Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Nucleic Acids Res ; 50(10): 5688-5712, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35641110

RESUMEN

Elongation by RNA polymerase is dynamically modulated by accessory factors. The transcription-repair coupling factor (TRCF) recognizes paused/stalled RNAPs and either rescues transcription or initiates transcription termination. Precisely how TRCFs choose to execute either outcome remains unclear. With Escherichia coli as a model, we used single-molecule assays to study dynamic modulation of elongation by Mfd, the bacterial TRCF. We found that nucleotide-bound Mfd converts the elongation complex (EC) into a catalytically poised state, presenting the EC with an opportunity to restart transcription. After long-lived residence in this catalytically poised state, ATP hydrolysis by Mfd remodels the EC through an irreversible process leading to loss of the RNA transcript. Further, biophysical studies revealed that the motor domain of Mfd binds and partially melts DNA containing a template strand overhang. The results explain pathway choice determining the fate of the EC and provide a molecular mechanism for transcription modulation by TRCF.


Asunto(s)
Proteínas Bacterianas , Reparación del ADN , Escherichia coli , Factores de Transcripción , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
2.
Biochem Biophys Res Commun ; 590: 7-13, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-34959192

RESUMEN

Contamination of drinking water with toxic inorganic arsenic is a major public health issue. The mechanisms of enzymes and transporters in arsenic elimination are therefore of interest. The human omega-class glutathione transferases have been previously shown to possess monomethylarsonate (V) reductase activity. To further understanding of this activity, molecular dynamics of human GSTO1-1 bound to glutathione with a monomethylarsonate isostere were simulated to reveal putative monomethylarsonate binding sites on the enzyme. The major binding site is in the active site, adjacent to the glutathione binding site. Based on this and previously reported biochemical data, a reaction mechanism for this enzyme is proposed. Further insights were gained from comparison of the human omega-class GSTs to homologs from a range of animals.


Asunto(s)
Glutatión Transferasa/metabolismo , Oxidorreductasas/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Glutatión Transferasa/química , Humanos , Modelos Moleculares
3.
Nature ; 525(7569): 394-8, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26322585

RESUMEN

In all domains of life, DNA synthesis occurs bidirectionally from replication origins. Despite variable rates of replication fork progression, fork convergence often occurs at specific sites. Escherichia coli sets a 'replication fork trap' that allows the first arriving fork to enter but not to leave the terminus region. The trap is set by oppositely oriented Tus-bound Ter sites that block forks on approach from only one direction. However, the efficiency of fork blockage by Tus-Ter does not exceed 50% in vivo despite its apparent ability to almost permanently arrest replication forks in vitro. Here we use data from single-molecule DNA replication assays and structural studies to show that both polarity and fork-arrest efficiency are determined by a competition between rates of Tus displacement and rearrangement of Tus-Ter interactions that leads to blockage of slower moving replisomes by two distinct mechanisms. To our knowledge this is the first example where intrinsic differences in rates of individual replisomes have different biological outcomes.


Asunto(s)
Replicación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Complejos Multienzimáticos/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Secuencia de Bases , Unión Competitiva , Cromosomas Bacterianos/genética , Cromosomas Bacterianos/metabolismo , Cristalografía por Rayos X , ADN Polimerasa Dirigida por ADN/química , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Cinética , Modelos Biológicos , Modelos Moleculares , Movimiento , Complejos Multienzimáticos/química , Conformación Proteica , Resonancia por Plasmón de Superficie , Factores de Tiempo
4.
J Comput Chem ; 41(6): 587-603, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-31840840

RESUMEN

Bioluminescence in marine systems is dominated by the use of coelenterazine for light production. The bioluminescent reaction of coelenterazine is an enzyme catalyzed oxidative decarboxylation: coelenterazine reacts with molecular oxygen to form carbon dioxide, coelenteramide, and light. One such class is the Ca2+ -regulated photoproteins. These proteins bind coelenterazine and oxygen, and trap 2-hydroperoxycoelenterazine, an intermediate along the reaction pathway. The reaction is halted until Ca2+ binding triggers the completion of the reaction. There are currently no reported experimental, atomistic descriptions of this ternary Michaelis complex. This study utilized computational techniques to develop an atomistic model of the Michaelis complex. Extensive molecular dynamics simulations were carried out to study the interactions between four tautomeric/protonation states of coelenterazine and wide-type and mutant obelin. Only minor differences in binding modes were observed across all systems. Interestingly, no basic residues were identified in the vicinity of the N7-nitrogen of coelenterazine. This observation was surprising considering that deprotonation at this position is a key mechanistic step in the proposed bioluminescent reaction. This work suggests that coelenterazine binds either as the O10H tautomer, or in the deprotonated form. Implicit ligand sampling simulations were used to identify potential O2 binding and migration pathways within obelin. A key oxygen binding site was identified close to the coelenterazine imidazopyrazinone core. The O2 binding free energy was observed to be dependent on the protonation state of coelenterazine. Taken together, the description of the obelin-coelenterazine-O2 complexes established in this study provides the basis for future computational studies of the bioluminescent mechanism. © 2019 Wiley Periodicals, Inc.


Asunto(s)
Imidazoles/química , Proteínas Luminiscentes/síntesis química , Simulación de Dinámica Molecular , Oxígeno/química , Pirazinas/química , Sitios de Unión , Proteínas Luminiscentes/química , Estructura Molecular
5.
J Struct Biol ; 204(3): 396-405, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30366028

RESUMEN

Bacterial sliding clamps bind to DNA and act as protein-protein interaction hubs for several proteins involved in DNA replication and repair. The partner proteins all bind to a common pocket on sliding clamps via conserved linear peptide sequence motifs, which suggest the pocket as an attractive target for development of new antibiotics. Herein we report the X-ray crystal structures and biochemical characterization of ß sliding clamps from the Gram-negative pathogens Pseudomonas aeruginosa, Acinetobacter baumannii and Enterobacter cloacae. The structures reveal close similarity between the pathogen and Escherichia coli clamps and similar patterns of binding to linear clamp-binding motif peptides. The results suggest that linear motif-sliding clamp interactions are well conserved and an antibiotic targeting the sliding clamp should have broad-spectrum activity against Gram-negative pathogens.


Asunto(s)
Acinetobacter baumannii/genética , ADN Bacteriano/química , Enterobacter cloacae/genética , Pseudomonas aeruginosa/genética , Algoritmos , Secuencias de Aminoácidos/genética , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacología , Cristalografía por Rayos X , Replicación del ADN/efectos de los fármacos , Replicación del ADN/genética , ADN Bacteriano/metabolismo , Escherichia coli/genética , Modelos Moleculares , Conformación de Ácido Nucleico , Péptidos/química , Péptidos/genética , Péptidos/metabolismo , Unión Proteica , Conformación Proteica
6.
J Struct Biol ; 203(3): 205-218, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29885491

RESUMEN

Apolipoprotein-D is a 25 kDa glycosylated member of the lipocalin family that folds into an eight-stranded ß-barrel with a single adjacent α-helix. Apolipoprotein-D specifically binds a range of small hydrophobic ligands such as progesterone and arachidonic acid and has an antioxidant function that is in part due to the reduction of peroxidised lipids by methionine-93. Therefore, apolipoprotein-D plays multiple roles throughout the body and is protective in Alzheimer's disease, where apolipoprotein-D overexpression reduces the amyloid-ß burden in Alzheimer's disease mouse models. Oligomerisation is a common feature of lipocalins that can influence ligand binding. The native structure of apolipoprotein-D, however, has not been conclusively defined. Apolipoprotein-D is generally described as a monomeric protein, although it dimerises when reducing peroxidised lipids. Here, we investigated the native structure of apolipoprotein-D derived from plasma, breast cyst fluid (BCF) and cerebrospinal fluid. In plasma and cerebrospinal fluid, apolipoprotein-D was present in high-molecular weight complexes, potentially in association with lipoproteins. In contrast, apolipoprotein-D in BCF formed distinct oligomeric species. We assessed apolipoprotein-D oligomerisation using native apolipoprotein-D purified from BCF and a suite of complementary methods, including multi-angle laser light scattering, analytical ultracentrifugation and small-angle X-ray scattering. Our analyses showed that apolipoprotein-D predominantly forms a ∼95 to ∼100 kDa tetramer. Small-angle X-ray scattering analysis confirmed these findings and provided a structural model for apolipoprotein-D tetramer. These data indicate apolipoprotein-D rarely exists as a free monomer under physiological conditions and provide insights into novel native structures of apolipoprotein-D and into oligomerisation behaviour in the lipocalin family.


Asunto(s)
Enfermedad de Alzheimer/genética , Apolipoproteínas D/química , Conformación Proteica , Multimerización de Proteína , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Animales , Apolipoproteínas D/líquido cefalorraquídeo , Apolipoproteínas D/genética , Quiste Mamario/química , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Humanos , Ligandos , Lipocalinas/química , Ratones , Unión Proteica , Dispersión del Ángulo Pequeño
7.
J Biol Chem ; 292(20): 8401-8411, 2017 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-28351836

RESUMEN

Excitation-contraction (EC) coupling in skeletal muscle requires a physical interaction between the voltage-gated calcium channel dihydropyridine receptor (DHPR) and the ryanodine receptor Ca2+ release channel. Although the exact molecular mechanism that initiates skeletal EC coupling is unresolved, it is clear that both the α1 and ß subunits of DHPR are essential for this process. Here, we employed a series of techniques, including size-exclusion chromatography-multi-angle light scattering, differential scanning fluorimetry, and isothermal calorimetry, to characterize various biophysical properties of the skeletal DHPR ß subunit ß1a Removal of the intrinsically disordered N and C termini and the hook region of ß1a prevented oligomerization, allowing for its structural determination by X-ray crystallography. The structure had a topology similar to that of previously determined ß isoforms, which consist of SH3 and guanylate kinase domains. However, transition melting temperatures derived from the differential scanning fluorimetry experiments indicated a significant difference in stability of ∼2-3 °C between the ß1a and ß2a constructs, and the addition of the DHPR α1s I-II loop (α-interaction domain) peptide stabilized both ß isoforms by ∼6-8 °C. Similar to other ß isoforms, ß1a bound with nanomolar affinity to the α-interaction domain, but binding affinities were influenced by amino acid substitutions in the adjacent SH3 domain. These results suggest that intramolecular interactions between the SH3 and guanylate kinase domains play a role in the stability of ß1a while also providing a conduit for allosteric signaling events.


Asunto(s)
Canales de Calcio Tipo L/química , Guanilato-Quinasas/química , Regulación Alostérica , Animales , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Cristalografía por Rayos X , Guanilato-Quinasas/genética , Guanilato-Quinasas/metabolismo , Ratones , Estructura Secundaria de Proteína , Transducción de Señal , Dominios Homologos src
8.
Chemistry ; 24(44): 11325-11331, 2018 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-29917264

RESUMEN

The human sliding clamp (PCNA) controls access to DNA for many proteins involved in DNA replication and repair. Proteins are recruited to the PCNA surface by means of a short, conserved peptide motif known as the PCNA-interacting protein box (PIP-box). Inhibitors of these essential protein-protein interactions may be useful as cancer therapeutics by disrupting DNA replication and repair in these highly proliferative cells. PIP-box peptide mimetics have been identified as a potentially rapid route to potent PCNA inhibitors. Here we describe the rational design and synthesis of the first PCNA peptidomimetic ligands, based on the high affinity PIP-box sequence from the natural PCNA inhibitor p21. These mimetics incorporate covalent i,i+4 side-chain/side-chain lactam linkages of different lengths, designed to constrain the peptides into the 310 -helical structure required for PCNA binding. NMR studies confirmed that while the unmodified p21 peptide had little defined structure in solution, mimetic ACR2 pre-organized into 310 -helical structure prior to interaction with PCNA. ACR2 displayed higher affinity binding than most known PIP-box peptides, and retains the native PCNA binding mode, as observed in the co-crystal structure of ACR2 bound to PCNA. This study offers a promising new strategy for PCNA inhibitor design for use as anti-cancer therapeutics.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/química , Péptidos/química , Antígeno Nuclear de Célula en Proliferación/química , Secuencias de Aminoácidos , Sitios de Unión , Fenómenos Bioquímicos , Cristalografía por Rayos X , Humanos , Lactamas/química , Ligandos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Peptidomiméticos/química , Conformación Proteica en Hélice alfa
9.
Biochem J ; 474(14): 2475-2487, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28592682

RESUMEN

Over time, the long-lived proteins that are present throughout the human body deteriorate. Typically, they become racemized, truncated, and covalently cross-linked. One reaction responsible for age-related protein cross-linking in the lens was elucidated recently and shown to involve spontaneous formation of dehydroalanine (DHA) intermediates from phosphoserine. Cys residues are another potential source of DHA, and evidence for this was found in many lens crystallins. In the human lens, some sites were more prone to forming non-disulfide covalent cross-links than others. Foremost among them was Cys5 in ßA4 crystallin. The reason for this enhanced reactivity was investigated using peptides. Oxidation of Cys to cystine was a prerequisite for DHA formation, and DHA production was accelerated markedly by the presence of a Lys, one residue separated from Cys5. Modeling and direct investigation of the N-terminal sequence of ßA4 crystallin, as well as a variety of homologous peptides, showed that the epsilon amino group of Lys can promote DHA production by nucleophilic attack on the alpha proton of cystine. Once a DHA residue was generated, it could form intermolecular cross-links with Lys and Cys. In the lens, the most abundant cross-link involved Cys5 of ßA4 crystallin attached via a thioether bond to glutathione. These findings illustrate the potential of Cys and disulfide bonds to act as precursors for irreversible covalent cross-links and the role of nearby amino acids in creating 'hotpsots' for the spontaneous processes responsible for protein degradation in aged tissues.


Asunto(s)
Cisteína/química , Proteínas del Ojo/química , Cristalino/química , Factores de Edad , Alanina/análogos & derivados , Alanina/química , Bases de Datos de Proteínas , Disulfuros/química , Humanos , Modelos Moleculares , Oligopéptidos/química , Proteolisis , Espectrometría de Masas en Tándem , Cadena A de beta-Cristalina/química
10.
Nucleic Acids Res ; 42(4): 2750-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24288378

RESUMEN

Single-stranded DNA (ssDNA) binding protein (SSB) is an essential protein to protect ssDNA and recruit specific ssDNA-processing proteins. Escherichia coli SSB forms a tetramer at neutral pH, comprising a structurally well-defined ssDNA binding domain (OB-domain) and a disordered C-terminal domain (C-domain) of ∼ 64 amino acid residues. The C-terminal eight-residue segment of SSB (C-peptide) has been shown to interact with the OB-domain, but crystal structures failed to reveal any electron density of the C-peptide. Here we show that SSB forms a monomer at pH 3.4, which is suitable for studies by high-resolution nuclear magnetic resonance (NMR) spectroscopy. The OB-domain retains its 3D structure in the monomer, and the C-peptide is shown by nuclear Overhauser effects and lanthanide-induced pseudocontact shifts to bind to the OB-domain at a site that harbors ssDNA in the crystal structure of the SSB-ssDNA complex. (15)N relaxation data demonstrate high flexibility of the polypeptide segment linking the C-peptide to the OB-domain and somewhat increased flexibility of the C-peptide compared with the OB-domain, suggesting that the C-peptide either retains high mobility in the bound state or is in a fast equilibrium with an unbound state.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Escherichia coli/química , Sitios de Unión , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Péptidos/metabolismo , Unión Proteica
11.
Nucleic Acids Res ; 41(10): 5354-67, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23580545

RESUMEN

A complex of the three (αεθ) core subunits and the ß2 sliding clamp is responsible for DNA synthesis by Pol III, the Escherichia coli chromosomal DNA replicase. The 1.7 Å crystal structure of a complex between the PHP domain of α (polymerase) and the C-terminal segment of ε (proofreading exonuclease) subunits shows that ε is attached to α at a site far from the polymerase active site. Both α and ε contain clamp-binding motifs (CBMs) that interact simultaneously with ß2 in the polymerization mode of DNA replication by Pol III. Strengthening of both CBMs enables isolation of stable αεθ:ß2 complexes. Nuclear magnetic resonance experiments with reconstituted αεθ:ß2 demonstrate retention of high mobility of a segment of 22 residues in the linker that connects the exonuclease domain of ε with its α-binding segment. In spite of this, small-angle X-ray scattering data show that the isolated complex with strengthened CBMs has a compact, but still flexible, structure. Photo-crosslinking with p-benzoyl-L-phenylalanine incorporated at different sites in the α-PHP domain confirm the conformational variability of the tether. Structural models of the αεθ:ß2 replicase complex with primer-template DNA combine all available structural data.


Asunto(s)
ADN Polimerasa III/química , Proteínas de Escherichia coli/química , Exodesoxirribonucleasas/química , Secuencia de Aminoácidos , ADN Polimerasa III/metabolismo , Proteínas de Escherichia coli/metabolismo , Exodesoxirribonucleasas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína
13.
Biochem J ; 442(3): 713-21, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22150111

RESUMEN

ApoD (apolipoprotein D) is up-regulated in AD (Alzheimer's disease) and upon oxidative stress. ApoD inhibits brain lipid peroxidation in vivo, but the mechanism is unknown. Specific methionine residues may inhibit lipid peroxidation by reducing radical-propagating L-OOHs (lipid hydroperoxides) to non-reactive hydroxides via a reaction that generates MetSO (methionine sulfoxide). Since apoD has three conserved methionine residues (Met(49), Met(93) and Met(157)), we generated recombinant proteins with either one or all methionine residues replaced by alanine and assessed their capacity to reduce HpETEs (hydroperoxyeicosatetraenoic acids) to their HETE (hydroxyeicosatetraenoic acid) derivatives. ApoD, apoD(M49-A) and apoD(M157-A) all catalysed the reduction of HpETEs to their corresponding HETEs. Amino acid analysis of HpETE-treated apoD revealed a loss of one third of the methionine residues accompanied by the formation of MetSO. Additional studies using apoD(M93-A) indicated that Met(93) was required for HpETE reduction. We also assessed the impact that apoD MetSO formation has on protein aggregation by Western blotting of HpETE-treated apoD and human brain samples. ApoD methionine oxidation was associated with formation of apoD aggregates that were also detected in the hippocampus of AD patients. In conclusion, conversion of HpETE into HETE is mediated by apoD Met(93), a process that may contribute to apoD antioxidant function.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Antioxidantes/metabolismo , Apolipoproteínas D/metabolismo , Ácidos Hidroxieicosatetraenoicos/química , Animales , Apolipoproteínas D/química , Células CHO , Cricetinae , Células HEK293 , Hipocampo/metabolismo , Humanos , Ácidos Hidroxieicosatetraenoicos/metabolismo , Metionina/análogos & derivados , Metionina/metabolismo , Oxidación-Reducción
14.
Proc Natl Acad Sci U S A ; 107(2): 657-62, 2010 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-20080731

RESUMEN

Enzymatic catalysis of oxygenation reactions in the absence of metal or organic cofactors is a considerable biochemical challenge. The CO-forming 1-H-3-hydroxy-4-oxoquinaldine 2,4-dioxygenase (HOD) from Arthrobacter nitroguajacolicus Rü61a and 1-H-3-hydroxy-4-oxoquinoline 2,4-dioxygenase (QDO) from Pseudomonas putida 33/1 are homologous cofactor-independent dioxygenases involved in the breakdown of N-heteroaromatic compounds. To date, they are the only dioxygenases suggested to belong to the alpha/beta-hydrolase fold superfamily. Members of this family typically catalyze hydrolytic processes rather than oxygenation reactions. We present here the crystal structures of both HOD and QDO in their native state as well as the structure of HOD in complex with its natural 1-H-3-hydroxy-4-oxoquinaldine substrate, its N-acetylanthranilate reaction product, and chloride as dioxygen mimic. HOD and QDO are structurally very similar. They possess a classical alpha/beta-hydrolase fold core domain additionally equipped with a cap domain. Organic substrates bind in a preorganized active site with an orientation ideally suited for selective deprotonation of their hydroxyl group by a His/Asp charge-relay system affording the generation of electron-donating species. The "oxyanion hole" of the alpha/beta-hydrolase fold, typically employed to stabilize the tetrahedral intermediate in ester hydrolysis reactions, is utilized here to host and control oxygen chemistry, which is proposed to involve a peroxide anion intermediate. Product release by proton back transfer from the catalytic histidine is driven by minimization of intramolecular charge repulsion. Structural and kinetic data suggest a nonnucleophilic general-base mechanism. Our analysis provides a framework to explain cofactor-independent dioxygenation within a protein architecture generally employed to catalyze hydrolytic reactions.


Asunto(s)
Dioxigenasas/química , Dioxigenasas/metabolismo , Pseudomonas putida/enzimología , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Cinética , Modelos Moleculares , Conformación Proteica , Pliegue de Proteína , Especificidad por Sustrato , Propiedades de Superficie
15.
Biomolecules ; 13(8)2023 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-37627305

RESUMEN

With the development of accurate protein structure prediction algorithms, artificial intelligence (AI) has emerged as a powerful tool in the field of structural biology. AI-based algorithms have been used to analyze large amounts of protein sequence data including the human proteome, complementing experimental structure data found in resources such as the Protein Data Bank. The EBI AlphaFold Protein Structure Database (for example) contains over 230 million structures. In this study, these data have been analyzed to find all human proteins containing (or predicted to contain) the cytosolic glutathione transferase (cGST) fold. A total of 39 proteins were found, including the alpha-, mu-, pi-, sigma-, zeta- and omega-class GSTs, intracellular chloride channels, metaxins, multisynthetase complex components, elongation factor 1 complex components and others. Three broad themes emerge: cGST domains as enzymes, as chloride ion channels and as protein-protein interaction mediators. As the majority of cGSTs are dimers, the AI-based structure prediction algorithm AlphaFold-multimer was used to predict structures of all pairwise combinations of these cGST domains. Potential homo- and heterodimers are described. Experimental biochemical and structure data is used to highlight the strengths and limitations of AI-predicted structures.


Asunto(s)
Genoma Humano , Glutatión Transferasa , Humanos , Glutatión Transferasa/genética , Inteligencia Artificial , Algoritmos , Secuencia de Aminoácidos
16.
J Biol Chem ; 286(6): 4271-9, 2011 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-21106529

RESUMEN

The polymorphic deletion of Glu-155 from human glutathione transferase omega1 (GSTO1-1) occurs in most populations. Although the recombinant ΔGlu-155 enzyme expressed in Escherichia coli is active, the deletion causes a deficiency of the active enzyme in vivo. The crystal structure and the folding/unfolding kinetics of the ΔGlu-155 variant were determined in order to investigate the cause of the rapid loss of the enzyme in human cells. The crystal structure revealed altered packing around the Glu-155 deletion, an increase in the predicted solvent-accessible area and a corresponding reduction in the buried surface area. This increase in solvent accessibility was consistent with an elevated Stern-Volmer constant. The unfolding of both the wild type and ΔGlu-155 enzyme in urea is best described by a three-state model, and there is evidence for the more pronounced population of an intermediate state by the ΔGlu-155 enzymes. Studies using intrinsic fluorescence revealed a free energy change around 14.4 kcal/mol for the wild type compared with around 8.6 kcal/mol for the ΔGlu-155 variant, which indicates a decrease in stability associated with the Glu-155 deletion. Urea induced unfolding of the wild type GSTO1-1 was reversible through an initial fast phase followed by a second slow phase. In contrast, the ΔGlu-155 variant lacks the slow phase, indicating a refolding defect. It is possible that in some conditions in vivo, the increased solvent-accessible area and the low stability of the ΔGlu-155 variant may promote its unfolding, whereas the refolding defect limits its refolding, resulting in GSTO1-1 deficiency.


Asunto(s)
Glutatión Transferasa/química , Glutatión Transferasa/deficiencia , Modelos Moleculares , Pliegue de Proteína , Cristalografía por Rayos X , Estabilidad de Enzimas/genética , Escherichia coli , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Humanos , Mutación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Urea/química
17.
Int J Mol Sci ; 13(10): 12857-79, 2012 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-23202926

RESUMEN

Fragment-based drug discovery (FBDD) concerns the screening of low-molecular weight compounds against macromolecular targets of clinical relevance. These compounds act as starting points for the development of drugs. FBDD has evolved and grown in popularity over the past 15 years. In this paper, the rationale and technology behind the use of X-ray crystallography in fragment based screening (FBS) will be described, including fragment library design and use of synchrotron radiation and robotics for high-throughput X-ray data collection. Some recent uses of crystallography in FBS will be described in detail, including interrogation of the drug targets ß-secretase, phenylethanolamine N-methyltransferase, phosphodiesterase 4A and Hsp90. These examples provide illustrations of projects where crystallography is straightforward or difficult, and where other screening methods can help overcome the limitations of crystallography necessitated by diffraction quality.


Asunto(s)
Proteínas/química , Secretasas de la Proteína Precursora del Amiloide/química , Sitios de Unión , Cristalografía por Rayos X , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/química , Diseño de Fármacos , Proteínas HSP90 de Choque Térmico/química , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Feniletanolamina N-Metiltransferasa/química , Estructura Terciaria de Proteína , Proteínas/metabolismo
18.
Protein Sci ; 31(5): e4311, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35481653

RESUMEN

Excitation-contraction coupling (ECC) is the physiological process in which an electrical signal originating from the central nervous system is converted into muscle contraction. In skeletal muscle tissue, the key step in the molecular mechanism of ECC initiated by the muscle action potential is the cooperation between two Ca2+ channels, dihydropyridine receptor (DHPR; voltage-dependent L-type calcium channel) and ryanodine receptor 1 (RyR1). These two channels were originally postulated to communicate with each other via direct mechanical interactions; however, the molecular details of this cooperation have remained ambiguous. Recently, it has been proposed that one or more supporting proteins are in fact required for communication of DHPR with RyR1 during the ECC process. One such protein that is increasingly believed to play a role in this interaction is the SH3 and cysteine-rich domain-containing protein 3 (STAC3), which has been proposed to bind a cytosolic portion of the DHPR α1S subunit known as the II-III loop. In this work, we present direct evidence for an interaction between a small peptide sequence of the II-III loop and several residues within the SH3 domains of STAC3 as well as the neuronal isoform STAC2. Differences in this interaction between STAC3 and STAC2 suggest that STAC3 possesses distinct biophysical features that are potentially important for its physiological interactions with the II-III loop. Therefore, this work demonstrates an isoform-specific interaction between STAC3 and the II-III loop of DHPR and provides novel insights into a putative molecular mechanism behind this association in the skeletal muscle ECC process.


Asunto(s)
Canales de Calcio Tipo L , Canal Liberador de Calcio Receptor de Rianodina , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Acoplamiento Excitación-Contracción/fisiología , Músculo Esquelético/fisiología , Isoformas de Proteínas/metabolismo
19.
J Biol Chem ; 285(13): 9642-9648, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20110353

RESUMEN

Gamma-glutamylamine cyclotransferase (GGACT) is an enzyme that converts gamma-glutamylamines to free amines and 5-oxoproline. GGACT shows high activity toward gamma-glutamyl-epsilon-lysine, derived from the breakdown of fibrin and other proteins cross-linked by transglutaminases. The enzyme adopts the newly identified cyclotransferase fold, observed in gamma-glutamylcyclotransferase (GGCT), an enzyme with activity toward gamma-glutamyl-alpha-amino acids (Oakley, A. J., Yamada, T., Liu, D., Coggan, M., Clark, A. G., and Board, P. G. (2008) J. Biol. Chem. 283, 22031-22042). Despite the absence of significant sequence identity, several residues are conserved in the active sites of GGCT and GGACT, including a putative catalytic acid/base residue (GGACT Glu(82)). The structure of GGACT in complex with the reaction product 5-oxoproline provides evidence for a common catalytic mechanism in both enzymes. The proposed mechanism, combined with the three-dimensional structures, also explains the different substrate specificities of these enzymes. Despite significant sequence divergence, there are at least three subfamilies in prokaryotes and eukaryotes that have conserved the GGCT fold and GGCT enzymatic activity.


Asunto(s)
Dipéptidos/química , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , gamma-Glutamilciclotransferasa/química , gamma-Glutamilciclotransferasa/genética , Secuencia de Aminoácidos , Catálisis , Dominio Catalítico , Línea Celular Tumoral , Clonación Molecular , Reactivos de Enlaces Cruzados/química , Cristalografía por Rayos X/métodos , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Conformación Proteica , Ácido Pirrolidona Carboxílico/química , Homología de Secuencia de Aminoácido
20.
Transcription ; 12(4): 92-102, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34403307

RESUMEN

The low G + C Gram-positive bacteria represent some of the most medically and industrially important microorganisms. They are relied on for the production of food and dietary supplements, enzymes and antibiotics, as well as being responsible for the majority of nosocomial infections and serving as a reservoir for antibiotic resistance. Control of gene expression in this group is more highly studied than in any bacteria other than the Gram-negative model  Escherichia coli, yet until recently no structural information on RNA polymerase (RNAP) from this group was available. This review will summarize recent reports on the high-resolution structure of RNAP from the model low G + C representative  Bacillus subtilis, including the role of auxiliary subunits δ and ε, and outline approaches for the development of antimicrobials to target RNAP from this group.


Asunto(s)
Proteínas Bacterianas , ARN Polimerasas Dirigidas por ADN , Bacterias Grampositivas , Bacillus subtilis/química , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas Bacterianas/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA