Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 299(9): 105189, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37625592

RESUMEN

Xanthine oxidoreductase is a metalloenzyme that catalyzes the final steps in purine metabolism by converting hypoxanthine to xanthine and then uric acid. Allopurinol, an analog of hypoxanthine, is widely used as an antigout drug, as xanthine oxidoreductase-mediated metabolism of allopurinol to oxypurinol leads to oxypurinol rotation in the enzyme active site and reduction of the molybdenum Mo(VI) active center to Mo(IV), inhibiting subsequent urate production. However, when oxypurinol is administered directly to a mouse model of hyperuricemia, it yields a weaker urate-lowering effect than allopurinol. To better understand its mechanism of inhibition and inform patient dosing strategies, we performed kinetic and structural analyses of the inhibitory activity of oxypurinol. Our results demonstrated that oxypurinol was less effective than allopurinol both in vivo and in vitro. We show that upon reoxidation to Mo(VI), oxypurinol binding is greatly weakened, and reduction by xanthine, hypoxanthine, or allopurinol is required for reformation of the inhibitor-enzyme complex. In addition, we show oxypurinol only weakly inhibits the conversion of hypoxanthine to xanthine and is therefore unlikely to affect the feedback inhibition of de novo purine synthesis. Furthermore, we observed weak allosteric inhibition of purine nucleoside phosphorylase by oxypurinol which has potentially adverse effects for patients. Considering these results, we propose the single-dose method currently used to treat hyperuricemia can result in unnecessarily high levels of allopurinol. While the short half-life of allopurinol in blood suggests that oxypurinol is responsible for enzyme inhibition, we anticipate multiple, smaller doses of allopurinol would reduce the total allopurinol patient load.

2.
J Biomol Struct Dyn ; 40(24): 13823-13832, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34705594

RESUMEN

Protein tyrosine phosphatases constitute a family of cytosolic and receptor-like signal transducing enzymes that catalyze the hydrolysis of phospho-tyrosine residues of phosphorylated proteins. PTP1B, encoded by PTPN1, is a key negative regulator of insulin and leptin receptor signaling, linking it to two widespread diseases: type 2 diabetes mellitus and obesity. Here, we present crystal structures of the PTP1B apo-enzyme and a complex with a newly identified allosteric inhibitor, 2-(2,5-dimethyl-pyrrol-1-yl)-5-hydroxy-benzoic acid, designated as P00058. The inhibitor binding site is located about 18 Å away from the active center. However, the inhibitor causes significant re-arrangements in the active center of enzyme: residues 45-50 of catalytic Tyr-loop are shifted at their Cα-atom positions by 2.6 to 5.8 Å. We have identified an event of allosteric signal transfer from the inhibitor to the catalytic area using molecular dynamic simulation. Analyzing change of complex structure along the fluctuation trajectory we have found the large Cα-atom shifts in external strand, residues 25-40, which occur at the same time with the shifts in adjacent catalytic p-Tyr-loop. Coming of the signal to this loop arises due to dynamic fluctuation of protein structure at about 4.0 nanoseconds after the inhibitor takes up its space. Communicated by Ramaswamy H. Sarma.


Asunto(s)
Diabetes Mellitus Tipo 2 , Humanos , Sitios de Unión , Transducción de Señal , Simulación de Dinámica Molecular , Obesidad , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química
3.
Chembiochem ; 23(1): e202100414, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34643018

RESUMEN

l-2-Haloacid dehalogenases, industrially and environmentally important enzymes that catalyse cleavage of the carbon-halogen bond in S-2-halocarboxylic acids, were known to hydrolyse chlorinated, brominated and iodinated substrates but no activity towards fluorinated compounds had been reported. A screen for novel dehalogenase activities revealed four l-2-haloacid dehalogenases capable of defluorination. We now report crystal structures for two of these enzymes, Bpro0530 and Rha0230, as well as for the related proteins PA0810 and RSc1362, which hydrolyse chloroacetate but not fluoroacetate, all at ∼2.2 Šresolution. Overall structure and active sites of these enzymes are highly similar. In molecular dynamics (MD) calculations, only the defluorinating enzymes sample more compact conformations, which in turn allow more effective interactions with the small fluorine atom. Structural constraints, based on X-ray structures and MD calculations, correctly predict the defluorination activity of the homologous enzyme ST2570.


Asunto(s)
Hidrolasas/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Halogenación , Hidrolasas/química , Hidrolasas/aislamiento & purificación , Simulación de Dinámica Molecular , Conformación Proteica , Alineación de Secuencia
4.
ACS Infect Dis ; 6(12): 3224-3236, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33237740

RESUMEN

Evolving antimicrobial resistance has motivated the search for novel targets and alternative therapies. Caseinolytic protease (ClpP) has emerged as an enticing new target since its function is conserved and essential for bacterial fitness, and because its inhibition or dysregulation leads to bacterial cell death. ClpP protease function controls global protein homeostasis and is, therefore, crucial for the maintenance of the bacterial proteome during growth and infection. Previously, acyldepsipeptides (ADEPs) were discovered to dysregulate ClpP, leading to bactericidal activity against both actively growing and dormant Gram-positive pathogens. Unfortunately, these compounds had very low efficacy against Gram-negative bacteria. Hence, we sought to develop non-ADEP ClpP-targeting compounds with activity against Gram-negative species and called these activators of self-compartmentalizing proteases (ACPs). These ACPs bind and dysregulate ClpP in a manner similar to ADEPs, effectively digesting bacteria from the inside out. Here, we performed further ACP derivatization and testing to improve the efficacy and breadth of coverage of selected ACPs against Gram-negative bacteria. We observed that a diverse collection of Neisseria meningitidis and Neisseria gonorrhoeae clinical isolates were exquisitely sensitive to these ACP analogues. Furthermore, based on the ACP-ClpP cocrystal structure solved here, we demonstrate that ACPs could be designed to be species specific. This validates the feasibility of drug-based targeting of ClpP in Gram-negative bacteria.


Asunto(s)
Antibacterianos , Depsipéptidos , Péptido Hidrolasas , Antibacterianos/farmacología , Bacterias , Depsipéptidos/farmacología , Bacterias Gramnegativas
5.
Commun Biol ; 2: 410, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31754640

RESUMEN

Bacterial ClpP is a highly conserved, cylindrical, self-compartmentalizing serine protease required for maintaining cellular proteostasis. Small molecule acyldepsipeptides (ADEPs) and activators of self-compartmentalized proteases 1 (ACP1s) cause dysregulation and activation of ClpP, leading to bacterial cell death, highlighting their potential use as novel antibiotics. Structural changes in Neisseria meningitidis and Escherichia coli ClpP upon binding to novel ACP1 and ADEP analogs were probed by X-ray crystallography, methyl-TROSY NMR, and small angle X-ray scattering. ACP1 and ADEP induce distinct conformational changes in the ClpP structure. However, reorganization of electrostatic interaction networks at the ClpP entrance pores is necessary and sufficient for activation. Further activation is achieved by formation of ordered N-terminal axial loops and reduction in the structural heterogeneity of the ClpP cylinder. Activating mutations recapitulate the structural effects of small molecule activator binding. Our data, together with previous findings, provide a structural basis for a unified mechanism of compound-based ClpP activation.


Asunto(s)
Endopeptidasa Clp/química , Modelos Moleculares , Electricidad Estática , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Endopeptidasa Clp/metabolismo , Activación Enzimática , Espectroscopía de Resonancia Magnética , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Proteínas Tirosina Fosfatasas/química
6.
Science ; 365(6458): 1167-1170, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31515393

RESUMEN

A comprehensive understanding of protein function demands correlating structure and dynamic changes. Using time-resolved serial synchrotron crystallography, we visualized half-of-the-sites reactivity and correlated molecular-breathing motions in the enzyme fluoroacetate dehalogenase. Eighteen time points from 30 milliseconds to 30 seconds cover four turnover cycles of the irreversible reaction. They reveal sequential substrate binding, covalent-intermediate formation, setup of a hydrolytic water molecule, and product release. Small structural changes of the protein mold and variations in the number and placement of water molecules accompany the various chemical steps of catalysis. Triggered by enzyme-ligand interactions, these repetitive changes in the protein framework's dynamics and entropy constitute crucial components of the catalytic machinery.


Asunto(s)
Proteínas Bacterianas/química , Dominio Catalítico , Hidrolasas/química , Rhodopseudomonas/enzimología , Catálisis , Entropía , Cinética , Ligandos , Modelos Moleculares , Conformación Proteica , Multimerización de Proteína
7.
Chemistry ; 25(49): 11416-11421, 2019 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-31407832

RESUMEN

Trypanothione reductase (TR) plays a key role in the unique redox metabolism of trypanosomatids, the causative agents of human African trypanosomiasis (HAT), Chagas' disease, and leishmaniases. Introduction of a new, lean propargylic vector to a known class of TR inhibitors resulted in the strongest reported competitive inhibitor of Trypanosoma (T.) brucei TR, with an inhibition constant Ki of 73 nm, which is fully selective against human glutathione reductase (hGR). The best ligands exhibited in vitro IC50 values (half-maximal inhibitory concentration) against the HAT pathogen, T. brucei rhodesiense, in the mid-nanomolar range, reaching down to 50 nm. X-Ray co-crystal structures confirmed the binding mode of the ligands and revealed the presence of a HEPES buffer molecule in the large active site. Extension of the propargylic vector, guided by structure-based design, to replace the HEPES buffer molecule should give inhibitors with low nanomolar Ki and IC50 values for in vivo studies.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/química , NADH NADPH Oxidorreductasas/antagonistas & inhibidores , Proteínas Protozoarias/antagonistas & inhibidores , Trypanosoma brucei brucei/enzimología , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Concentración 50 Inhibidora , Ligandos , Simulación de Dinámica Molecular , NADH NADPH Oxidorreductasas/metabolismo , Proteínas Protozoarias/metabolismo , Relación Estructura-Actividad
8.
J Am Chem Soc ; 141(29): 11540-11556, 2019 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-31188575

RESUMEN

Many enzymes operate through half-of-the sites reactivity wherein a single protomer is catalytically engaged at one time. In the case of the homodimeric enzyme, fluoroacetate dehalogenase, substrate binding triggers closing of a regulatory cap domain in the empty protomer, preventing substrate access to the remaining active site. However, the empty protomer serves a critical role by acquiring more disorder upon substrate binding, thereby entropically favoring the forward reaction. Empty protomer dynamics are also allosterically coupled to the bound protomer, driving conformational exchange at the active site and progress along the reaction coordinate. Here, we show that at high concentrations, a second substrate binds along the substrate-access channel of the occupied protomer, thereby dampening interprotomer dynamics and inhibiting catalysis. While a mutation (K152I) abrogates second site binding and removes inhibitory effects, it also precipitously lowers the maximum catalytic rate, implying a role for the allosteric pocket at low substrate concentrations, where only a single substrate engages the enzyme at one time. We show that this outer pocket first desolvates the substrate, whereupon it is deposited in the active site. Substrate binding to the active site then triggers the empty outer pocket to serve as an interprotomer allosteric conduit, enabling enhanced dynamics and sampling of activation states needed for catalysis. These allosteric networks and the ensuing changes resulting from second substrate binding are delineated using rigidity-based allosteric transmission theory and validated by nuclear magnetic resonance and functional studies. The results illustrate the role of dynamics along allosteric networks in facilitating function.


Asunto(s)
Hidrolasas/química , Hidrolasas/metabolismo , Regulación Alostérica , Sitios de Unión , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Entropía , Glicolatos/metabolismo , Hidrolasas/genética , Modelos Moleculares , Simulación de Dinámica Molecular , Resonancia Magnética Nuclear Biomolecular , Multimerización de Proteína , Subunidades de Proteína/metabolismo , Rhodopseudomonas/enzimología
9.
Cancer Cell ; 35(5): 721-737.e9, 2019 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-31056398

RESUMEN

The mitochondrial caseinolytic protease P (ClpP) plays a central role in mitochondrial protein quality control by degrading misfolded proteins. Using genetic and chemical approaches, we showed that hyperactivation of the protease selectively kills cancer cells, independently of p53 status, by selective degradation of its respiratory chain protein substrates and disrupts mitochondrial structure and function, while it does not affect non-malignant cells. We identified imipridones as potent activators of ClpP. Through biochemical studies and crystallography, we show that imipridones bind ClpP non-covalently and induce proteolysis by diverse structural changes. Imipridones are presently in clinical trials. Our findings suggest a general concept of inducing cancer cell lethality through activation of mitochondrial proteolysis.


Asunto(s)
Endopeptidasa Clp/genética , Endopeptidasa Clp/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Leucemia Mieloide Aguda/tratamiento farmacológico , Mitocondrias/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Endopeptidasa Clp/química , Femenino , Células HCT116 , Células HEK293 , Compuestos Heterocíclicos de 4 o más Anillos/química , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Imidazoles , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Ratones , Modelos Moleculares , Mutación Puntual , Conformación Proteica/efectos de los fármacos , Proteolisis , Piridinas , Pirimidinas , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Leukemia ; 33(1): 37-51, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29884901

RESUMEN

Acute myeloid leukemia (AML) is an aggressive hematologic malignancy for which new therapeutic approaches are required. One such potential therapeutic strategy is to target the ubiquitin-like modifier-activating enzyme 1 (UBA1), the initiating enzyme in the ubiquitylation cascade in which proteins are tagged with ubiquitin moieties to regulate their degradation or function. Here, we evaluated TAK-243, a first-in-class UBA1 inhibitor, in preclinical models of AML. In AML cell lines and primary AML samples, TAK-243 induced cell death and inhibited clonogenic growth. In contrast, normal hematopoietic progenitor cells were more resistant. TAK-243 preferentially bound to UBA1 over the related E1 enzymes UBA2, UBA3, and UBA6 in intact AML cells. Inhibition of UBA1 with TAK-243 decreased levels of ubiquitylated proteins, increased markers of proteotoxic stress and DNA damage stress. In vivo, TAK-243 reduced leukemic burden and targeted leukemic stem cells without evidence of toxicity. Finally, we selected populations of AML cells resistant to TAK-243 and identified missense mutations in the adenylation domain of UBA1. Thus, our data demonstrate that TAK-243 targets AML cells and stem cells and support a clinical trial of TAK-243 in this patient population. Moreover, we provide insight into potential mechanisms of acquired resistance to UBA1 inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Nucleósidos/farmacología , Sulfonamidas/farmacología , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Proliferación Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Ratones , Ratones SCID , Pirazoles , Pirimidinas , Sulfuros , Células Tumorales Cultivadas
11.
Nat Methods ; 15(11): 901-904, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30377366

RESUMEN

We present a 'hit-and-return' (HARE) method for time-resolved serial synchrotron crystallography with time resolution from milliseconds to seconds or longer. Timing delays are set mechanically, using the regular pattern in fixed-target crystallography chips and a translation stage system. Optical pump-probe experiments to capture intermediate structures of fluoroacetate dehalogenase binding to its ligand demonstrated that data can be collected at short (30 ms), medium (752 ms) and long (2,052 ms) intervals.


Asunto(s)
Cristalografía por Rayos X , Hidrolasas/química , Conformación Proteica , Rhodopseudomonas/enzimología , Sincrotrones/instrumentación , Diseño de Equipo , Modelos Moleculares , Factores de Tiempo
12.
MAbs ; 10(7): 1018-1029, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29969365

RESUMEN

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic growth factor that can stimulate a variety of cells, but its overexpression leads to excessive production and activation of granulocytes and macrophages with many pathogenic effects. This cytokine is a therapeutic target in inflammatory diseases, and several anti-GM-CSF antibodies have advanced to Phase 2 clinical trials in patients with such diseases, e.g., rheumatoid arthritis. GM-CSF is also an essential factor in preventing pulmonary alveolar proteinosis (PAP), a disease associated with GM-CSF malfunction arising most typically through the presence of GM-CSF neutralizing auto-antibodies. Understanding the mechanism of action for neutralizing antibodies that target GM-CSF is important for improving their specificity and affinity as therapeutics and, conversely, in devising strategies to reduce the effects of GM-CSF auto-antibodies in PAP. We have solved the crystal structures of human GM-CSF bound to antigen-binding fragments of two neutralizing antibodies, the human auto-antibody F1 and the mouse monoclonal antibody 4D4. Coordinates and structure factors of the crystal structures of the GM-CSF:F1 Fab and the GM-CSF:4D4 Fab complexes have been deposited in the RCSB Protein Data Bank under the accession numbers 6BFQ and 6BFS, respectively. The structures show that these antibodies bind to mutually exclusive epitopes on GM-CSF; however, both prevent the cytokine from interacting with its alpha receptor subunit and hence prevent receptor activation. Importantly, identification of the F1 epitope together with functional analyses highlighted modifications to GM-CSF that would abolish auto-antibody recognition whilst retaining GM-CSF function. These results provide a framework for developing novel GM-CSF molecules for PAP treatment and for optimizing current anti-GM-CSF antibodies for use in treating inflammatory disorders.


Asunto(s)
Anticuerpos Neutralizantes/química , Complejo Antígeno-Anticuerpo/química , Artritis Reumatoide/terapia , Autoanticuerpos/química , Epítopos/química , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunoterapia/métodos , Anticuerpos Neutralizantes/metabolismo , Artritis Reumatoide/inmunología , Autoanticuerpos/metabolismo , Autoanticuerpos/farmacología , Cristalografía por Rayos X , Citocinas/metabolismo , Epítopos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Estructura Molecular , Unión Proteica , Conformación Proteica
13.
Sci Rep ; 8(1): 8654, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29872131

RESUMEN

At times, it can be difficult to discern if a lack of overlap in reported interactions for a protein-of-interest reflects differences in methodology or biology. In such instances, systematic analyses of protein-protein networks across diverse paradigms can provide valuable insights. Here, we interrogated the interactome of the prion protein (PrP), best known for its central role in prion diseases, in four mouse cell lines. Analyses made use of identical affinity capture and sample processing workflows. Negative controls were generated from PrP knockout lines of the respective cell models, and the relative levels of peptides were quantified using isobaric labels. The study uncovered 26 proteins that reside in proximity to PrP. All of these proteins are predicted to have access to the outer face of the plasma membrane, and approximately half of them were not reported to interact with PrP before. Strikingly, although several proteins exhibited profound co-enrichment with PrP in a given model, except for the neural cell adhesion molecule 1, no protein was highly enriched in all PrP-specific interactomes. However, Gene Ontology analyses revealed a shared association of the majority of PrP candidate interactors with cellular events at the intersection of transforming growth factor ß and integrin signaling.


Asunto(s)
Integrinas/metabolismo , Proteínas Priónicas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Línea Celular , Ratones , Proteínas Priónicas/genética , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas
14.
ChemMedChem ; 13(9): 957-967, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29624890

RESUMEN

The tropical diseases human African trypanosomiasis, Chagas disease, and the various forms of leishmaniasis are caused by parasites of the family of trypanosomatids. These protozoa possess a unique redox metabolism based on trypanothione and trypanothione reductase (TR), making TR a promising drug target. We report the optimization of properties and potency of cyclohexylpyrrolidine inhibitors of TR by structure-based design. The best inhibitors were freely soluble and showed competitive inhibition constants (Ki ) against Trypanosoma (T.) brucei TR and T. cruzi TR and in vitro activities (half-maximal inhibitory concentration, IC50 ) against these parasites in the low micromolar range, with high selectivity against human glutathione reductase. X-ray co-crystal structures confirmed the binding of the ligands to the hydrophobic wall of the "mepacrine binding site" with the new, solubility-providing vectors oriented toward the surface of the large active site.


Asunto(s)
Inhibidores Enzimáticos/farmacología , NADH NADPH Oxidorreductasas/antagonistas & inhibidores , Pirrolidinas/farmacología , Tripanocidas/farmacología , Trypanosoma/efectos de los fármacos , Cristalografía por Rayos X , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Ligandos , Modelos Moleculares , Estructura Molecular , NADH NADPH Oxidorreductasas/metabolismo , Oxidación-Reducción , Pruebas de Sensibilidad Parasitaria , Pirrolidinas/síntesis química , Pirrolidinas/química , Solubilidad , Tripanocidas/síntesis química , Tripanocidas/química , Trypanosoma/metabolismo
15.
J Biomol Struct Dyn ; 36(2): 376-386, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28034013

RESUMEN

The crystal structures of protein SA0856 from Staphylococcus aureus in its apo-form and in complex with a Zn2+-ion have been presented. The 152 amino acid protein consists of two similar domains with α + ß topology. In both crystalline state and in solution, the protein forms a dimer with monomers related by a twofold pseudo-symmetry rotation axis. A sequence homology search identified the protein as a member of the structural family Glyoxalase I. We have shown that the enzyme possesses glyoxalase I activity in the presence of Zn2+, Mg2+, Ni2+, and Co2+, in this order of preference. Sequence and structure comparisons revealed that human glyoxalase I should be assigned to a subfamily A, while S. aureus glyoxalase I represents a new subfamily B, which includes also proteins from other bacteria. Both subfamilies have a similar protein chain fold but rather diverse sequences. The active sites of human and staphylococcus glyoxalases I are also different: the former contains one Zn-ion per chain; the latter incorporates two of these ions. In the active site of SA0856, the first Zn-ion is well coordinated by His58, Glu60 from basic molecule and Glu40*, His44* from adjacent symmetry-related molecule. The second Zn3-ion is coordinated only by residue His143 from protein molecule and one acetate ion. We suggest that only single Zn1-ion plays the role of catalytic center. The newly found differences between the two subfamilies could guide the design of new drugs against S. aureus, an important pathogenic micro-organism.


Asunto(s)
Lactoilglutatión Liasa/química , Staphylococcus aureus/química , Zinc/química , Secuencia de Aminoácidos/genética , Sitios de Unión , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Lactoilglutatión Liasa/genética , Modelos Moleculares , Conformación Proteica , Staphylococcus aureus/enzimología , Staphylococcus aureus/patogenicidad
16.
J Inherit Metab Dis ; 40(3): 423-431, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28205048

RESUMEN

BACKGROUND: Elevated urinary excretion of orotic acid is associated with treatable disorders of the urea cycle and pyrimidine metabolism. Establishing the correct and timely diagnosis in a patient with orotic aciduria is key to effective treatment. Uridine monophosphate synthase is involved in de novo pyrimidine synthesis. Uridine monophosphate synthase deficiency (or hereditary orotic aciduria), due to biallelic mutations in UMPS, is a rare condition presenting with megaloblastic anemia in the first months of life. If not treated with the pyrimidine precursor uridine, neutropenia, failure to thrive, growth retardation, developmental delay, and intellectual disability may ensue. METHODS AND RESULTS: We identified mild and isolated orotic aciduria in 11 unrelated individuals with diverse clinical signs and symptoms, the most common denominator being intellectual disability/developmental delay. Of note, none had blood count abnormalities, relevant hyperammonemia or altered plasma amino acid profile. All individuals were found to have heterozygous alterations in UMPS. Four of these variants were predicted to be null alleles with complete loss of function. The remaining variants were missense changes and predicted to be damaging to the normal encoded protein. Interestingly, family screening revealed heterozygous UMPS variants in combination with mild orotic aciduria in 19 clinically asymptomatic family members. CONCLUSIONS: We therefore conclude that heterozygous UMPS-mutations can lead to mild and isolated orotic aciduria without clinical consequence. Partial UMPS-deficiency should be included in the differential diagnosis of mild orotic aciduria. The discovery of heterozygotes manifesting clinical symptoms such as hypotonia and developmental delay are likely due to ascertainment bias.


Asunto(s)
Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Orotato Fosforribosiltransferasa/deficiencia , Orotidina-5'-Fosfato Descarboxilasa/deficiencia , Errores Innatos del Metabolismo de la Purina-Pirimidina/metabolismo , Anemia Megaloblástica/genética , Anemia Megaloblástica/metabolismo , Niño , Preescolar , Femenino , Heterocigoto , Humanos , Lactante , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Masculino , Mutación/genética , Orotato Fosforribosiltransferasa/genética , Orotato Fosforribosiltransferasa/metabolismo , Ácido Orótico/metabolismo , Orotidina-5'-Fosfato Descarboxilasa/genética , Orotidina-5'-Fosfato Descarboxilasa/metabolismo , Errores Innatos del Metabolismo de la Purina-Pirimidina/genética , Pirimidinas/metabolismo , Trastornos Innatos del Ciclo de la Urea/genética , Trastornos Innatos del Ciclo de la Urea/metabolismo , Uridina/metabolismo
17.
Science ; 355(6322)2017 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-28104837

RESUMEN

Freeze-trapping x-ray crystallography, nuclear magnetic resonance, and computational techniques reveal the distribution of states and their interconversion rates along the reaction pathway of a bacterial homodimeric enzyme, fluoroacetate dehalogenase (FAcD). The crystal structure of apo-FAcD exhibits asymmetry around the dimer interface and cap domain, priming one protomer for substrate binding. This asymmetry is dynamically averaged through conformational exchange on a millisecond time scale. During catalysis, the protomer conformational exchange rate becomes enhanced, the empty protomer exhibits increased local disorder, and water egresses. Computational studies identify allosteric pathways between protomers. Water release and enhanced dynamics associated with catalysis compensate for entropic losses from substrate binding while facilitating sampling of the transition state. The studies provide insights into how substrate-coupled allosteric modulation of structure and dynamics facilitates catalysis in a homodimeric enzyme.


Asunto(s)
Proteínas Bacterianas/química , Biocatálisis , Hidrolasas/química , Estructura Cuaternaria de Proteína , Rhodopseudomonas/enzimología , Regulación Alostérica , Cristalografía por Rayos X , Entropía , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Multimerización de Proteína , Especificidad por Sustrato , Agua/química
18.
J Immunol ; 196(11): 4723-30, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27183571

RESUMEN

The human Ab response to certain pathogens is oligoclonal, with preferred IgV genes being used more frequently than others. A pair of such preferred genes, IGVK3-11 and IGVH3-30, contributes to the generation of protective Abs directed against the 23F serotype of the pneumonococcal capsular polysaccharide of Streptococcus pneumoniae and against the AD-2S1 peptide of the gB membrane protein of human CMV. Structural analyses of Fab fragments of mAbs 023.102 and pn132p2C05 in complex with portions of the 23F polysaccharide revealed five germline-encoded residues in contact with the key component, l-rhamnose. In the case of the AD-2S1 peptide, the KE5 Fab fragment complex identified nine germline-encoded contact residues. Two of these germline-encoded residues, Arg91L and Trp94L, contact both the l-rhamnose and the AD-2S1 peptide. Comparison of the respective paratopes that bind to carbohydrate and protein reveals that stochastic diversity in both CDR3 loops alone almost exclusively accounts for their divergent specificity. Combined evolutionary pressure by human CMV and the 23F serotype of S. pneumoniae acted on the IGVK3-11 and IGVH3-30 genes as demonstrated by the multiple germline-encoded amino acids that contact both l-rhamnose and AD-2S1 peptide.


Asunto(s)
Antígenos/inmunología , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/genética , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Antígenos/química , Citomegalovirus/inmunología , Variación Genética , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Región Variable de Inmunoglobulina/química , Conformación Proteica , Streptococcus pneumoniae/inmunología
19.
PLoS One ; 11(3): e0149830, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26934472

RESUMEN

BACKGROUND: EspP (E. coli secreted serine protease, large plasmid encoded) is an extracellular serine protease produced by enterohemorrhagic E. coli (EHEC) O157:H7, a causative agent of diarrhea-associated Hemolytic Uremic Syndrome (D+HUS). The mechanism by which EHEC induces D+HUS has not been fully elucidated. OBJECTIVES: We investigated the effects of EspP on clot formation and lysis in human blood. METHODS: Human whole blood and plasma were incubated with EspP(WT )at various concentrations and sampled at various time points. Thrombin time (TT), prothrombin time (PT), and activated partial thromboplastin time (aPTT), coagulation factor activities, and thrombelastgraphy (TEG) were measured. RESULTS AND CONCLUSIONS: Human whole blood or plasma incubated with EspP(WT) was found to have prolonged PT, aPTT, and TT. Furthermore, human whole blood or plasma incubated with EspP(WT) had reduced activities of coagulation factors V, VII, VIII, and XII, as well as prothrombin. EspP did not alter the activities of coagulation factors IX, X, or XI. When analyzed by whole blood TEG, EspP decreased the maximum amplitude of the clot, and increased the clot lysis. Our results indicate that EspP alters hemostasis in vitro by decreasing the activities of coagulation factors V, VII, VIII, and XII, and of prothrombin, by reducing the clot strength and accelerating fibrinolysis, and provide further evidence of a functional role for this protease in the virulence of EHEC and the development of D+HUS.


Asunto(s)
Factores de Coagulación Sanguínea/metabolismo , Coagulación Sanguínea/fisiología , Escherichia coli Enterohemorrágica/metabolismo , Proteínas de Escherichia coli/metabolismo , Serina Endopeptidasas/metabolismo , Serina Proteasas/metabolismo , Trastornos de la Coagulación Sanguínea/metabolismo , Escherichia coli O157/metabolismo , Tiempo de Lisis del Coágulo de Fibrina/métodos , Fibrinólisis/fisiología , Síndrome Hemolítico-Urémico/metabolismo , Humanos , Tiempo de Tromboplastina Parcial/métodos , Protrombina/metabolismo , Tiempo de Protrombina/métodos , Trombosis/metabolismo
20.
Nat Commun ; 7: 10882, 2016 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-26947396

RESUMEN

Access to experimental X-ray diffraction image data is fundamental for validation and reproduction of macromolecular models and indispensable for development of structural biology processing methods. Here, we established a diffraction data publication and dissemination system, Structural Biology Data Grid (SBDG; data.sbgrid.org), to preserve primary experimental data sets that support scientific publications. Data sets are accessible to researchers through a community driven data grid, which facilitates global data access. Our analysis of a pilot collection of crystallographic data sets demonstrates that the information archived by SBDG is sufficient to reprocess data to statistics that meet or exceed the quality of the original published structures. SBDG has extended its services to the entire community and is used to develop support for other types of biomedical data sets. It is anticipated that access to the experimental data sets will enhance the paradigm shift in the community towards a much more dynamic body of continuously improving data analysis.


Asunto(s)
Bases de Datos Genéticas , Sustancias Macromoleculares/química , Publicaciones , Cristalografía por Rayos X , Internet , Programas Informáticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...