzymes. These proteins use a generalized substrate, nucleoside diphosphate linked to a chemical group X (NDP-X), to produce nucleoside monophosphate (NMP) and the moiety X linked with phosphate (XP). E. coli MutT (EcoMutT) and mycobacterial MutT1 (MsmMutT1) belong to the Nudix hydrolase superfamily that utilize 8-oxo-(d)GTP (referring to both 8-oxo-GTP or 8-oxo-dGTP). However, predominant products of their activities are different. While EcoMutT produces 8-oxo-(d)GMP, MsmMutT1 gives rise to 8-oxo-(d)GDP. Here, we show that the altered cleavage specificities of the two proteins are largely a consequence of the variation at the equivalent of Gly37 (G37) in EcoMutT to Lys (K65) in the MsmMutT1. Remarkably, mutations of G37K (EcoMutT) and K65G (MsmMutT1) switch their cleavage specificities to produce 8-oxo-(d)GDP, and 8-oxo-(d)GMP, respectively. Further, a time course analysis using 8-oxo-GTP suggests that MsmMutT1(K65G) hydrolyses 8-oxo-(d)GTP to 8-oxo-(d)GMP in a two-step reaction via 8-oxo-(d)GDP intermediate. Expectedly, unlike EcoMutT (G37K) and MsmMutT1, EcoMutT and MsmMutT1 (K65G) rescue an E. coli ΔmutT strain, better by decreasing A to C mutations.


Asunto(s)
Nucleótidos de Desoxiguanina , Proteínas de Escherichia coli , Escherichia coli , Mycobacterium smegmatis , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/química , Mycobacterium smegmatis/enzimología , Mycobacterium smegmatis/metabolismo , Mycobacterium smegmatis/genética , Especificidad por Sustrato , Nucleótidos de Desoxiguanina/metabolismo , Escherichia coli/metabolismo , Escherichia coli/genética , Escherichia coli/enzimología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Sustitución de Aminoácidos , Pirofosfatasas/metabolismo , Pirofosfatasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Guanosina Trifosfato/metabolismo , Guanosina Trifosfato/análogos & derivados
2.
Anal Biochem ; 631: 114338, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34433016

RESUMEN

The Ras superfamily of small G proteins play central roles in diverse signaling pathways. Superfamily members act as molecular on-off switches defined by their occupancy with GTP or GDP, respectively. In vitro functional studies require loading with a hydrolysis-resistant GTP analogue to increase the on-state lifetime, as well as knowledge of fractional loading with activating and inactivating nucleotides. The present study describes a method combining elements of previous approaches with new, optimized features to analyze the bound nucleotide composition of a G protein loaded with activating (GMPPNP) or inactivating (GDP) nucleotide. After nucleotide loading, the complex is washed to remove unbound nucleotides then bound nucleotides are heat-extracted and subjected to ion-paired, reverse-phase HPLC-UV to resolve, identify and quantify the individual nucleotide components. These data enable back-calculation to the nucleotide composition and fractional activation of the original, washed G protein population prior to heat extraction. The method is highly reproducible. Application to multiple HRas preparations and mutants confirms its ability to fully extract and analyze bound nucleotides, and to resolve the fractional on- and off-state populations. Furthermore, the findings yield a novel hypothesis for the molecular disease mechanism of Ras mutations at the E63 and Y64 positions.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Nucleótidos de Guanina/análisis , Nucleótidos de Guanina/metabolismo , Proteínas ras/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/metabolismo , Calor , Hidrólisis , Mutación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reproducibilidad de los Resultados , Rayos Ultravioleta , Proteínas ras/genética
3.
Methods Mol Biol ; 2302: 37-48, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33877621

RESUMEN

The complex of G protein-coupled receptors (GPCR) and G proteins is the core assembly in GPCR signaling in eukaryotes. With the recent development of cryo-electron microscopy, there has been a rapid growth in structures of GPCR-G protein complexes solved to near-atomic resolution, giving important insights into this signaling complex. Here we describe the biochemical protocol to study the interaction between GPCRs and G proteins before preparation of GPCR-G protein complexes for structural studies. We use gel filtration to analyze the binding properties between GPCR and G protein with the presence of agonist or antagonist, as well as the complex dissociation in the presence of GTP analogue. Methods used in the protocol are affinity purification and gel filtration, which are also commonly used in protein sample preparation for structural work. Therefore, the protocol can be easily adapted for large-scale sample preparation.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Guanosina Trifosfato/análogos & derivados , Receptores Acoplados a Proteínas G/metabolismo , Membrana Celular/metabolismo , Cromatografía en Gel , Proteínas de Unión al GTP/química , Células HEK293 , Humanos , Unión Proteica , Receptores Acoplados a Proteínas G/química , Transducción de Señal
4.
RNA ; 27(6): 665-675, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33758037

RESUMEN

Human tRNAHis guanylyltransferase (HsThg1) catalyzes the 3'-5' addition of guanosine triphosphate (GTP) to the 5'-end (-1 position) of tRNAHis, producing mature tRNAHis In human cells, cytoplasmic and mitochondrial tRNAHis have adenine (A) or cytidine (C), respectively, opposite to G-1 Little attention has been paid to the structural requirements of incoming GTP in 3'-5' nucleotidyl addition by HsThg1. In this study, we evaluated the incorporation efficiencies of various GTP analogs by HsThg1 and compared the reaction mechanism with that of Candida albicans Thg1 (CaThg1). HsThg1 incorporated GTP opposite A or C in the template most efficiently. In contrast to CaThg1, HsThg1 could incorporate UTP opposite A, and guanosine diphosphate (GDP) opposite C. These results suggest that HsThg1 could transfer not only GTP, but also other NTPs, by forming Watson-Crick (WC) hydrogen bonds between the incoming NTP and the template base. On the basis of the molecular mechanism, HsThg1 succeeded in labeling the 5'-end of tRNAHis with biotinylated GTP. Structural analysis of HsThg1 was also performed in the presence of the mitochondrial tRNAHis Structural comparison of HsThg1 with other Thg1 family enzymes suggested that the structural diversity of the carboxy-terminal domain of the Thg1 enzymes might be involved in the formation of WC base-pairing between the incoming GTP and template base. These findings provide new insights into an unidentified biological function of HsThg1 and also into the applicability of HsThg1 to the 5'-terminal modification of RNAs.


Asunto(s)
Guanosina Trifosfato/metabolismo , Nucleotidiltransferasas/metabolismo , Biotinilación , Candida albicans/enzimología , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/análogos & derivados , Humanos , Methanosarcina/enzimología , Mitocondrias/enzimología , Modelos Moleculares , Nucleótidos/metabolismo , Nucleotidiltransferasas/química , ARN de Transferencia de Histidina/metabolismo
5.
Chembiochem ; 22(6): 1079-1083, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33140496

RESUMEN

The millisecond timescale dynamics of activated Ras transiently sample a low-populated conformational state that has distinct surface property from the major state and represents a promising target for binding of small-molecule compounds. To avoid the complications of hydrolysis, dynamics and other properties of active Ras have so far been routinely investigated by using non-hydrolyzable GTP analogues, which, however, were previously reported to alter both the kinetics and distribution of the conformational exchange. In this study, we quantitatively measured and validated the internal dynamics of Ras complexed with a slowly hydrolyzable GTP analogue, GTPγS, which increases the lifetime of active Ras by 23 times relative to that of native GTP. It was found that GTPγS, in addition to its better mimicking of the exchange kinetics than the commonly used non-hydrolyzable analogues GppNHp and GppCH2 p, can rigorously reproduce the natural dynamics network in active Ras, thus indicating its fitness for use in the development of allosteric inhibitors.


Asunto(s)
Guanosina Trifosfato/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina Trifosfato/análogos & derivados , Humanos , Hidrólisis , Cinética , Espectroscopía de Resonancia Magnética , Mutagénesis Sitio-Dirigida , Proteínas Proto-Oncogénicas p21(ras)/genética
6.
Sci Rep ; 10(1): 13661, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32788644

RESUMEN

Dynamic instability of microtubules is characterized by stochastically alternating phases of growth and shrinkage and is hypothesized to be controlled by the conformation and nucleotide state of tubulin dimers within the microtubule lattice. Specifically, conformation changes (compression) in the tubulin dimer following the hydrolysis of GTP have been suggested to generate stress and drive depolymerization. In the present study, molecular dynamics simulations were used in tandem with in vitro experiments to investigate changes in depolymerization based on the presence of islands of uncompressed (GMPCPP) dimers in the microtubule lattice. Both methods revealed an exponential decay in the kinetic rate of depolymerization corresponding to the relative level of uncompressed (GMPCPP) dimers, beginning at approximately 20% incorporation. This slowdown was accompanied by a distinct morphological change from unpeeling "ram's horns" to blunt-ended dissociation at the microtubule end. Collectively these data demonstrated that islands of uncompressed dimers can alter the mechanism and kinetics of depolymerization in a manner consistent with promoting rescue events.


Asunto(s)
Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Animales , Hidrólisis , Cinética , Simulación de Dinámica Molecular , Polimerizacion , Multimerización de Proteína , Porcinos
7.
FEBS J ; 287(3): 496-514, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31330084

RESUMEN

Human guanylate-binding protein 1 (hGBP1) belongs to the family of dynamin-like proteins and is activated by addition of nucleotides, leading to protein oligomerization and stimulated GTPase activity. In vivo, hGBP1 is post-translationally modified by attachment of a farnesyl group yielding farn-hGBP1. In this study, hydrodynamic differences in farn-hGBP1 and unmodified hGBP1 were investigated using dynamic light scattering (DLS), analytical ultracentrifugation (AUC) and analytical size-exclusion chromatography (SEC). In addition, we performed small-angle X-ray scattering (SAXS) experiments coupled with a SEC setup (SEC-SAXS) to investigate structural properties of nonmodified hGBP1 and farn-hGBP1 in solution. SEC-SAXS measurements revealed that farnesylation keeps hGBP1 in its inactive monomeric and crystal-like conformation in nucleotide-free solution, whereas unmodified hGBP1 forms a monomer-dimer equilibrium both in the inactive ground state in nucleotide-free solution as well as in the activated state that is trapped by addition of the nonhydrolysable GTP analogue GppNHp. Nonmodified hGBP1 is structurally perturbed as compared to farn-hGBP. In particular, GppNHp binding leads to large structural rearrangements and higher conformational flexibility of the monomer and the dimer. Structural changes observed in the nonmodified protein are prerequisites for further oligomer assemblies of farn-hGBP1 that occur in the presence of nucleotides. DATABASE: All SEC-SAXS data, corresponding fits to the data and structural models are deposited in the Small Angle Scattering Biological Data Bank [SASBDB (Nucleic Acids Res, 43, 2015, D357)] with project IDs: SASDEE8, SASDEF8, SASDEG8, SASDEH8, SASDEJ8, SASDEK8, SASDEL8 and SASDEM8.


Asunto(s)
Proteínas de Unión al GTP/química , Prenilación , Multimerización de Proteína , Cromatografía , Dispersión Dinámica de Luz , Proteínas de Unión al GTP/metabolismo , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/metabolismo , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Dispersión del Ángulo Pequeño , Difracción de Rayos X
8.
Sci Rep ; 9(1): 13910, 2019 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-31558768

RESUMEN

DNA polymerases often incorporate non-canonical nucleotide, i.e., ribonucleoside triphosphates into the genomic DNA. Aberrant accumulation of ribonucleotides in the genome causes various cellular abnormalities. Here, we show the possible role of human nucleotide excision repair (NER) and DNA polymerase η (Pol η) in processing of a single ribonucleotide embedded into DNA. We found that the reconstituted NER system can excise the oxidized ribonucleotide on the plasmid DNA. Taken together with the evidence that Pol η accurately bypasses a ribonucleotide, i.e., riboguanosine (rG) or its oxidized derivative (8-oxo-rG) in vitro, we further assessed the mutagenic potential of the embedded ribonucleotide in human cells lacking NER or Pol η. A single rG on the supF reporter gene predominantly induced large deletion mutations. An embedded 8-oxo-rG caused base substitution mutations at the 3'-neighboring base rather than large deletions in wild-type cells. The disruption of XPA, an essential factor for NER, or Pol η leads to the increased mutant frequency of 8-oxo-rG. Furthermore, the frequency of 8-oxo-rG-mediated large deletions was increased by the loss of Pol η, but not XPA. Collectively, our results suggest that base oxidation of the embedded ribonucleotide enables processing of the ribonucleotide via alternative DNA repair and damage tolerance pathways.


Asunto(s)
Reparación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Guanosina Trifosfato/análogos & derivados , Línea Celular Tumoral , ADN Polimerasa Dirigida por ADN/genética , Guanosina Trifosfato/metabolismo , Humanos , Proteína de la Xerodermia Pigmentosa del Grupo A/genética , Proteína de la Xerodermia Pigmentosa del Grupo A/metabolismo
9.
Chemistry ; 25(27): 6728-6740, 2019 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-30801798

RESUMEN

The m7 G cap is a unique nucleotide structure at the 5'-end of all eukaryotic mRNAs. The cap specifically interacts with numerous cellular proteins and participates in biological processes that are essential for cell growth and function. To provide small molecular probes to study important cap-recognizing proteins, we synthesized m7 G nucleotides labeled with fluorescent tags via the terminal phosph(on)ate group and studied how their emission properties changed upon protein binding or enzymatic cleavage. Only the pyrene-labeled compounds behaved as sensitive turn-on probes. A pyrene-labeled m7 GTP analogue showed up to eightfold enhanced fluorescence emission upon binding to eukaryotic translation initiation factor 4E (eIF4E) and over 30-fold enhancement upon cleavage by decapping scavenger (DcpS) enzyme. These observations served as the basis for developing binding- and hydrolytic-activity assays. The assay utility was validated with previously characterized libraries of eIF4E ligands and DcpS inhibitors. The DcpS assay was also applied to study hydrolytic activity and inhibition of endogenous enzyme in cytoplasmic extracts from HeLa and HEK cells.


Asunto(s)
Endorribonucleasas/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Colorantes Fluorescentes/química , Animales , Endorribonucleasas/antagonistas & inhibidores , Endorribonucleasas/genética , Factor 4E Eucariótico de Iniciación/química , Factor 4E Eucariótico de Iniciación/genética , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/metabolismo , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/síntesis química , Guanosina Trifosfato/metabolismo , Células HEK293 , Células HeLa , Humanos , Hidrólisis , Concentración 50 Inhibidora , Cinética , Ligandos , Ratones , Unión Proteica , Pirenos/química , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Espectrometría de Fluorescencia
10.
Neuropharmacology ; 148: 347-357, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30710569

RESUMEN

Spinal cord injury results in sensation dysfunction. This study explored miR-142-3p, which acts a critical role in sciatic nerve conditioning injury (SNCI) promoting the repair of the dorsal column injury and validated its function on primary sensory neuron(DRG). miR-142-3p expression increased greatly in the spinal cord dorsal column lesion (SDCL) group and increased slightly in the SNCI group. Subsequently, the expression of adenylate cyclase 9 (AC9), the target gene of miR-142-3p, declined sharply in the SDCL group and declined limitedly in the SNCI group. The expression trend of cAMP was opposite to that of miR-142-3p. MiR-142-3p inhibitor improved the axon length, upregulated the expression of AC9, cAMP, p-CREB, IL-6, and GAP43, and downregulated the expression of GTP-RhoA. miR-142-3p inhibitor combined with AC9 siRNA showed shorter axon length, the expression of AC9, cAMP, p-CREB, IL-6, and GAP43 was decreased, and the expression of GTP-RhoA was increased. H89 and AG490, inhibitors of cAMP/PKA pathway and IL6/STAT3/GAP43 axis, respectively, declined the enhanced axonal growth by miR-142-3p inhibitor and altered the expression level of the corresponding proteins. Thus, a substitution therapy using Sorafenib that downregulates the miR-142-3p expression for SNCI was investigated. The results showed the effect of Sorafenib was similar to that of miR-142-3p inhibitor and SNCI on both axon growth in vitro and sensory conduction function recovery in vivo. In conclusion, miR-142-3p acts a pivotal role in SNCI promoting the repair of dorsal column injury. Sorafenib mimics the treatment effect of SNCI via downregulation of miR-142-3p, subsequently, promoting sensory conduction function recovery post dorsal column injury.


Asunto(s)
Adenilil Ciclasas/fisiología , AMP Cíclico/fisiología , MicroARNs/fisiología , Sensación/efectos de los fármacos , Sorafenib/farmacología , Traumatismos de la Médula Espinal/fisiopatología , Adenilil Ciclasas/biosíntesis , Animales , AMP Cíclico/biosíntesis , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Proteína GAP-43/biosíntesis , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/biosíntesis , Interleucina-6/biosíntesis , Isoquinolinas/farmacología , MicroARNs/antagonistas & inhibidores , MicroARNs/biosíntesis , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Ratas , Recuperación de la Función/efectos de los fármacos , Rodaminas , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Transducción de Señal/efectos de los fármacos , Traumatismos de la Médula Espinal/metabolismo , Sulfonamidas/farmacología , Tirfostinos/farmacología , Regulación hacia Arriba/efectos de los fármacos
11.
Nucleic Acids Res ; 46(20): 10589-10597, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30289487

RESUMEN

A previously developed RNA polymerase ribozyme uses nucleoside triphosphates (NTPs) to extend a primer 3'-terminus, templated by an RNA template with good fidelity, forming 3'-5'-phosphordiester bonds. Indirect evidence has suggested that the ribozyme's accessory domain binds the NTP with a highly conserved purine-rich loop. To determine the NTP binding site more precisely we evolved the ribozyme for efficient use of 6-thio guanosine triphosphate (6sGTP). 6sGTP never appeared in the evolutionary history of the ribozyme, therefore it was expected that mutations would appear at the NTP binding site, adapting to more efficient binding of 6sGTP. Indeed, the evolution identified three mutations that mediate 200-fold improved incorporation kinetics for 6sGTP. A >50-fold effect resulted from mutation A156U in the purine-rich loop, identifying the NTP binding site. This mutation acted weakly cooperative with two other beneficial mutations, C113U in the P2 stem near the catalytic site, and C79U on the surface of the catalytic domain. The preference pattern of the ribozyme for different NTPs changed when position 156 was mutated, confirming a direct contact between position 156 and the NTP. The results suggest that A156 stabilizes the NTP in the active site by a hydrogen bond to the Hoogsteen face of the NTP.


Asunto(s)
Guanosina Trifosfato/análogos & derivados , Nucleósidos/química , Fosfatos/química , ARN Catalítico/química , ARN/química , Tionucleótidos/química , Sitios de Unión , Dominio Catalítico , ARN Polimerasas Dirigidas por ADN/química , Guanosina Trifosfato/química , Cinética , Modelos Moleculares , Mutación , Conformación de Ácido Nucleico , Nucleótidos/química , Purinas/química
12.
Nature ; 560(7717): 258-262, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30069048

RESUMEN

Membrane fission is a fundamental process in the regulation and remodelling of cell membranes. Dynamin, a large GTPase, mediates membrane fission by assembling around, constricting and cleaving the necks of budding vesicles1. Here we report a 3.75 Å resolution cryo-electron microscopy structure of the membrane-associated helical polymer of human dynamin-1 in the GMPPCP-bound state. The structure defines the helical symmetry of the dynamin polymer and the positions of its oligomeric interfaces, which were validated by cell-based endocytosis assays. Compared to the lipid-free tetramer form2, membrane-associated dynamin binds to the lipid bilayer with its pleckstrin homology domain (PHD) and self-assembles across the helical rungs via its guanine nucleotide-binding (GTPase) domain3. Notably, interaction with the membrane and helical assembly are accommodated by a severely bent bundle signalling element (BSE), which connects the GTPase domain to the rest of the protein. The BSE conformation is asymmetric across the inter-rung GTPase interface, and is unique compared to all known nucleotide-bound states of dynamin. The structure suggests that the BSE bends as a result of forces generated from the GTPase dimer interaction that are transferred across the stalk to the PHD and lipid membrane. Mutations that disrupted the BSE kink impaired endocytosis. We also report a 10.1 Å resolution cryo-electron microscopy map of a super-constricted dynamin polymer showing localized conformational changes at the BSE and GTPase domains, induced by GTP hydrolysis, that drive membrane constriction. Together, our results provide a structural basis for the mechanism of action of dynamin on the lipid membrane.


Asunto(s)
Biopolímeros/química , Biopolímeros/metabolismo , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Microscopía por Crioelectrón , Dinamina I/metabolismo , Dinamina I/ultraestructura , Biopolímeros/genética , Membrana Celular/química , Dinamina I/química , Dinamina I/genética , Endocitosis/genética , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Humanos , Hidrólisis , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestructura , Mutación , Dominios Proteicos , Multimerización de Proteína
13.
Nucleic Acids Res ; 46(16): 8641-8650, 2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-30107565

RESUMEN

According to the traditional view, GTPases act as molecular switches, which cycle between distinct 'on' and 'off' conformations bound to GTP and GDP, respectively. Translation elongation factor EF-Tu is a GTPase essential for prokaryotic protein synthesis. In its GTP-bound form, EF-Tu delivers aminoacylated tRNAs to the ribosome as a ternary complex. GTP hydrolysis is thought to cause the release of EF-Tu from aminoacyl-tRNA and the ribosome due to a dramatic conformational change following Pi release. Here, the crystal structure of Escherichia coli EF-Tu in complex with a non-hydrolysable GTP analogue (GDPNP) has been determined. Remarkably, the overall conformation of EF-Tu·GDPNP displays the classical, open GDP-bound conformation. This is in accordance with an emerging view that the identity of the bound guanine nucleotide is not 'locking' the GTPase in a fixed conformation. Using a single-molecule approach, the conformational dynamics of various ligand-bound forms of EF-Tu were probed in solution by fluorescence resonance energy transfer. The results suggest that EF-Tu, free in solution, may sample a wider set of conformations than the structurally well-defined GTP- and GDP-forms known from previous X-ray crystallographic studies. Only upon binding, as a ternary complex, to the mRNA-programmed ribosome, is the well-known, closed GTP-bound conformation, observed.


Asunto(s)
Escherichia coli/química , Guanosina Trifosfato/química , Factor Tu de Elongación Peptídica/química , Conformación Proteica , Cristalografía por Rayos X , Escherichia coli/genética , GTP Fosfohidrolasas/química , GTP Fosfohidrolasas/genética , Guanosina Difosfato/química , Guanosina Trifosfato/análogos & derivados , Factor Tu de Elongación Peptídica/genética , Biosíntesis de Proteínas/genética , ARN Mensajero/química , ARN Mensajero/genética , Ribosomas/química , Ribosomas/genética
14.
Structure ; 26(6): 810-820.e4, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29706533

RESUMEN

Structures of wild-type K-Ras from crystals obtained in the presence of guanosine triphosphate (GTP) or its analogs have remained elusive. Of the K-Ras mutants, only K-RasG12D and K-RasQ61H are available in the PDB representing the activated form of the GTPase not in complex with other proteins. We present the crystal structure of wild-type K-Ras bound to the GTP analog GppCH2p, with K-Ras in the state 1 conformation. Signatures of conformational states obtained by one-dimensional proton NMR confirm that K-Ras has a more substantial population of state 1 in solution than H-Ras, which predominantly favors state 2. The oncogenic mutant K-RasG12D favors state 2, changing the balance of conformational states in favor of interactions with effector proteins. Differences in the population of conformational states between K-Ras and H-Ras, as well as between K-Ras and its mutants, can provide a structural basis for focused targeting of the K-Ras isoform in cancer-specific strategies.


Asunto(s)
Mutación , Proteínas Proto-Oncogénicas p21(ras)/química , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/metabolismo , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Proteínas Proto-Oncogénicas p21(ras)/genética , Espectroscopía de Protones por Resonancia Magnética
15.
Biochim Biophys Acta Proteins Proteom ; 1866(5-6): 702-711, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29653192

RESUMEN

Nitric oxide sensitive guanylyl cyclase (NOsGC) is a heterodimeric enzyme consisting of one α and one ß subunit. Each subunit consists of four domains: the N-terminal heme-nitric oxide oxygen binding (HNOX) domain, a PAS domain, a coiled-coil domain and the C-terminal catalytic domain. Upon activation by the endogenous ligand NO or activating drugs, NOsGC catalyses the conversion of GTP to cGMP. Although several crystal structures of the isolated domains are known, the structure of the full-length enzyme and the interdomain conformational changes during activation remain unsolved to date. In the current study, we performed protein thermal shift assays of purified NOsGC to identify discrete conformational states amenable to further analysis e.g. by crystallisation. A non-hydrolysable substrate analogue binding to the catalytic domain led to a subtle change in melting temperature. An activator drug binding to the HNOX domain led to a small increase. However, the combination of substrate analogue and activator drug led to a marked synergistic increase from 51 °C to 60 °C. This suggests reciprocal communication between HNOX domain and catalytic domain and formation of a stable activated conformation amenable to further biophysical characterization.


Asunto(s)
Benzoatos/farmacología , Activadores de Enzimas/farmacología , Guanosina Trifosfato/farmacología , Hemo/metabolismo , Guanilil Ciclasa Soluble/metabolismo , Sitios de Unión , Dominio Catalítico , Cloruros/farmacología , GMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Activación Enzimática , Estabilidad de Enzimas , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/metabolismo , Hemo/química , Humanos , Compuestos de Manganeso/farmacología , Unión Proteica , Conformación Proteica , Desnaturalización Proteica , Guanilil Ciclasa Soluble/química , Guanilil Ciclasa Soluble/genética , Relación Estructura-Actividad , Temperatura de Transición
16.
Nucleic Acids Res ; 46(6): 3232-3244, 2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29471537

RESUMEN

Archaea and eukaryotes have ribosomal P stalks composed of anchor protein P0 and aP1 homodimers (archaea) or P1•P2 heterodimers (eukaryotes). These P stalks recruit translational GTPases to the GTPase-associated center in ribosomes to provide energy during translation. The C-terminus of the P stalk is known to selectively recognize GTPases. Here we investigated the interaction between the P stalk and elongation factor 2 by determining the structures of Pyrococcus horikoshii EF-2 (PhoEF-2) in the Apo-form, GDP-form, GMPPCP-form (GTP-form), and GMPPCP-form bound with 11 C-terminal residues of P1 (P1C11). Helical structured P1C11 binds to a hydrophobic groove between domain G and subdomain G' of PhoEF-2, where is completely different from that of aEF-1α in terms of both position and sequence, implying that such interaction characteristic may be requested by how GTPases perform their functions on the ribosome. Combining PhoEF-2 P1-binding assays with a structural comparison of current PhoEF-2 structures and molecular dynamics model of a P1C11-bound GDP form, the conformational changes of the P1C11-binding groove in each form suggest that in response to the translation process, the groove has three states: closed, open, and release for recruiting and releasing GTPases.


Asunto(s)
Proteínas Arqueales/metabolismo , Factor 2 de Elongación Peptídica/metabolismo , Pyrococcus horikoshii/metabolismo , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo , Secuencia de Aminoácidos , Proteínas Arqueales/química , Proteínas Arqueales/genética , Guanosina Difosfato/química , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Factor 2 de Elongación Peptídica/química , Factor 2 de Elongación Peptídica/genética , Unión Proteica , Conformación Proteica , Pyrococcus horikoshii/genética , Proteínas Ribosómicas/química , Proteínas Ribosómicas/genética , Ribosomas/química , Homología de Secuencia de Aminoácido
17.
J Med Virol ; 90(1): 13-18, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28922464

RESUMEN

In the year 2015, new Zika virus (ZIKV) broke out in Brazil and spread away in more than 80 countries. Scientists directed their efforts toward viral polymerase in attempt to find inhibitors that might interfere with its function. In this study, molecular dynamics simulation (MDS) was performed over 444 ns for a ZIKV polymerase model. Molecular docking (MD) was then performed every 10 ns during the MDS course to ensure the binding of small molecules to the polymerase over the entire time of the simulation. MD revealed the binding ability of four suggested guanosine inhibitors (GIs); (Guanosine substituted with OH and SH (phenyl) oxidanyl in the 2' carbon of the ribose ring). The GIs were compared to guanosine triphosphate (GTP) and five anti-hepatitis C virus drugs (either approved or under clinical trials). The mode of binding and the binding performance of GIs to ZIKV polymerase were found to be the same as GTP. Hence, these compounds were capable of competing GTP for the active site. Moreover, GIs bound to ZIKV active site more tightly compared to ribavirin, the wide-range antiviral drug.


Asunto(s)
Antivirales/metabolismo , Antivirales/farmacología , Nucleótidos/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/metabolismo , Virus Zika/efectos de los fármacos , Virus Zika/metabolismo , Antivirales/química , Sitios de Unión , Brasil , Guanosina/antagonistas & inhibidores , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/química , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Virus Zika/enzimología
18.
Biochem J ; 475(1): 99-115, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29138260

RESUMEN

Chloroplasts evolved from cyanobacterial endosymbiotic ancestors and their division is a complex process initiated by the assembly of cytoskeletal FtsZ (Filamentous temperature sensitive Z) proteins into a ring structure at the division site (Z-ring). The cyanobacterial Z-ring positioning system (MinCDE proteins) is also conserved in chloroplasts, except that MinC was lost and replaced by the eukaryotic ARC3 (accumulation and replication of chloroplasts). Both MinC and ARC3 act as negative regulators of FtsZ assembly, but ARC3 bears little sequence similarity with MinC. Here, light scattering assays, co-sedimentation, GTPase assay and transmission electron microscopy in conjunction with single-particle analysis have been used to elucidate the structure of ARC3 and its effect on its main target in chloroplast division, FtsZ2. Analysis of FtsZ2 in vitro assembly reactions in the presence and absence of GMPCPP showed that ARC3 promotes FtsZ2 debundling and disassembly of existing filaments in a concentration-dependent manner and requires GTP hydrolysis. Three-dimensional reconstruction of ARC3 revealed an almost circular molecule in which the FtsZ-binding N-terminus and the C-terminal PARC6 (paralog of ARC6)-binding MORN (Membrane Occupation and Recognition Nexus) domain are in close proximity and suggest a model for PARC6-enabled binding of ARC3 to FtsZ2. The latter is corroborated by in vivo data.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/genética , Cloroplastos/metabolismo , Regulación de la Expresión Génica de las Plantas , Guanosina Trifosfato/metabolismo , Arabidopsis/efectos de los fármacos , Arabidopsis/metabolismo , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , División Celular , Cloroplastos/efectos de los fármacos , Cloroplastos/genética , Cloroplastos/ultraestructura , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Células Eucariotas/metabolismo , Células Eucariotas/ultraestructura , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/farmacología , Cinética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Pichia/genética , Pichia/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
19.
Chem Res Toxicol ; 30(11): 1993-2001, 2017 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-28862449

RESUMEN

Members of the nucleoside analogue class of cancer therapeutics compete with canonical nucleotides to disrupt numerous cellular processes, including nucleotide homeostasis, DNA and RNA synthesis, and nucleotide metabolism. Nucleoside analogues are triphosphorylated and subsequently inserted into genomic DNA, contributing to the efficacy of therapeutic nucleosides in multiple ways. In some cases, the altered base acts as a mutagen, altering the DNA sequence to promote cellular death; in others, insertion of the altered nucleotide triggers DNA repair pathways, which produce lethal levels of cytotoxic intermediates such as single and double stranded DNA breaks. As a prerequisite to many of these biological outcomes, the modified nucleotide must be accommodated in the DNA polymerase active site during nucleotide insertion. Currently, the molecular contacts that mediate DNA polymerase insertion of modified nucleotides remain unknown for multiple therapeutic compounds, despite decades of clinical use. To determine how modified bases are inserted into duplex DNA, we used mammalian DNA polymerase ß (pol ß) to visualize the structural conformations of four therapeutically relevant modified nucleotides, 6-thio-2'-deoxyguanosine-5'-triphosphate (6-TdGTP), 5-fluoro-2'-deoxyuridine-5'-triphosphate (5-FdUTP), 5-formyl-deoxycytosine-5'-triphosphate (5-FodCTP), and 5-formyl-deoxyuridine-5'-triphosphate (5-FodUTP). Together, the structures reveal a pattern in which the modified nucleotides utilize Watson-Crick base pairing interactions similar to that of unmodified nucleotides. The nucleotide modifications were consistently positioned in the major groove of duplex DNA, accommodated by an open cavity in pol ß. These results provide novel information for the rational design of new therapeutic nucleoside analogues and a greater understanding of how modified nucleotides are tolerated by polymerases.


Asunto(s)
ADN Polimerasa beta/química , ADN/química , Nucleótidos de Desoxicitosina/química , Nucleótidos de Desoxiuracil/química , Guanosina Trifosfato/análogos & derivados , Emparejamiento Base , Sitios de Unión , Cristalografía por Rayos X , ADN/metabolismo , ADN Polimerasa beta/metabolismo , Nucleótidos de Desoxicitosina/metabolismo , Nucleótidos de Desoxiuracil/metabolismo , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Humanos , Modelos Moleculares , Conformación Molecular , Conformación de Ácido Nucleico
20.
J Phys Chem B ; 121(36): 8427-8436, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28820593

RESUMEN

Microtubule (MT) is made of αß-tubulin heterodimers that dynamically assemble into a hollow nanotube composed of straight protofilaments. MT dynamics is facilitated by hydrolysis of guanosine-5'-triphosphate (GTP) and can be inhibited by either anticancer agents like taxol or the nonhydrolyzable GTP analogues like GMPPCP. Using high-resolution synchrotron X-ray scattering, we have measured and analyzed the scattering curves from solutions of dynamic MT (in other words, in the presence of excess GTP and free of dynamic-inhibiting agents) and examined the effect of two MT stabilizers: taxol and GMPPCP. Previously, we have analyzed the structure of dynamic MT by docking the atomic model of tubulin dimer onto a 3-start left handed helical lattice, derived from the PDB ID 3J6F . 3J6F corresponds to a MT with 14 protofilaments. In this paper, we took into account the possibility of having MT structures containing between 12 and 15 protofilaments. MTs with 12 protofilaments were never observed. We determined the radii, the pitch, and the distribution of protofilament number that best fit the scattering data from dynamic MT or stabilized MT by taxol or GMPPCP. We found that the protofilament number distribution shifted when the MT was stabilized. Taxol increased the mass fraction of MT with 13 protofilaments and decreased the mass fraction of MT with 14 protofilaments. GMPPCP reduced the mass fraction of MT with 15 protofilaments and increased the mass fraction of MT with 14 protofilaments. The pitch, however, remained unchanged regardless of whether the MT was dynamic or stabilized. Higher tubulin concentrations increased the fraction of dynamic MT with 14 protofilaments.


Asunto(s)
Guanosina Trifosfato/análogos & derivados , Microtúbulos/química , Paclitaxel/química , Tubulina (Proteína)/química , Guanosina Trifosfato/química , Microscopía Electrónica de Transmisión , Simulación del Acoplamiento Molecular , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Dispersión de Radiación , Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...