Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Proteins ; 85(4): 647-656, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28066922

RESUMEN

The retroviral integrase (IN) carries out the integration of a dsDNA copy of the viral genome into the host DNA, an essential step for viral replication. All IN proteins have three general domains, the N-terminal domain (NTD), the catalytic core domain, and the C-terminal domain. The NTD includes an HHCC zinc finger-like motif, which is conserved in all retroviral IN proteins. Two crystal structures of Moloney murine leukemia virus (M-MuLV) IN N-terminal region (NTR) constructs that both include an N-terminal extension domain (NED, residues 1-44) and an HHCC zinc-finger NTD (residues 45-105), in two crystal forms are reported. The structures of IN NTR constructs encoding residues 1-105 (NTR1-105 ) and 8-105 (NTR8-105 ) were determined at 2.7 and 2.15 Å resolution, respectively and belong to different space groups. While both crystal forms have similar protomer structures, NTR1-105 packs as a dimer and NTR8-105 packs as a tetramer in the asymmetric unit. The structure of the NED consists of three anti-parallel ß-strands and an α-helix, similar to the NED of prototype foamy virus (PFV) IN. These three ß-strands form an extended ß-sheet with another ß-strand in the HHCC Zn2+ binding domain, which is a unique structural feature for the M-MuLV IN. The HHCC Zn2+ binding domain structure is similar to that in HIV and PFV INs, with variations within the loop regions. Differences between the PFV and MLV IN NEDs localize at regions identified to interact with the PFV LTR and are compared with established biochemical and virological data for M-MuLV. Proteins 2017; 85:647-656. © 2016 Wiley Periodicals, Inc.


Asunto(s)
ADN Viral/química , Integrasas/química , Virus de la Leucemia Murina de Moloney/química , Proteínas Virales/química , Dedos de Zinc , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Clonación Molecular , Cristalografía por Rayos X , ADN Viral/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Integrasas/genética , Integrasas/metabolismo , Modelos Moleculares , Virus de la Leucemia Murina de Moloney/enzimología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Proteínas Virales/genética , Proteínas Virales/metabolismo
2.
J Am Chem Soc ; 137(40): 13106-13, 2015 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-26374198

RESUMEN

Celiac disease is characterized by intestinal inflammation triggered by gliadin, a component of dietary gluten. Oral administration of proteases that can rapidly degrade gliadin in the gastric compartment has been proposed as a treatment for celiac disease; however, no protease has been shown to specifically reduce the immunogenic gliadin content, in gastric conditions, to below the threshold shown to be toxic for celiac patients. Here, we used the Rosetta Molecular Modeling Suite to redesign the active site of the acid-active gliadin endopeptidase KumaMax. The resulting protease, Kuma030, specifically recognizes tripeptide sequences that are found throughout the immunogenic regions of gliadin, as well as in homologous proteins in barley and rye. Indeed, treatment of gliadin with Kuma030 eliminates the ability of gliadin to stimulate a T cell response. Kuma030 is capable of degrading >99% of the immunogenic gliadin fraction in laboratory-simulated gastric digestions within physiologically relevant time frames, to a level below the toxic threshold for celiac patients, suggesting great potential for this enzyme as an oral therapeutic for celiac disease.


Asunto(s)
Mucosa Gástrica/metabolismo , Gliadina/metabolismo , Péptido Hidrolasas/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Células Cultivadas , Humanos , Datos de Secuencia Molecular , Péptido Hidrolasas/química
3.
Proc Natl Acad Sci U S A ; 108(25): 10104-8, 2011 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-21636784

RESUMEN

The enzyme guanylyltransferase (GTase) plays a central role in the three-step catalytic process of adding an (m7)GpppN cap cotranscriptionally to nascent mRNA (pre-mRNAs). The 5'-mRNA capping process is functionally and evolutionarily conserved from unicellular organisms to human. However, the GTases from viruses and yeast have low amino acid sequence identity (∼25%) with GTases from mammals that, in contrast, are highly conserved (∼98%). We have defined by limited proteolysis of human capping enzyme residues 229-567 as comprising the minimum enzymatically active human GTase (hGTase) domain and have determined the structure by X-ray crystallography. Seven related conformational states of hGTase exist in the crystal. The GTP-binding site is evolutionarily and structurally conserved. The positional variations of the oligonucleotide/oligosaccharide binding fold lid domain over the GTP-binding site provide snapshots of the opening and closing of the active site cleft through a swivel motion. The pattern of conserved surface residues in mammals, but not in yeast, supports the finding that the recognition of the capping apparatus by RNA polymerase II and associated transcription factors is highly conserved in mammals, and the mechanism may differ somewhat from that in yeast. The hGTase structure should help in the design of biochemical and molecular biology experiments to explore the proteinprotein and proteinRNA interactions that ensure regulated transcription of genes in humans and other mammals.


Asunto(s)
Nucleotidiltransferasas/química , Animales , Sitios de Unión , Cristalografía por Rayos X , Análisis Mutacional de ADN , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Humanos , Datos de Secuencia Molecular , Nucleotidiltransferasas/genética , Estructura Terciaria de Proteína
4.
Proc Natl Acad Sci U S A ; 108(33): 13468-73, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21808041

RESUMEN

Interferon-induced ISG15 conjugation plays an important antiviral role against several viruses, including influenza viruses. The NS1 protein of influenza B virus (NS1B) specifically binds only human and nonhuman primate ISG15s and inhibits their conjugation. To elucidate the structural basis for the sequence-specific recognition of human ISG15, we determined the crystal structure of the complex formed between human ISG15 and the N-terminal region of NS1B (NS1B-NTR). The NS1B-NTR homodimer interacts with two ISG15 molecules in the crystal and also in solution. The two ISG15-binding sites on the NS1B-NTR dimer are composed of residues from both chains, namely residues in the RNA-binding domain (RBD) from one chain, and residues in the linker between the RBD and the effector domain from the other chain. The primary contact region of NS1B-NTR on ISG15 is composed of residues at the junction of the N-terminal ubiquitin-like (Ubl) domain and the short linker region between the two Ubl domains, explaining why the sequence of the short linker in human and nonhuman primate ISG15s is essential for the species-specific binding of these ISG15s. In addition, the crystal structure identifies NS1B-NTR binding sites in the N-terminal Ubl domain of ISG15, and shows that there are essentially no contacts with the C-terminal Ubl domain of ISG15. Consequently, NS1B-NTR binding to ISG15 would not occlude access of the C-terminal Ubl domain of ISG15 to its conjugating enzymes. Nonetheless, transfection assays show that NS1B-NTR binding of ISG15 is responsible for the inhibition of interferon-induced ISG15 conjugation in cells.


Asunto(s)
Citocinas/metabolismo , Virus de la Influenza B/química , Ubiquitinas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Humanos , Interferones/farmacología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas
5.
J Nucl Med ; 64(9): 1392-1398, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37385670

RESUMEN

223Ra, a targeted α-therapy, is approved for the treatment of patients with metastatic castration-resistant prostate cancer (mCRPC) who have bone metastases. In the phase 3 ALSYMPCA study, 223Ra prolonged survival and improved quality of life versus placebo. Our real-world study, PARABO, investigated pain and bone pain-related quality of life in patients with mCRPC and symptomatic bone metastases receiving 223Ra in clinical practice. Methods: PARABO was a prospective, observational, noninterventional single-arm study conducted in nuclear medicine centers across Germany (NCT02398526). The primary endpoint was a clinically meaningful pain response (≥2-point improvement from baseline for the worst-pain item score in the Brief Pain Inventory-Short Form). Results: The analysis included 354 patients, who received a median of 6 223Ra injections (range, 1-6). Sixty-seven percent (236/354) received 5-6 injections, and 33% (118/354) received 1-4 injections. Of 216 patients with a baseline worst-pain score of more than 1, 59% (128) had a clinically meaningful pain response during treatment. Corresponding rates were 67% (range, 98/146) with 5-6 223Ra injections versus 43% (range, 30/70) with 1-4 injections, 60% (range, 60/100) in patients with no more than 20 lesions versus 59% (range, 65/111) in those with more than 20 lesions, and 65% (range, 69/106) in patients without prior or concomitant opioid use versus 54% (range, 59/110) in those with prior or concomitant opioid use. Mean subscale scores (pain severity and pain interference) on the Brief Pain Inventory-Short Form improved during treatment. Conclusion: 223Ra reduced pain in patients with mCRPC and symptomatic bone metastases, particularly in patients who received 5-6 injections. The extent of metastatic disease did not impact pain response.


Asunto(s)
Neoplasias Óseas , Neoplasias de la Próstata Resistentes a la Castración , Radio (Elemento) , Humanos , Masculino , Analgésicos Opioides/uso terapéutico , Neoplasias Óseas/secundario , Dolor/complicaciones , Estudios Prospectivos , Neoplasias de la Próstata Resistentes a la Castración/patología , Calidad de Vida , Radio (Elemento)/uso terapéutico
6.
Thyroid ; 32(9): 1059-1068, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35950621

RESUMEN

Background: Sorafenib and lenvatinib are multikinase inhibitors (MKIs) approved for patients with radioactive iodine-refractory (RAI-R) differentiated thyroid cancer (DTC). There is no consensus on when to initiate MKI treatment. The objective of this study was to evaluate time to symptomatic progression (TTSP) in patients with RAI-R DTC for whom the decision to treat with an MKI was made at study entry. Methods: International, prospective, open-label, noninterventional cohort study (NCT02303444). Eligible patients had asymptomatic progressive RAI-R DTC, with ≥1 lesion ≥1 cm in diameter and life expectancy ≥6 months. The decision to treat with an MKI was at the treating physician's discretion. Primary endpoint was TTSP from study entry. Two cohorts were evaluated: patients for whom a decision to initiate an MKI was made at study entry (Cohort 1) and patients for whom there was a decision not to initiate an MKI at study entry (Cohort 2). Cohorts were compared descriptively. Results: The full analysis set (FAS) comprised 647 patients. The median duration of observation was 35.5 months (range <1-59.4). Of 344 MKI-treated patients, 209 received sorafenib, 191 received lenvatinib, and 19 received another MKI at some point. Median TTSP was 55.4 months (interquartile range [IQR] 18.6-not estimable [NE]) overall, 55.4 months (IQR 15.2-NE) in Cohort 1 (n = 169), and 51.4 months (IQR 20.0-NE) in Cohort 2 (n = 478). TTSP ≥36 months was achieved in 64.5% of patients overall, 59.5% of patients in Cohort 1, and 66.4% of patients in Cohort 2. Median overall survival from classification as RAI-R was 167 months and median progression-free survival from start of MKI therapy was 19.2 months and from start of sorafenib therapy 16.7 months. Among sorafenib-treated patients, 70% had dose modifications, 35% had a dose reduction, 89% experienced ≥1 treatment-emergent adverse event (TEAE), and 82% experienced ≥1 drug-related TEAE. Conclusions: This real-world study provides valuable insight into outcomes in patients with asymptomatic, progressive RAI-R DTC under observation or receiving MKI treatment. TTSP in the FAS provides insight into the current prognosis for patients with RAI-R DTC in the era of MKIs. Registration: NCT02303444.


Asunto(s)
Adenocarcinoma , Antineoplásicos , Neoplasias de la Tiroides , Antineoplásicos/uso terapéutico , Estudios de Cohortes , Humanos , Radioisótopos de Yodo/uso terapéutico , Compuestos de Fenilurea/uso terapéutico , Estudios Prospectivos , Inhibidores de Proteínas Quinasas/efectos adversos , Quinolinas , Sorafenib/uso terapéutico , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/patología , Neoplasias de la Tiroides/radioterapia
7.
Biochemistry ; 49(43): 9256-68, 2010 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-20836565

RESUMEN

Many functional proteins are at least partially disordered prior to binding. Although the structural transitions upon binding of disordered protein regions can influence the affinity and specificity of protein complexes, their precise energetic contributions to binding are unknown. Here, we use a model protein-protein interaction system in which a locally disordered region has been modified by directed evolution to quantitatively assess the thermodynamic and structural contributions to binding of disorder-to-order transitions. Through X-ray structure determination of the protein binding partners before and after complex formation and isothermal titration calorimetry of the interactions, we observe a correlation between protein ordering and binding affinity for complexes along this affinity maturation pathway. Additionally, we show that discrepancies between observed and calculated heat capacities based on buried surface area changes in the protein complexes can be explained largely by heat capacity changes that would result solely from folding the locally disordered region. Previously developed algorithms for predicting binding energies of protein-protein interactions, however, are unable to correctly model the energetic contributions of the structural transitions in our model system. While this highlights the shortcomings of current computational methods in modeling conformational flexibility, it suggests that the experimental methods used here could provide training sets of molecular interactions for improving these algorithms and further rationalizing molecular recognition in protein-protein interactions.


Asunto(s)
Unión Proteica , Proteínas/química , Proteínas/metabolismo , Termodinámica , Animales , Calorimetría , Evolución Molecular Dirigida , Ratones , Conformación Proteica , Proteínas/genética
8.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 66(Pt 12): 1567-71, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21139197

RESUMEN

Phosphatidylinositol 3-kinase (PI3K) proteins actively trigger signaling pathways leading to cell growth, proliferation and survival. These proteins have multiple isoforms and consist of a catalytic p110 subunit and a regulatory p85 subunit. The iSH2 domain of the p85ß isoform has been implicated in the binding of nonstructural protein 1 (NS1) of influenza A viruses. Here, the crystal structure of human p85ß iSH2 determined to 3.3 Šresolution is reported. The structure reveals that this domain mainly consists of a coiled-coil motif. Comparison with the published structure of the bovine p85ß iSH2 domain bound to the influenza A virus nonstructural protein 1 indicates that little or no structural change occurs upon complex formation. By comparing this human p85ß iSH2 structure with the bovine p85ß iSH2 domain, which shares 99% sequence identity, and by comparing the multiple conformations observed within the asymmetric unit of the bovine iSH2 structure, it was found that this coiled-coil domain exhibits a certain degree of conformational variability or `plasticity' in the interhelical turn region. It is speculated that this plasticity of p85ß iSH2 may play a role in regulating its functional and molecular-recognition properties.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ia/química , Animales , Bovinos , Cristalografía por Rayos X , Humanos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Homología Estructural de Proteína
9.
Trends Microbiol ; 15(3): 127-34, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17275309

RESUMEN

Peptidoglycan (PGN) is the major component of bacterial cell walls and one of the main microbial products recognized by the innate immune system. PGN recognition is mediated by several families of pattern recognition molecules, including Toll-like receptors, nucleotide-binding oligomerization domain-containing proteins, and peptidoglycan recognition proteins (PGRPs). However, only the interaction of PGN with PGRPs, which are highly conserved from insects to mammals, has so far been characterized at the molecular level. Here, we describe recent structural studies of PGRPs that reveal the basis for PGN recognition and provide insights into the signal transduction and antibacterial activities of these innate immune proteins.


Asunto(s)
Proteínas Portadoras/química , Inmunidad Innata , Peptidoglicano/química , Animales , Antibacterianos/química , Antibacterianos/metabolismo , Bacterias/efectos de los fármacos , Bacterias/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Portadoras/farmacología , Drosophila/metabolismo , Humanos , Ratones , Modelos Moleculares , Peptidoglicano/metabolismo , Transducción de Señal
10.
J Mol Biol ; 358(1): 157-71, 2006 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-16500675

RESUMEN

Large DNA viruses of the herpesvirus family produce proteins that mimic host MHC-I molecules as part of their immunoevasive strategy. The m144 glycoprotein, expressed by murine cytomegalovirus, is thought to be an MHC-I homolog whose expression prolongs viral survival in vivo by preventing natural killer cell activation. To explore the structural basis of this m144 function, we have determined the three-dimensional structure of an m144/beta2-microglobulin (beta2m) complex at 1.9A resolution. This structure reveals the canonical features of MHC-I molecules including readily identifiable alpha1, alpha2, and alpha3 domains. A unique disulfide bond links the alpha1 helix to the beta-sheet floor, explaining the known thermal stability of m144. Close juxtaposition of the alpha1 and alpha2 helices and the lack of critical residues that normally contribute to anchoring the peptide N and C termini eliminates peptide binding. A region of 13 amino acid residues, corresponding to the amino-terminal portion of the alpha2 helix, is missing in the electron density map, suggesting an area of structural flexibility that may be involved in ligand binding.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/química , Muromegalovirus/química , Proteínas Virales/química , Miembro 3 de la Subfamilia B de Transportadores de Casetes de Unión a ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Antígenos CD8/metabolismo , Carbohidratos/química , Línea Celular , Cristalografía por Rayos X , Expresión Génica , Células Asesinas Naturales/metabolismo , Ligandos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Homología de Secuencia , Microglobulina beta-2/metabolismo
11.
Structure ; 13(12): 1775-87, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16338406

RESUMEN

Although protein-protein interactions are involved in nearly all cellular processes, general rules for describing affinity and selectivity in protein-protein complexes are lacking, primarily because correlations between changes in protein structure and binding energetics have not been well determined. Here, we establish the structural basis of affinity maturation for a protein-protein interaction system that we had previously characterized energetically. This model system exhibits a 1500-fold affinity increase. Also, its affinity maturation is restricted by negative intramolecular cooperativity. With three complex and six unliganded variant X-ray crystal structures, we provide molecular snapshots of protein interface remodeling events that span the breadth of the affinity maturation process and present a comprehensive structural view of affinity maturation. Correlating crystallographically observed structural changes with measured energetic changes reveals molecular bases for affinity maturation, intramolecular cooperativity, and context-dependent binding.


Asunto(s)
Enterotoxinas/química , Modelos Moleculares , Fragmentos de Péptidos/química , Mapeo de Interacción de Proteínas , Receptores de Antígenos de Linfocitos T alfa-beta/química , Sustitución de Aminoácidos , Animales , Cristalografía por Rayos X , Enterotoxinas/genética , Ratones , Mutación , Fragmentos de Péptidos/genética , Conformación Proteica , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Agua/química
12.
Sci Rep ; 7(1): 3063, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28596570

RESUMEN

Crops expressing Bacillus thuringiensis (Bt)-derived insecticidal protein genes have been commercially available for over 15 years and are providing significant value to growers. However, there remains the need for alternative insecticidal actives due to emerging insect resistance to certain Bt proteins. A screen of bacterial strains led to the discovery of a two-component insecticidal protein named AfIP-1A/1B from an Alcaligenes faecalis strain. This protein shows selectivity against coleopteran insects including western corn rootworm (WCR). Transgenic maize plants expressing AfIP-1A/1B demonstrate strong protection from rootworm injury. Surprisingly, although little sequence similarity exists to known insecticidal proteins, efficacy tests using WCR populations resistant to two different Cry proteins show that AfIP-1A/1B and mCry3A differ in their mode of action while AfIP-1A/1B and the binary Cry34Ab1/Cry35Ab1 protein share a similar mode. These findings are supported by results of competitive binding assays and the similarity of the x-ray structure of AfIP-1A to Cry34Ab1. Our work indicates that insecticidal proteins obtained from a non-Bt bacterial source can be useful for developing genetically modified crops and can function similarly to familiar proteins from Bt.


Asunto(s)
Alcaligenes/genética , Proteínas Bacterianas/genética , Agentes de Control Biológico/toxicidad , Escarabajos/efectos de los fármacos , Endotoxinas/genética , Proteínas Hemolisinas/genética , Alcaligenes/metabolismo , Animales , Toxinas de Bacillus thuringiensis , Proteínas Bacterianas/toxicidad , Agentes de Control Biológico/metabolismo , Clonación Molecular , Escarabajos/patogenicidad , Endotoxinas/toxicidad , Proteínas Hemolisinas/toxicidad
13.
Science ; 355(6321): 201-206, 2017 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-28082595

RESUMEN

Active sites and ligand-binding cavities in native proteins are often formed by curved ß sheets, and the ability to control ß-sheet curvature would allow design of binding proteins with cavities customized to specific ligands. Toward this end, we investigated the mechanisms controlling ß-sheet curvature by studying the geometry of ß sheets in naturally occurring protein structures and folding simulations. The principles emerging from this analysis were used to design, de novo, a series of proteins with curved ß sheets topped with α helices. Nuclear magnetic resonance and crystal structures of the designs closely match the computational models, showing that ß-sheet curvature can be controlled with atomic-level accuracy. Our approach enables the design of proteins with cavities and provides a route to custom design ligand-binding and catalytic sites.


Asunto(s)
Conformación Proteica en Lámina beta , Ingeniería de Proteínas/métodos , Dominio Catalítico , Cristalografía por Rayos X , Ligandos , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Pliegue de Proteína
14.
Protein Sci ; 15(5): 1199-206, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16641493

RESUMEN

Peptidoglycan recognition proteins (PGRPs) are pattern recognition receptors of the innate immune system that bind bacterial peptidoglycans (PGNs). We determined the crystal structure, to 2.1 A resolution, of the C-terminal PGN-binding domain of human PGRP-I alpha in complex with a muramyl pentapeptide (MPP) from Gram-positive bacteria containing a complete peptide stem (L-Ala-D-isoGln-L-Lys-D-Ala-D-Ala). The structure reveals important features not observed previously in the complex between PGRP-I alpha and a muramyl tripeptide lacking D-Ala at stem positions 4 and 5. Most notable are ligand-induced structural rearrangements in the PGN-binding site that are essential for entry of the C-terminal portion of the peptide stem and for locking MPP in the binding groove. We propose that similar structural rearrangements to accommodate the PGN stem likely characterize many PGRPs, both mammalian and insect.


Asunto(s)
Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Bacterias Grampositivas/química , Peptidoglicano/química , Peptidoglicano/metabolismo , Pared Celular/metabolismo , Cristalografía por Rayos X , Humanos , Ligandos , Modelos Moleculares , Estructura Molecular , Ácidos Murámicos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína
15.
J Mol Biol ; 347(4): 683-91, 2005 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-15769462

RESUMEN

Peptidoglycan recognition proteins (PGRPs) are pattern recognition receptors of the innate immune system that bind peptidoglycans (PGNs) of bacterial cell walls. These molecules, which are highly conserved from insects to mammals, contribute to host defense against infections by both Gram-positive and Gram-negative bacteria. Here, we present the crystal structure of human PGRP-S at 1.70A resolution. The overall structure of PGRP-S, which participates in intracellular killing of Gram-positive bacteria, is similar to that of other PGRPs, including Drosophila PGRP-LB and PGRP-SA and human PGRP-Ialpha. However, comparison with these PGRPs reveals important differences in both the PGN-binding site and a groove formed by the PGRP-specific segment on the opposite face of the molecule. This groove, which may constitute a binding site for effector or signaling proteins, is less hydrophobic and deeper in PGRP-S than in PGRP-IalphaC, whose PGRP-specific segments vary considerably in amino acid sequence. By docking a PGN ligand into the PGN-binding cleft of PGRP-S based on the known structure of a PGRP-Ialpha-PGN complex, we identified potential PGN-binding residues in PGRP-S. Differences in PGN-contacting residues and interactions suggest that, although PGRPs may engage PGNs in a similar mode, structural differences exist that likely regulate the affinity and fine specificity of PGN recognition.


Asunto(s)
Proteínas Portadoras/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas Portadoras/metabolismo , Cristalografía por Rayos X , Proteínas de Drosophila/química , Humanos , Modelos Moleculares , Estructura Molecular , Peptidoglicano/química , Peptidoglicano/metabolismo , Estructura Terciaria de Proteína , Alineación de Secuencia
16.
Structure ; 24(9): 1562-72, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27545620

RESUMEN

Influenza viruses cause a highly contagious respiratory disease in humans. The NS1 proteins of influenza A and B viruses (NS1A and NS1B proteins, respectively) are composed of two domains, a dimeric N-terminal domain and a C-terminal domain, connected by a flexible polypeptide linker. Here we report the 2.0-Å X-ray crystal structure and nuclear magnetic resonance studies of the NS1B C-terminal domain, which reveal a novel and unexpected basic RNA-binding site that is not present in the NS1A protein. We demonstrate that single-site alanine replacements of basic residues in this site lead to reduced RNA-binding activity, and that recombinant influenza B viruses expressing these mutant NS1B proteins are severely attenuated in replication. This novel RNA-binding site of NS1B is required for optimal influenza B virus replication. Most importantly, this study reveals an unexpected RNA-binding function in the C-terminal domain of NS1B, a novel function that distinguishes influenza B viruses from influenza A viruses.


Asunto(s)
Virus de la Influenza A/genética , Virus de la Influenza B/genética , Mutación , ARN Viral/química , Proteínas de Unión al ARN/química , Proteínas no Estructurales Virales/química , Células A549 , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Secuencia Conservada , Cristalografía por Rayos X , Expresión Génica , Humanos , Virus de la Influenza A/metabolismo , Virus de la Influenza B/metabolismo , Cinética , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Especificidad por Sustrato , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
17.
Protein Sci ; 25(1): 30-45, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26293815

RESUMEN

We have developed an online NMR / X-ray Structure Pair Data Repository. The NIGMS Protein Structure Initiative (PSI) has provided many valuable reagents, 3D structures, and technologies for structural biology. The Northeast Structural Genomics Consortium was one of several PSI centers. NESG used both X-ray crystallography and NMR spectroscopy for protein structure determination. A key goal of the PSI was to provide experimental structures for at least one representative of each of hundreds of targeted protein domain families. In some cases, structures for identical (or nearly identical) constructs were determined by both NMR and X-ray crystallography. NMR spectroscopy and X-ray diffraction data for 41 of these "NMR / X-ray" structure pairs determined using conventional triple-resonance NMR methods with extensive sidechain resonance assignments have been organized in an online NMR / X-ray Structure Pair Data Repository. In addition, several NMR data sets for perdeuterated, methyl-protonated protein samples are included in this repository. As an example of the utility of this repository, these data were used to revisit questions about the precision and accuracy of protein NMR structures first outlined by Levy and coworkers several years ago (Andrec et al., Proteins 2007;69:449-465). These results demonstrate that the agreement between NMR and X-ray crystal structures is improved using modern methods of protein NMR spectroscopy. The NMR / X-ray Structure Pair Data Repository will provide a valuable resource for new computational NMR methods development.


Asunto(s)
Cristalografía por Rayos X , Bases de Datos de Proteínas , Resonancia Magnética Nuclear Biomolecular , Modelos Moleculares , Conformación Proteica , Proteínas/química
18.
J Endotoxin Res ; 11(1): 41-6, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15826377

RESUMEN

Peptidoglycan recognition proteins (PGRPs) are pattern recognition receptors of the innate immune system that bind, and in some cases hydrolyse, bacterial peptidoglycans (PGNs). We determined the crystal structure of the C-terminal PGN-binding domain of human PGRP-Ialpha in complex with a muramyl tripeptide representing the conserved core of lysine-type PGNs. The peptide stem of the ligand is buried at the deep end of a long binding groove, with N-acetylmuramic acid situated in the middle of the groove, whose shallow end could accommodate N-acetylglucosamine. Both peptide and glycan moieties are essential for binding by PGRPs. Conservation of key PGN-contacting residues indicates that all PGRPs employ this basic PGN-binding mode. The structure identifies variable residues that likely mediate discrimination between lysine- and diaminopimelic acid-type PGNs. In addition, we propose a mechanism for PGN hydrolysis by Zn2+-containing catalytic PGRPs.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/análogos & derivados , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Proteínas Portadoras/metabolismo , Cristalografía por Rayos X , Proteínas de Drosophila/metabolismo , Bacterias Grampositivas/química , Secuencia de Aminoácidos , Sitios de Unión , Proteínas Portadoras/química , Secuencia Conservada , Disulfuros/química , Glicina/química , Humanos , Enlace de Hidrógeno , Ligandos , Modelos Moleculares , Estructura Molecular , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
19.
J Mol Biol ; 341(5): 1189-204, 2004 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-15321715

RESUMEN

Non-proline cis peptide bonds have been observed in numerous protein crystal structures even though the energetic barrier to this conformation is significant and no non-prolyl-cis/trans-isomerase has been identified to date. While some external factors, such as metal binding or co-factor interaction, have been identified that appear to induce cis/trans isomerization of non-proline peptide bonds, the intrinsic structural basis for their existence and the mechanism governing cis/trans isomerization in proteins remains poorly understood. Here, we report the crystal structure of a newly isolated neurotoxin, the scorpion alpha-like toxin Buthus martensii Karsch (BmK) M7, at 1.4A resolution. BmK M7 crystallizes as a dimer in which the identical non-proline peptide bond between residues 9 and 10 exists either in the cis conformation or as a mixture of cis and trans conformations in either monomer. We also determined the crystal structures of several mutants of BmK M1, a representative scorpion alpha-like toxin that contains an identical non-proline cis peptide bond as that observed in BmK M7, in which residues within or neighboring the cis peptide bond were altered. Substitution of an aspartic acid residue for lysine at residue 8 in the BmK M1 (K8D) mutant converted the cis form of the non-proline peptide bond 9-10 into the trans form, revealing an intramolecular switch for cis-to-trans isomerization. Cis/trans interconversion of the switch residue at position 8 appears to be sequence-dependent as the peptide bond between residues 9 and 10 retains its wild-type cis conformation in the BmK M1 (K8Q) mutant structure. The structural interconversion of the isomeric states of the BmK M1 non-proline cis peptide bond may relate to the conversion of the scorpion alpha-toxins subgroups.


Asunto(s)
Neurotoxinas/química , Conformación Proteica , Venenos de Escorpión/química , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Dimerización , Isomerismo , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Estructura Molecular , Neurotoxinas/genética , Neurotoxinas/metabolismo , Prolina/química , Venenos de Escorpión/genética , Venenos de Escorpión/metabolismo , Escorpiones/química , Alineación de Secuencia
20.
Protein Pept Lett ; 9(5): 441-9, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12370033

RESUMEN

An alpha-like toxin named BmK M7 active on both mammals and insects has been purified from the venom of scorpion Buthus martensii Karsch (BmK) recently. The electrophysiological experiments showed that M7 can bind to human cardiac Na+-channel and modify its normal properties, hence can be considered as a cardiotoxin. Single crystals of M7 have been obtained by hanging-drop vapor diffusion method using ammonium sulfate as precipitant in Tris-HCl buffer at pH 8.5. A data set to 1.40 A resolution was collected using synchrotron radiation and CCD detector in Photon Factory in Japan. Data analysis showed that the crystals belonged to space group P3(1)21/P3(1)21, with cell dimensions a=b=32.76 A, c=176.82 A. Assuming two molecules per asymmetric unit, the Vm value is 1.92 A3/Da. The initial structural analysis was carried out by molecular replacement, which showed the correct space group (P3(1)21), and the orientations and positions of the two molecules in the asymmetric unit.


Asunto(s)
Corazón/efectos de los fármacos , Venenos de Escorpión/química , Animales , Bioensayo , Cristalización , Cristalografía por Rayos X , Moscas Domésticas/efectos de los fármacos , Larva/efectos de los fármacos , Venenos de Escorpión/aislamiento & purificación , Venenos de Escorpión/toxicidad , Bloqueadores de los Canales de Sodio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA