Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Surg ; 218(3): 462-466, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31288926

RESUMEN

BACKGROUND: The "weekend effect," whereby surgeries performed during weekend haven been associated with poorer postoperative outcomes. We explored whether Saturday elective procedures at our hospital were associated with poorer post-operative outcomes when compared with weekday surgeries. METHODS: A retrospective cohort study of patients undergoing elective surgery on the abdomen or perineum from 2008 to 2015 was performed. Procedures were classified by day (Group 1: Monday, Tuesday, Wednesday; Group 2: Saturday). Multivariate regression analyses were performed to determine group differences in procedure duration, length-of-stay (LOS) and complications. RESULTS: In adjusted analyses, there were no statistically significant differences between Group 1 (n = 816) and Group 2 (n = 269) procedures in terms of procedure duration (Group 2 - Group 1 = 13.6 min, p = .19), LOS (Group 2 - Group 1 = 1.9 days, p = .14) and complications (OR 0.58, p = .46). CONCLUSION: Saturday elective procedures were not associated with poorer outcomes.


Asunto(s)
Abdomen/cirugía , Atención Posterior/estadística & datos numéricos , Procedimientos Quirúrgicos Electivos , Tiempo de Internación/estadística & datos numéricos , Perineo/cirugía , Complicaciones Posoperatorias/epidemiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
2.
Anesth Analg ; 129(2): 418-425, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30320650

RESUMEN

BACKGROUND: We implemented a previously described barcode-based drug safety system in all of our anesthetizing locations. Providers were instructed to scan the barcode on syringes using our Anesthesia Information Management System before drug administration, but the rate of provider adherence was low. We studied an implementation intervention intended to increase the rate of scanning. METHODS: Using our Anesthesia Information Management System and Smart Anesthesia Manager software, we quantified syringe drug administrations by anesthesia providers with and without barcode scanning. We use an anesthesia team model in which an attending anesthesiologist is paired with a certified registered nurse anesthetist (CRNA) or a resident. Our system identified the pair of providers associated with a particular drug administration, but did not distinguish which providers actually administered the drug. Therefore, the rate of barcode scanning for a particular case was assigned to both providers equally. A baseline rate of scanning was established over a period of 17 months. An audit and feedback intervention was then performed that consisted of monthly performance reports sent by email to individual providers along with coffee gift card awards for top performers. The coffee gift cards were awarded in only the first 2 months of the intervention, while the email performance reports continued on a monthly basis. The coffee card awards were made public. The monthly emails reported the individual provider's rank order of performance relative to other providers, but was otherwise anonymous. The baseline rate of scanning was compared to the rate of scanning after the intervention for a period of 7 months. RESULTS: From November 2014 to March 2017, we accumulated 60,197 cases performed by 88 attending anesthesiologists, 65 CRNAs, and 148 residents. The total number of syringe drug administrations was 653,355. Average scanning performance improved from 8.7% of syringe barcodes scanned during the baseline period from November 2014 to February 2016 to 64.4% scanned during the period September 2016 to March 2017 (P < .001). Variation in performance among individuals was marked, ranging from 0% to 100% of syringes scanned. The performance of some individuals showed marked oscillation over time. There was greater variation in performance attributable to residents than in performance attributable to CRNAs. CONCLUSIONS: Feedback of individual provider performance data from the anesthesia information system to providers can be used in conjunction with other measures to improve performance. Despite improved average performance, there was marked variation in performance between individuals, and some individuals had marked oscillation of their performance over time.


Asunto(s)
Anestesiólogos/normas , Anestésicos/administración & dosificación , Etiquetado de Medicamentos/normas , Retroalimentación Formativa , Adhesión a Directriz/normas , Sistemas de Medicación en Hospital/normas , Enfermeras Anestesistas/normas , Pautas de la Práctica en Enfermería/normas , Pautas de la Práctica en Medicina/normas , Recompensa , Servicio de Anestesia en Hospital/normas , Anestesiólogos/educación , Anestesiólogos/psicología , Anestésicos/efectos adversos , Actitud del Personal de Salud , Conocimientos, Actitudes y Práctica en Salud , Humanos , Internado y Residencia , Auditoría Médica , Enfermeras Anestesistas/psicología , Estudios Prospectivos , Mejoramiento de la Calidad/normas , Indicadores de Calidad de la Atención de Salud/normas
3.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 9): 1194-200, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22948920

RESUMEN

The genome of the human intestinal parasite Giardia lamblia contains only a single aminoacyl-tRNA synthetase gene for each amino acid. The Giardia prolyl-tRNA synthetase gene product was originally misidentified as a dual-specificity Pro/Cys enzyme, in part owing to its unexpectedly high off-target activation of cysteine, but is now believed to be a normal representative of the class of archaeal/eukaryotic prolyl-tRNA synthetases. The 2.2 Å resolution crystal structure of the G. lamblia enzyme presented here is thus the first structure determination of a prolyl-tRNA synthetase from a eukaryote. The relative occupancies of substrate (proline) and product (prolyl-AMP) in the active site are consistent with half-of-the-sites reactivity, as is the observed biphasic thermal denaturation curve for the protein in the presence of proline and MgATP. However, no corresponding induced asymmetry is evident in the structure of the protein. No thermal stabilization is observed in the presence of cysteine and ATP. The implied low affinity for the off-target activation product cysteinyl-AMP suggests that translational fidelity in Giardia is aided by the rapid release of misactivated cysteine.


Asunto(s)
Aminoacil-ARNt Sintetasas/química , Giardia lamblia/química , Modelos Moleculares , Estructura Terciaria de Proteína
4.
J Appl Crystallogr ; 45(Pt 3): 598-602, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22675232

RESUMEN

The growth of diffracting crystals from purified proteins is often a major bottleneck in determining structures of biological and medical interest. The PROSPERO web server, http://skuld.bmsc.washington.edu/prospero, is intended both to provide a means of organizing the potentially large numbers of experimental characterizations measured from such proteins, and to provide useful guidance for structural biologists who have succeeded in purifying their target protein but have reached an impasse in the difficult and poorly understood process of turning purified protein into well diffracting crystals. These researchers need to decide which of many possible rescue options are worth pursuing, given finite resources. This choice is even more crucial when attempting to solve high-priority but relatively difficult structures of eukaryotic proteins. The site currently uses the HyGX1 predictor, which was trained and validated on protein samples from pathogenic protozoa (eukaryotes) using results from six types of experiment. PROSPERO allows users to store, analyze and display multiple results for each sample, to group samples into projects, and to share results and predictions with collaborators.

5.
J Med Chem ; 55(14): 6342-51, 2012 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-22720744

RESUMEN

Urea-based methionyl-tRNA synthetase inhibitors were designed, synthesized, and evaluated for their potential toward treating human African trypanosomiasis (HAT). With the aid of a homology model and a structure-activity-relationship approach, low nM inhibitors were discovered that show high selectivity toward the parasite enzyme over the closest human homologue. These compounds inhibit parasite growth with EC(50) values as low as 0.15 µM while having low toxicity to mammalian cells. Two compounds (2 and 26) showed excellent membrane permeation in the MDR1-MDCKII model and encouraging oral pharmacokinetic properties in mice. Compound 2 was confirmed to enter the CNS in mice. Compound 26 had modest suppressive activity against Trpanosoma brucei rhodesiense in the mouse model, suggesting that more potent analogues or compounds with higher exposures need to be developed. The urea-based inhibitors are thus a promising starting point for further optimization toward the discovery of orally available and CNS active drugs to treat HAT.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Metionina-ARNt Ligasa/antagonistas & inhibidores , Trypanosoma brucei brucei/enzimología , Urea/química , Urea/farmacología , Administración Oral , Aminoquinolinas/química , Animales , Disponibilidad Biológica , Encéfalo/metabolismo , Línea Celular , Permeabilidad de la Membrana Celular , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Femenino , Humanos , Metionina-ARNt Ligasa/química , Metionina-ARNt Ligasa/metabolismo , Ratones , Modelos Moleculares , Conformación Proteica , Especificidad por Sustrato , Urea/metabolismo , Urea/farmacocinética
6.
Anal Bioanal Chem ; 401(5): 1585-91, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21750879

RESUMEN

Ultrafiltration provides a generic method to discover ligands for protein drug targets with millimolar to micromolar K(d), the typical range of fragment-based drug discovery. This method was tailored to a 96-well format, and cocktails of fragment-sized molecules, with molecular masses between 150 and 300 Da, were screened against medical structural genomics target proteins. The validity of the method was confirmed through competitive binding assays in the presence of ligands known to bind the target proteins.


Asunto(s)
Descubrimiento de Drogas/métodos , Proteínas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Ultrafiltración/métodos , Unión Competitiva , Escherichia coli/metabolismo , Ligandos , Plasmodium yoelii/metabolismo , Unión Proteica , Trypanosoma brucei brucei/metabolismo
7.
J Mol Biol ; 409(2): 159-76, 2011 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-21420975

RESUMEN

The single tyrosyl-tRNA synthetase (TyrRS) gene in trypanosomatid genomes codes for a protein that is twice the length of TyrRS from virtually all other organisms. Each half of the double-length TyrRS contains a catalytic domain and an anticodon-binding domain; however, the two halves retain only 17% sequence identity to each other. The structural and functional consequences of this duplication and divergence are unclear. TyrRS normally forms a homodimer in which the active site of one monomer pairs with the anticodon-binding domain from the other. However, crystal structures of Leishmania major TyrRS show that, instead, the two halves of a single molecule form a pseudo-dimer resembling the canonical TyrRS dimer. Curiously, the C-terminal copy of the catalytic domain has lost the catalytically important HIGH and KMSKS motifs characteristic of class I aminoacyl-tRNA synthetases. Thus, the pseudo-dimer contains only one functional active site (contributed by the N-terminal half) and only one functional anticodon recognition site (contributed by the C-terminal half). Despite biochemical evidence for negative cooperativity between the two active sites of the usual TyrRS homodimer, previous structures have captured a crystallographically-imposed symmetric state. As the L. major TyrRS pseudo-dimer is inherently asymmetric, conformational variations observed near the active site may be relevant to understanding how the state of a single active site is communicated across the dimer interface. Furthermore, substantial differences between trypanosomal TyrRS and human homologs are promising for the design of inhibitors that selectively target the parasite enzyme.


Asunto(s)
Flavonoides/metabolismo , Leishmania major/enzimología , Tirosina-ARNt Ligasa/química , Tirosina-ARNt Ligasa/metabolismo , Tirosina/análogos & derivados , Secuencia de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Flavonoles , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Multimerización de Proteína , Homología de Secuencia de Aminoácido , Tirosina/metabolismo
8.
Antimicrob Agents Chemother ; 55(5): 1982-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21282428

RESUMEN

Human African trypanosomiasis continues to be an important public health threat in extensive regions of sub-Saharan Africa. Treatment options for infected patients are unsatisfactory due to toxicity, difficult administration regimes, and poor efficacy of available drugs. The aminoacyl-tRNA synthetases were selected as attractive drug targets due to their essential roles in protein synthesis and cell survival. Comparative sequence analysis disclosed differences between the trypanosome and mammalian methionyl-tRNA synthetases (MetRSs) that suggested opportunities for selective inhibition using drug-like molecules. Experiments using RNA interference on the single MetRS of Trypanosoma brucei demonstrated that this gene product was essential for normal cell growth. Small molecules (diaryl diamines) similar to those shown to have potent activity on prokaryotic MetRS enzymes were synthesized and observed to have inhibitory activity on the T. brucei MetRS (50% inhibitory concentration, <50 nM) and on bloodstream forms of T. brucei cultures (50% effective concentration, as low as 4 nM). Twenty-one compounds had a close correlation between enzyme binding/inhibition and T. brucei growth inhibition, indicating that they were likely to be acting on the intended target. The compounds had minimal effects on mammalian cell growth at 20 µM, demonstrating a wide therapeutic index. The most potent compound was tested in the murine model of trypanosomiasis and demonstrated profound parasite suppression and delayed mortality. A homology model of the T. brucei MetRS based on other MetRS structures was used to model binding of the lead diaryl diamine compounds. Future studies will focus on improving the pharmacological properties of the MetRS inhibitors.


Asunto(s)
Metionina-ARNt Ligasa/antagonistas & inhibidores , Trypanosoma brucei brucei/efectos de los fármacos , Animales , Northern Blotting , Proliferación Celular/efectos de los fármacos , Diaminas/farmacología , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Tripanocidas/uso terapéutico , Trypanosoma brucei brucei/enzimología
9.
Mol Biochem Parasitol ; 177(1): 20-8, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21255615

RESUMEN

Tryptophanyl-tRNA synthetase (TrpRS) is an essential enzyme that is recognizably conserved across all forms of life. It is responsible for activating and attaching tryptophan to a cognate tRNA(Trp) molecule for use in protein synthesis. In some eukaryotes this original core function has been supplemented or modified through the addition of extra domains or the expression of variant TrpRS isoforms. The three TrpRS structures from pathogenic protozoa described here represent three illustrations of this malleability in eukaryotes. The Cryptosporidium parvum genome contains a single TrpRS gene, which codes for an N-terminal domain of uncertain function in addition to the conserved core TrpRS domains. Sequence analysis indicates that this extra domain, conserved among several apicomplexans, is related to the editing domain of some AlaRS and ThrRS. The C. parvum enzyme remains fully active in charging tRNA(Trp) after truncation of this extra domain. The crystal structure of the active, truncated enzyme is presented here at 2.4Å resolution. The Trypanosoma brucei genome contains separate cytosolic and mitochondrial isoforms of TrpRS that have diverged in their respective tRNA recognition domains. The crystal structure of the T. brucei cytosolic isoform is presented here at 2.8Å resolution. The Entamoeba histolytica genome contains three sequences that appear to be TrpRS homologs. However one of these, whose structure is presented here at 3.0Å resolution, has lost the active site motifs characteristic of the Class I aminoacyl-tRNA synthetase catalytic domain while retaining the conserved features of a fully formed tRNA(Trp) recognition domain. The biological function of this variant E. histolytica TrpRS remains unknown, but, on the basis of a completely conserved tRNA recognition region and evidence for ATP but not tryptophan binding, it is tempting to speculate that it may perform an editing function. Together with a previously reported structure of an unusual TrpRS from Giardia, these protozoan structures broaden our perspective on the extent of structural variation found in eukaryotic TrpRS homologs.


Asunto(s)
Cryptosporidium parvum/enzimología , Entamoeba histolytica/enzimología , Proteínas Protozoarias/química , Trypanosoma brucei brucei/enzimología , Triptófano-ARNt Ligasa/química , Secuencia de Aminoácidos , Sitios de Unión , Cryptosporidium parvum/química , Cryptosporidium parvum/genética , Cristalografía por Rayos X , Entamoeba histolytica/química , Entamoeba histolytica/genética , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Alineación de Secuencia , Trypanosoma brucei brucei/química , Trypanosoma brucei brucei/genética , Triptófano-ARNt Ligasa/genética , Triptófano-ARNt Ligasa/metabolismo
10.
Biochimie ; 93(3): 570-82, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21144880

RESUMEN

Leishmania parasites cause two million new cases of leishmaniasis each year with several hundreds of millions of people at risk. Due to the paucity and shortcomings of available drugs, we have undertaken the crystal structure determination of a key enzyme from Leishmania major in hopes of creating a platform for the rational design of new therapeutics. Crystals of the catalytic core of methionyl-tRNA synthetase from L. major (LmMetRS) were obtained with the substrates MgATP and methionine present in the crystallization medium. These crystals yielded the 2.0 Å resolution structure of LmMetRS in complex with two products, methionyladenylate and pyrophosphate, along with a Mg(2+) ion that bridges them. This is the first class I aminoacyl-tRNA synthetase (aaRS) structure with pyrophosphate bound. The residues of the class I aaRS signature sequence motifs, KISKS and HIGH, make numerous contacts with the pyrophosphate. Substantial differences between the LmMetRS structure and previously reported complexes of Escherichia coli MetRS (EcMetRS) with analogs of the methionyladenylate intermediate product are observed, even though one of these analogs only differs by one atom from the intermediate. The source of these structural differences is attributed to the presence of the product pyrophosphate in LmMetRS. Analysis of the LmMetRS structure in light of the Aquifex aeolicus MetRS-tRNA(Met) complex shows that major rearrangements of multiple structural elements of enzyme and/or tRNA are required to allow the CCA acceptor triplet to reach the methionyladenylate intermediate in the active site. Comparison with sequences of human cytosolic and mitochondrial MetRS reveals interesting differences near the ATP- and methionine-binding regions of LmMetRS, suggesting that it should be possible to obtain compounds that selectively inhibit the parasite enzyme.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Difosfatos/metabolismo , Leishmania major/enzimología , Metionina-ARNt Ligasa/química , Metionina-ARNt Ligasa/metabolismo , Metionina/análogos & derivados , Nucleótidos de Adenina/metabolismo , Adenosina Monofosfato/química , Adenosina Monofosfato/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Difosfatos/química , Escherichia coli/enzimología , Bacterias Gramnegativas/enzimología , Humanos , Magnesio/metabolismo , Metionina/química , Metionina/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Homología de Secuencia de Aminoácido , Triptófano-ARNt Ligasa/metabolismo
11.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 8): 889-900, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20693688

RESUMEN

The use of TLS (translation/libration/screw) models to describe anisotropic displacement of atoms within a protein crystal structure has become increasingly common. These models may be used purely as an improved methodology for crystallographic refinement or as the basis for analyzing inter-domain and other large-scale motions implied by the crystal structure. In either case it is desirable to validate that the crystallographic model, including the TLS description of anisotropy, conforms to our best understanding of protein structures and their modes of flexibility. A set of validation tests has been implemented that can be integrated into ongoing crystallographic refinement or run afterwards to evaluate a previously refined structure. In either case validation can serve to increase confidence that the model is correct, to highlight aspects of the model that may be improved or to strengthen the evidence supporting specific modes of flexibility inferred from the refined TLS model. Automated validation checks have been added to the PARVATI and TLSMD web servers and incorporated into the CCP4i user interface.


Asunto(s)
Cristalografía por Rayos X/métodos , Anisotropía , Bases de Datos de Proteínas , Internet , Modelos Químicos
12.
J Struct Biol ; 171(2): 238-43, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20438846

RESUMEN

The 2.1A crystal structure of tryptophanyl-tRNA synthetase (TrpRS) from the diplomonad Giardia lamblia reveals that the N-terminus of this class I aminoacyl-tRNA synthetase forms a 16-residue alpha-helix. This helix replaces a beta-hairpin that is required by human TrpRS for normal activity and has been inferred to play a similar role in all eukaryotic TrpRS. The primary sequences of TrpRS homologs from several basal eukaryotes including Giardia lack a set of three residues observed to stabilize interactions with this beta-hairpin in the human TrpRS. Thus the present structure suggests that the activation reaction mechanism of TrpRS from the basal eukaryote G. lamblia differs from that of higher eukaryotes. Furthermore, the protein as observed in the crystal forms an (alpha(2))(2) homotetramer. The canonical dimer interface observed in all previous structures of tryptophanyl-tRNA synthetases is maintained, but in addition each N-terminal alpha-helix reciprocally interlocks with the equivalent helix from a second dimer to form a dimer of dimers. Although we have no evidence for tetramer formation in vivo, modeling indicates that the crystallographically observed tetrameric structure would be compatible with the tRNA binding mode used by dimeric TrpRS and TyrRS.


Asunto(s)
Giardia lamblia/enzimología , Triptófano-ARNt Ligasa/química , Humanos , Modelos Moleculares , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Difracción de Rayos X
13.
J Struct Biol ; 171(1): 64-73, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20347992

RESUMEN

The great power of protein crystallography to reveal biological structure is often limited by the tremendous effort required to produce suitable crystals. A hybrid crystal growth predictive model is presented that combines both experimental and sequence-derived data from target proteins, including novel variables derived from physico-chemical characterization such as R(30), the ratio between a protein's DSF intensity at 30°C and at T(m). This hybrid model is shown to be more powerful than sequence-based prediction alone - and more likely to be useful for prioritizing and directing the efforts of structural genomics and individual structural biology laboratories.


Asunto(s)
Modelos Moleculares , Proteínas/química , Cristalización , Cristalografía por Rayos X , Interpretación Estadística de Datos , Análisis de Secuencia de Proteína
14.
J Mol Biol ; 397(2): 481-94, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20132829

RESUMEN

Crystal structures of histidyl-tRNA synthetase (HisRS) from the eukaryotic parasites Trypanosoma brucei and Trypanosoma cruzi provide a first structural view of a eukaryotic form of this enzyme and reveal differences from bacterial homologs. HisRSs in general contain an extra domain inserted between conserved motifs 2 and 3 of the Class II aminoacyl-tRNA synthetase catalytic core. The current structures show that the three-dimensional topology of this domain is very different in bacterial and archaeal/eukaryotic forms of the enzyme. Comparison of apo and histidine-bound trypanosomal structures indicates substantial active-site rearrangement upon histidine binding but relatively little subsequent rearrangement after reaction of histidine with ATP to form the enzyme's first reaction product, histidyladenylate. The specific residues involved in forming the binding pocket for the adenine moiety differ substantially both from the previously characterized binding site in bacterial structures and from the homologous residues in human HisRSs. The essentiality of the single HisRS gene in T. brucei is shown by a severe depression of parasite growth rate that results from even partial suppression of expression by RNA interference.


Asunto(s)
Histidina-ARNt Ligasa/química , Trypanosoma brucei brucei/enzimología , Trypanosoma cruzi/enzimología , Secuencia de Aminoácidos , Animales , Dominio Catalítico , Cristalografía por Rayos X , Técnicas de Silenciamiento del Gen , Genes Esenciales , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , Alineación de Secuencia , Trypanosoma brucei brucei/química , Trypanosoma cruzi/química
15.
J Mol Biol ; 396(5): 1244-59, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20070944

RESUMEN

Purine nucleoside phosphorylases (PNPs) and uridine phosphorylases (UPs) are closely related enzymes involved in purine and pyrimidine salvage, respectively, which catalyze the removal of the ribosyl moiety from nucleosides so that the nucleotide base may be recycled. Parasitic protozoa generally are incapable of de novo purine biosynthesis; hence, the purine salvage pathway is of potential therapeutic interest. Information about pyrimidine biosynthesis in these organisms is much more limited. Though all seem to carry at least a subset of enzymes from each pathway, the dependency on de novo pyrimidine synthesis versus salvage varies from organism to organism and even from one growth stage to another. We have structurally and biochemically characterized a putative nucleoside phosphorylase (NP) from the pathogenic protozoan Trypanosoma brucei and find that it is a homodimeric UP. This is the first characterization of a UP from a trypanosomal source despite this activity being observed decades ago. Although this gene was broadly annotated as a putative NP, it was widely inferred to be a purine nucleoside phosphorylase. Our characterization of this trypanosomal enzyme shows that it is possible to distinguish between PNP and UP activity at the sequence level based on the absence or presence of a characteristic UP-specificity insert. We suggest that this recognizable feature may aid in proper annotation of the substrate specificity of enzymes in the NP family.


Asunto(s)
Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Trypanosoma brucei brucei/enzimología , Uridina Fosforilasa/química , Uridina Fosforilasa/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Cartilla de ADN/genética , ADN Protozoario/genética , Genes Protozoarios , Metales/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/genética , Interferencia de ARN , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Trypanosoma brucei brucei/genética , Uridina Fosforilasa/antagonistas & inhibidores , Uridina Fosforilasa/genética
16.
Mol Biochem Parasitol ; 169(2): 95-100, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19874856

RESUMEN

The crystal structure of the aspartyl-tRNA synthetase from the eukaryotic parasite Entamoeba histolytica has been determined at 2.8Aresolution. Relative to homologous sequences, the E. histolytica protein contains a 43-residue insertion between the N-terminal anticodon binding domain and the C-terminal catalytic domain. The present structure reveals that this insertion extends an arm of the hinge region that has previously been shown to mediate interaction of aspartyl-tRNA synthetase with the cognate tRNA D-stem. Modeling indicates that this Entamoeba-specific insertion is likely to increase the interaction surface with the cognate tRNA(Asp). In doing so it may substitute functionally for an RNA-binding motif located in N-terminal extensions found in AspRS sequences from lower eukaryotes but absent in Entamoeba. The E. histolytica AspRS structure shows a well-ordered N-terminus that contributes to the AspRS dimer interface.


Asunto(s)
Aspartato-ARNt Ligasa/química , Entamoeba histolytica/química , Entamoeba histolytica/enzimología , Proteínas Protozoarias/química , Secuencia de Aminoácidos , Animales , Aspartato-ARNt Ligasa/genética , Cristalografía por Rayos X , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Alineación de Secuencia
17.
Curr Top Med Chem ; 9(18): 1678-87, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19929835

RESUMEN

The history of fragment-based drug discovery, with an emphasis on crystallographic methods, is sketched, illuminating various contributions, including our own, which preceded the industrial development of the method. Subsequently, the creation of the BMSC fragment cocktails library is described. The BMSC collection currently comprises 68 cocktails of 10 compounds that are shape-wise diverse. The utility of these cocktails for initiating lead discovery in structure-based drug design has been explored by soaking numerous protein crystals obtained by our MSGPP (Medical Structural Genomics of Pathogenic Protozoa) consortium. Details of the fragment selection and cocktail design procedures, as well as examples of the successes obtained are given. The BMSC Fragment Cocktail recipes are available free of charge and are in use in over 20 academic labs.


Asunto(s)
Descubrimiento de Drogas/métodos , Infecciones por Protozoos/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/química , Cristalografía por Rayos X , Diseño de Fármacos , Genoma de Protozoos , Humanos , Bibliotecas de Moléculas Pequeñas/farmacología
18.
J Mol Biol ; 381(4): 975-88, 2008 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-18602399

RESUMEN

Plasmodium and other apicomplexan parasites are deficient in purine biosynthesis, relying instead on the salvage of purines from their host environment. Therefore, interference with the purine salvage pathway is an attractive therapeutic target. The plasmodial enzyme adenosine deaminase (ADA) plays a central role in purine salvage and, unlike mammalian ADA homologs, has a further secondary role in methylthiopurine recycling. For this reason, plasmodial ADA accepts a wider range of substrates, as it is responsible for deamination of both adenosine and 5'-methylthioadenosine. The latter substrate is not accepted by mammalian ADA homologs. The structural basis for this natural difference in specificity between plasmodial and mammalian ADA has not been well understood. We now report crystal structures of Plasmodium vivax ADA in complex with adenosine, guanosine, and the picomolar inhibitor 2'-deoxycoformycin. These structures highlight a drastic conformational change in plasmodial ADA upon substrate binding that has not been observed for mammalian ADA enzymes. Further, these complexes illuminate the structural basis for the differential substrate specificity and potential drug selectivity between mammalian and parasite enzymes.


Asunto(s)
Inhibidores de la Adenosina Desaminasa , Adenosina Desaminasa/química , Antimaláricos/química , Malaria/enzimología , Parásitos/enzimología , Plasmodium vivax/enzimología , Adenosina/metabolismo , Secuencia de Aminoácidos , Animales , Antimaláricos/metabolismo , Apoproteínas/química , Sitios de Unión , Catálisis/efectos de los fármacos , Cristalografía por Rayos X , Humanos , Canales Iónicos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Plasmodium falciparum/enzimología , Estructura Secundaria de Proteína , Compuestos de Amonio Cuaternario/metabolismo , Ribosa/metabolismo , Alineación de Secuencia , Especificidad por Sustrato/efectos de los fármacos
19.
Artículo en Inglés | MEDLINE | ID: mdl-18540054

RESUMEN

The structure of a structural genomics target protein, Tbru020260AAA from Trypanosoma brucei, has been determined to a resolution of 2.2 A using multiple-wavelength anomalous diffraction at the Se K edge. This protein belongs to Pfam sequence family PF08538 and is only distantly related to previously studied members of the alpha/beta-hydrolase fold family. Structural superposition onto representative alpha/beta-hydrolase fold proteins of known function indicates that a possible catalytic nucleophile, Ser116 in the T. brucei protein, lies at the expected location. However, the present structure and by extension the other trypanosomatid members of this sequence family have neither sequence nor structural similarity at the location of other active-site residues typical for proteins with this fold. Together with the presence of an additional domain between strands beta6 and beta7 that is conserved in trypanosomatid genomes, this suggests that the function of these homologs has diverged from other members of the fold family.


Asunto(s)
Hidrolasas/química , Pliegue de Proteína , Trypanosoma brucei brucei/enzimología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cristalografía por Rayos X , Dimerización , Hidrolasas/aislamiento & purificación , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Serina/química
20.
Methods Mol Biol ; 426: 497-513, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18542886

RESUMEN

The Structural Genomics of Pathogenic Protozoa (SGPP) Consortium aimed to determine crystal structures of proteins from trypanosomatid and malaria parasites in a high throughput manner. The pipeline of target selection, protein production, crystallization, and structure determination, is sketched. Special emphasis is given to a number of technology developments including domain prediction, the use of "co-crystallants," and capillary crystallization. "Fragment cocktail crystallography" for medical structural genomics is also described.


Asunto(s)
Genómica/métodos , Plasmodium/genética , Proteínas Protozoarias/química , Trypanosomatina/genética , Animales , Cristalización , Cristalografía por Rayos X/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...