Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33836586

RESUMEN

Intracellular protein homeostasis is maintained by a network of chaperones that function to fold proteins into their native conformation. The eukaryotic TRiC chaperonin (TCP1-ring complex, also called CCT for cytosolic chaperonin containing TCP1) facilitates folding of a subset of proteins with folding constraints such as complex topologies. To better understand the mechanism of TRiC folding, we investigated the biogenesis of an obligate TRiC substrate, the reovirus σ3 capsid protein. We discovered that the σ3 protein interacts with a network of chaperones, including TRiC and prefoldin. Using a combination of cryoelectron microscopy, cross-linking mass spectrometry, and biochemical approaches, we establish functions for TRiC and prefoldin in folding σ3 and promoting its assembly into higher-order oligomers. These studies illuminate the molecular dynamics of σ3 folding and establish a biological function for TRiC in virus assembly. In addition, our findings provide structural and functional insight into the mechanism by which TRiC and prefoldin participate in the assembly of protein complexes.


Asunto(s)
Proteínas de la Cápside/metabolismo , Chaperonina con TCP-1/metabolismo , Chaperonas Moleculares/metabolismo , Reoviridae/metabolismo , Proteínas de la Cápside/química , Chaperonina con TCP-1/química , Microscopía por Crioelectrón , Espectrometría de Masas , Chaperonas Moleculares/química , Conformación Proteica , Pliegue de Proteína , Proteostasis
2.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30626675

RESUMEN

Human noroviruses (NoVs) are the main cause of epidemic and sporadic gastroenteritis. Phylogenetically, noroviruses are divided into seven genogroups, with each divided into multiple genotypes. NoVs belonging to genogroup II and genotype 4 (GII.4) are globally most prevalent. Genetic diversity among the NoVs and the periodic emergence of novel strains present a challenge for the development of vaccines and antivirals to treat NoV infection. NoV protease is essential for viral replication and is an attractive target for the development of antivirals. The available structure of GI.1 protease provided a basis for the design of inhibitors targeting the active site of the protease. These inhibitors, although potent against the GI proteases, poorly inhibit the GII proteases, for which structural information is lacking. To elucidate the structural basis for this difference in the inhibitor efficiency, we determined the crystal structure of a GII.4 protease. The structure revealed significant changes in the S2 substrate-binding pocket, making it noticeably smaller, and in the active site, with the catalytic triad residues showing conformational changes. Furthermore, a conserved arginine is found inserted into the active site, interacting with the catalytic histidine and restricting substrate/inhibitor access to the S2 pocket. This interaction alters the relationships between the catalytic residues and may allow for a pH-dependent regulation of protease activity. The changes we observed in the GII.4 protease structure may explain the reduced potency of the GI-specific inhibitors against the GII protease and therefore must be taken into account when designing broadly cross-reactive antivirals against NoVs.IMPORTANCE Human noroviruses (NoVs) cause sporadic and epidemic gastroenteritis worldwide. They are divided into seven genogroups (GI to GVII), with each genogroup further divided into several genotypes. Human NoVs belonging to genogroup II and genotype 4 (GII.4) are the most prevalent. Currently, there are no vaccines or antiviral drugs available for NoV infection. The protease encoded by NoV is considered a valuable target because of its essential role in replication. NoV protease structures have only been determined for the GI genogroup. We show here that the structure of the GII.4 protease exhibits several significant changes from GI proteases, including a unique pairing of an arginine with the catalytic histidine that makes the proteolytic activity of GII.4 protease pH sensitive. A comparative analysis of NoV protease structures may provide a rational framework for structure-based drug design of broadly cross-reactive inhibitors targeting NoVs.


Asunto(s)
Arginina/metabolismo , Dominio Catalítico/genética , Histidina/metabolismo , Norovirus/metabolismo , Péptido Hidrolasas/metabolismo , Secuencia de Aminoácidos , Infecciones por Caliciviridae/metabolismo , Dominio Catalítico/fisiología , Variación Genética/genética , Genotipo , Humanos , Concentración de Iones de Hidrógeno , Norovirus/genética , Filogenia , Proteolisis
3.
Proc Natl Acad Sci U S A ; 113(40): E5830-E5837, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27647885

RESUMEN

Human noroviruses (HuNoVs) cause sporadic and epidemic gastroenteritis worldwide. They are classified into two major genogroups (GI and GII), with each genogroup further divided into multiple genotypes. Susceptibility to these viruses is influenced by genetically determined histo-blood group antigen (HBGA) expression. HBGAs function as cell attachment factors by binding to a surface-exposed region in the protruding (P) domain of the capsid protein. Sequence variations in this region that result in differential HBGA binding patterns and antigenicity are suggested to form a basis for strain diversification. Recent studies show that serum antibodies that block HBGA binding correlate with protection against illness. Although genogroup-dependent variation in HBGA binding specificity is structurally well characterized, an understanding of how antibodies block HBGA binding and how genotypic variations affect such blockade is lacking. Our crystallographic studies of the GI.1 P domain in complex with the Fab fragment of a human IgA monoclonal antibody (IgA 5I2) with HBGA blocking activity show that the antibody recognizes a conformational epitope formed by two surface-exposed loop clusters in the P domain. The antibody engulfs the HBGA binding site but does not affect its structural integrity. An unusual feature of the antigen recognition by IgA 5I2 is the predominant involvement of the CDR light chain 1 in contrast to the commonly observed CDR heavy chain 3, providing a unique perspective into antibody diversity in antigen recognition. Identification of the antigenic site in the P domain shows how genotypic variations might allow escape from antibody neutralization and exemplifies the interplay between antigenicity and HBGA specificity in HuNoV evolution.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Antígenos de Grupos Sanguíneos/inmunología , Inmunoglobulina A/metabolismo , Pruebas de Neutralización , Norovirus/inmunología , Secuencia de Aminoácidos , Antígenos/química , Cristalografía por Rayos X , Epítopos/química , Genotipo , Humanos , Fragmentos Fab de Inmunoglobulinas/metabolismo , Modelos Moleculares , Norovirus/efectos de los fármacos , Norovirus/genética , Dominios Proteicos , Proteínas Virales/química , Proteínas Virales/metabolismo
4.
J Biol Chem ; 292(52): 21366-21380, 2017 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-29123034

RESUMEN

Studies of virulence determinants in the bacterial phytopathogen Erwinia amylovora, the cause of devastating fire blight disease in apple and pear, have shown that HsvA, a putative amidinotransferase enzyme located in the Hrp pathogenicity island, is required for systemic infection in apple. However, the mechanism by which HsvA contributes to virulence is unclear. To investigate the role of HsvA in virulence, we carried out a series of biochemical and structural studies to characterize the amidinotransferase activity of HsvA. We found that HsvA displays a preference for linear aliphatic polyamines as the amidino acceptor substrate, especially for spermidine and putrescine (Km values of 33 µm and 3.9 mm, respectively). The three-dimensional structure, determined at 2.30 Å resolution using X-ray crystallography, revealed that the overall architecture of HsvA is similar to that of the human arginine-glycine amidinotransferase in the creatine biosynthesis pathway. The active site is located in the core of the protein at the base of a long, narrow substrate access channel. Specific amino acids near the entrance of the channel may serve as major determinants of the substrate specificity, including a glutamate residue at the rim of the channel entrance that appears to be positioned to interact with the distal primary amine in the putrescine substrate as well as the internal and distal amines in the spermidine substrate. These results suggest potential in vivo functions for HsvA as a virulence factor in fire blight and may also provide a basis for strategies to control fire blight by inhibiting HsvA activity.


Asunto(s)
Amidinotransferasas/metabolismo , Erwinia amylovora/metabolismo , Amidinotransferasas/fisiología , Cristalografía por Rayos X/métodos , Erwinia amylovora/patogenicidad , Islas Genómicas/genética , Islas Genómicas/fisiología , Malus/microbiología , Enfermedades de las Plantas/microbiología , Poliaminas/metabolismo , Pyrus/microbiología , Virulencia , Factores de Virulencia/metabolismo
5.
PLoS Pathog ; 12(6): e1005719, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27355511

RESUMEN

Noroviruses (NoV) are the most common cause of non-bacterial acute gastroenteritis and cause local outbreaks of illness, especially in confined situations. Despite being identified four decades ago, the correlates of protection against norovirus gastroenteritis are still being elucidated. Recent studies have shown an association of protection with NoV-specific serum histo-blood group antigen-blocking antibody and with serum IgA in patients vaccinated with NoV VLPs. Here, we describe the isolation and characterization of human monoclonal IgG and IgA antibodies against a GI.I NoV, Norwalk virus (NV). A higher proportion of the IgA antibodies blocked NV VLP binding to glycans than did IgG antibodies. We generated isotype-switched variants of IgG and IgA antibodies to study the effects of the constant domain on blocking and binding activities. The IgA form of antibodies appears to be more potent than the IgG form in blocking norovirus binding to histo-blood group antigens. These studies suggest a unique role for IgA antibodies in protection from NoV infections by blocking attachment to cell receptors.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Linfocitos B/inmunología , Antígenos de Grupos Sanguíneos/inmunología , Infecciones por Caliciviridae/inmunología , Western Blotting , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Gastroenteritis/inmunología , Humanos , Hibridomas , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Virus Norwalk/inmunología , Reacción en Cadena de la Polimerasa
6.
J Virol ; 88(11): 6168-80, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24648450

RESUMEN

UNLABELLED: Human noroviruses (NoVs) cause acute epidemic gastroenteritis. Susceptibility to the majority of NoV infections is determined by genetically controlled secretor-dependent expression of histo-blood group antigens (HBGAs), which are also critical for NoV attachment to host cells. Human NoVs are classified into two major genogroups (genogroup I [GI] and GII), with each genogroup further divided into several genotypes. GII NoVs are more prevalent and exhibit periodic emergence of new variants, suggested to be driven by altered HBGA binding specificities and antigenic drift. Recent epidemiological studies show increased activity among GI NoVs, with some members showing the ability to bind nonsecretor HBGAs. NoVs bind HBGAs through the protruding (P) domain of the major capsid protein VP1. GI NoVs, similar to GII, exhibit significant sequence variations in the P domain; it is unclear how these variations affect HBGA binding specificities. To understand the determinants of possible strain-specific HBGA binding among GI NoVs, we determined the structure of the P domain of a GI.7 clinical isolate and compared it to the previously determined P domain structures of GI.1 and GI.2 strains. Our crystallographic studies revealed significant structural differences, particularly in the loop regions of the GI.7 P domain, altering its surface topography and electrostatic landscape and potentially indicating antigenic variation. The GI.7 strain bound to H- and A-type, Lewis secretor, and Lewis nonsecretor families of HBGAs, allowing us to further elucidate the structural determinants of nonsecretor HBGA binding among GI NoVs and to infer several contrasting and generalizable features of HBGA binding in the GI NoVs. IMPORTANCE: Human noroviruses (NoVs) cause acute epidemic gastroenteritis. Recent epidemiological studies have shown increased prevalence of genogroup I (GI) NoVs. Although secretor-positive status is strongly correlated with NoV infection, cases of NoV infection associated with secretor-negative individuals are reported. Biochemical studies have shown that GI NoVs exhibit genotype-dependent binding to nonsecretor histo-blood group antigens (HBGAs). From our crystallographic studies of a GI.7 NoV, in comparison with previous studies on GI.1 and GI.2 NoVs, we show that genotypic differences translate to extensive structural changes in the loop regions that significantly alter the surface topography and electrostatic landscape of the P domain; these features may be indicative of antigenic variations contributing to serotypic differentiation in GI NoVs and also differential modulation of the HBGA binding characteristics. A significant finding is that the threshold length and the structure of one of the loops are critical determinants in the binding of GI NoVs to nonsecretor HBGAs.


Asunto(s)
Antígenos Virales/metabolismo , Epítopos/metabolismo , Modelos Moleculares , Norovirus/metabolismo , Proteínas Estructurales Virales/química , Acoplamiento Viral , Secuencia de Aminoácidos , Secuencia de Bases , Western Blotting , Cristalografía , Dimerización , Ensayo de Inmunoadsorción Enzimática , Epítopos/genética , Genotipo , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Alineación de Secuencia , Análisis de Secuencia de ADN , Proteínas Estructurales Virales/metabolismo , Difracción de Rayos X
7.
J Virol ; 87(8): 4281-92, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23365454

RESUMEN

Norwalk virus (NV), the prototype human calicivirus, is the leading cause of nonbacterial acute gastroenteritis. The NV protease cleaves the polyprotein encoded by open reading frame 1 of the viral genome at five nonhomologous sites, releasing six nonstructural proteins that are essential for viral replication. The structural details of how NV protease recognizes multiple substrates are unclear. In our X-ray structure of an NV protease construct, we observed that the C-terminal tail, representing the native substrate positions P5 to P1, is inserted into the active site cleft of the neighboring protease molecule, providing atomic details of how NV protease recognizes a substrate. The crystallographic structure of NV protease with the C-terminal tail redesigned to mimic P4 to P1 of another substrate site provided further structural details on how the active site accommodates sequence variations in the substrates. Based on these structural analyses, substrate-based aldehyde inhibitors were synthesized and screened for inhibition potency. Crystallographic structures of the protease in complex with each of the three most potent inhibitors were determined. These structures showed concerted conformational changes in the S4 and S2 pockets of the protease to accommodate variations in the P4 and P2 residues of the substrate/inhibitor, which could be a mechanism for how the NV protease recognizes multiple sites in the polyprotein with differential affinities during virus replication. These structures further indicate that the mechanism of inhibition by these inhibitors involves covalent bond formation with the side chain of the conserved cysteine in the active site by nucleophilic addition, and such substrate-based aldehydes could be effective protease inhibitors.


Asunto(s)
Virus Norwalk/efectos de los fármacos , Virus Norwalk/enzimología , Péptido Hidrolasas/química , Péptido Hidrolasas/metabolismo , Inhibidores de Proteasas/metabolismo , Cristalografía por Rayos X , Modelos Moleculares , Conformación Proteica , Especificidad por Sustrato
8.
Biochem J ; 451(1): 33-44, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23289588

RESUMEN

PKC (protein kinase C) θ is predominantly expressed in T-cells and is critically involved in immunity. Design of PKCθ-selective molecules to manage autoimmune disorders by targeting its activator-binding C1 domain requires the knowledge of its structure and the activator-binding residues. The C1 domain consists of twin C1 domains, C1A and C1B, of which C1B plays a critical role in the membrane translocation and activation of PKCθ. In the present study we determined the crystal structure of PKCθC1B to 1.63 Å (1 Å=0.1 nm) resolution, which showed that Trp(253) at the rim of the activator-binding pocket was orientated towards the membrane, whereas in PKCδC1B the homologous tryptophan residue was orientated away from the membrane. This particular orientation of Trp(253) affects the size of the activator-binding pocket and the membrane affinity. To further probe the structural constraints on activator-binding, five residues lining the activator-binding site were mutated (Y239A, T243A, W253G, L255G and Q258G) and the binding affinities of the PKCθC1B mutants were measured. These mutants showed reduced binding affinities for phorbol ester [PDBu (phorbol 12,13-dibutyrate)] and diacylglycerol [DOG (sn-1,2-dioctanoylglycerol), SAG (sn-1-stearoyl 2-arachidonyl glycerol)]. All five full-length PKCθ mutants exhibited reduced phorbol-ester-induced membrane translocation compared with the wild-type. These results provide insights into the PKCθ activator-binding domain, which will aid in future design of PKCθ-selective molecules.


Asunto(s)
Activadores de Enzimas/química , Isoenzimas/metabolismo , Proteína Quinasa C/metabolismo , Sustitución de Aminoácidos , Animales , Sitios de Unión , Activadores de Enzimas/metabolismo , Isoenzimas/química , Isoenzimas/genética , Ratones , Mutación Missense , Proteína Quinasa C/química , Proteína Quinasa C/genética , Proteína Quinasa C-theta , Estructura Terciaria de Proteína , Transporte de Proteínas , Triptófano/química , Triptófano/genética , Triptófano/metabolismo
9.
Nat Commun ; 14(1): 6516, 2023 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-37845211

RESUMEN

Acute gastroenteritis caused by human noroviruses (HuNoVs) is a significant global health and economic burden and is without licensed vaccines or antiviral drugs. The GII.4 HuNoV causes most epidemics worldwide. This virus undergoes epochal evolution with periodic emergence of variants with new antigenic profiles and altered specificity for histo-blood group antigens (HBGA), the determinants of cell attachment and susceptibility, hampering the development of immunotherapeutics. Here, we show that a llama-derived nanobody M4 neutralizes multiple GII.4 variants with high potency in human intestinal enteroids. The crystal structure of M4 complexed with the protruding domain of the GII.4 capsid protein VP1 revealed a conserved epitope, away from the HBGA binding site, fully accessible only when VP1 transitions to a "raised" conformation in the capsid. Together with dynamic light scattering and electron microscopy of the GII.4 VLPs, our studies suggest a mechanism in which M4 accesses the epitope by altering the conformational dynamics of the capsid and triggering its disassembly to neutralize GII.4 infection.


Asunto(s)
Antígenos de Grupos Sanguíneos , Infecciones por Caliciviridae , Norovirus , Humanos , Proteínas de la Cápside/química , Cápside/metabolismo , Norovirus/genética , Sitios de Unión , Epítopos/metabolismo , Antígenos de Grupos Sanguíneos/metabolismo
10.
J Biol Chem ; 286(6): 4329-40, 2011 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-21115497

RESUMEN

Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accordingly, it has been proposed for use as an anticoagulant. Full-length TFPI-2 or its isolated first Kunitz domain (KD1) also inhibits plasmin; therefore, it has been proposed for use as an antifibrinolytic agent. However, the anticoagulant properties of TFPI-2 or KD1 would diminish its antifibrinolytic function. In this study, structure-based investigations and analysis of the serine protease profiles revealed that coagulation enzymes prefer a hydrophobic residue at the P2' position in their substrates/inhibitors, whereas plasmin prefers a positively charged arginine residue at the corresponding position in its substrates/inhibitors. Based upon this observation, we changed the P2' residue Leu-17 in KD1 to Arg (KD1-L17R) and compared its inhibitory properties with wild-type KD1 (KD1-WT). Both WT and KD1-L17R were expressed in Escherichia coli, folded, and purified to homogeneity. N-terminal sequences and mass spectra confirmed proper expression of KD1-WT and KD1-L17R. Compared with KD1-WT, the KD1-L17R did not inhibit factor XIa, plasma kallikrein, or factor VIIa/tissue factor. Furthermore, KD1-L17R inhibited plasmin with ∼6-fold increased affinity and effectively prevented plasma clot fibrinolysis induced by tissue plasminogen activator. Similarly, in a mouse liver laceration bleeding model, KD1-L17R was ∼8-fold more effective than KD1-WT in preventing blood loss. Importantly, in this bleeding model, KD1-L17R was equally or more effective than aprotinin or tranexamic acid, which have been used as antifibrinolytic agents to prevent blood loss during major surgery/trauma. Furthermore, as compared with aprotinin, renal toxicity was not observed with KD1-L17R.


Asunto(s)
Sustitución de Aminoácidos , Antifibrinolíticos , Fibrinólisis/efectos de los fármacos , Glicoproteínas , Hemorragia/tratamiento farmacológico , Pliegue de Proteína , Animales , Antifibrinolíticos/química , Antifibrinolíticos/farmacología , Aprotinina/química , Aprotinina/farmacología , Factores de Coagulación Sanguínea/química , Modelos Animales de Enfermedad , Escherichia coli , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/farmacología , Humanos , Ratones , Mutación Missense , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Relación Estructura-Actividad , Ácido Tranexámico/química , Ácido Tranexámico/farmacología
11.
Antimicrob Agents Chemother ; 56(11): 5667-77, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22908171

RESUMEN

Metallo-ß-lactamases catalyze the hydrolysis of a broad range of ß-lactam antibiotics and are a concern for the spread of drug resistance. To analyze the determinants of enzyme structure and function, the sequence requirements for the subclass B1 IMP-1 ß-lactamase zinc binding residue Cys221 were tested by saturation mutagenesis and evaluated for protein expression, as well as hydrolysis of ß-lactam substrates. The results indicated that most substitutions at position 221 destabilized the enzyme. Only the enzymes containing C221D and C221G substitutions were expressed well in Escherichia coli and exhibited catalytic activity toward ß-lactam antibiotics. Despite the lack of a metal-chelating group at position 221, the C221G enzyme exhibited high levels of catalytic activity in the presence of exogenous zinc. Molecular modeling suggests the glycine substitution is unique among substitutions in that the complete removal of the cysteine side chain allows space for a water molecule to replace the thiol and coordinate zinc at the Zn2 zinc binding site to restore function. Multiple methods were used to estimate the C221G Zn2 binding constant to be 17 to 43 µM. Studies of enzyme function in vivo in E. coli grown on minimal medium showed that both IMP-1 and the C221G mutant exhibited compromised activity when zinc availability was low. Finally, substitutions at residue 121, which is the IMP-1 equivalent of the subclass B3 zinc-chelating position, failed to rescue C221G function, suggesting the coordination schemes of subclasses B1 and B3 are not interchangeable.


Asunto(s)
Cisteína/metabolismo , Escherichia coli/genética , Glicina/metabolismo , Zinc/química , beta-Lactamasas/metabolismo , beta-Lactamas/metabolismo , Sustitución de Aminoácidos , Dominio Catalítico , Cisteína/química , Cisteína/genética , Escherichia coli/enzimología , Glicina/química , Glicina/genética , Cinética , Ligandos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Mutagénesis , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Agua/química , Zinc/metabolismo , beta-Lactamasas/química , beta-Lactamasas/genética , beta-Lactamas/química
12.
J Virol ; 85(17): 8635-45, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21715503

RESUMEN

Susceptibility to norovirus (NoV), a major pathogen of epidemic gastroenteritis, is associated with histo-blood group antigens (HBGAs), which are also cell attachment factors for this virus. GII.4 NoV strains are predominantly associated with worldwide NoV epidemics with a periodic emergence of new variants. The sequence variations in the surface-exposed P domain of the capsid protein resulting in differential HBGA binding patterns and antigenicity are suggested to drive GII.4 epochal evolution. To understand how temporal sequence variations affect the P domain structure and contribute to epochal evolution, we determined the P domain structure of a 2004 variant with ABH and secretor Lewis HBGAs and compared it with the previously determined structure of a 1996 variant. We show that temporal sequence variations do not affect the binding of monofucosyl ABH HBGAs but that they can modulate the binding strength of difucosyl Lewis HBGAs and thus could contribute to epochal evolution by the potentiated targeting of new variants to Lewis-positive, secretor-positive individuals. The temporal variations also result in significant differences in the electrostatic landscapes, likely reflecting antigenic variations. The proximity of some of these changes to the HBGA binding sites suggests the possibility of a coordinated interplay between antigenicity and HBGA binding in epochal evolution. From the observation that the regions involved in the formation of the HBGA binding sites can be conformationally flexible, we suggest a plausible mechanism for how norovirus disassociates from salivary mucin-linked HBGA before reassociating with HBGAs linked to intestinal epithelial cells during its passage through the gastrointestinal tract.


Asunto(s)
Antígenos de Grupos Sanguíneos/metabolismo , Proteínas de la Cápside/metabolismo , Norovirus/fisiología , Receptores Virales/metabolismo , Acoplamiento Viral , Secuencia de Aminoácidos , Sitios de Unión , Antígenos de Grupos Sanguíneos/química , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Cristalografía por Rayos X , Genotipo , Humanos , Lactante , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Norovirus/química , Norovirus/genética , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Electricidad Estática
13.
Blood ; 115(14): 2852-63, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20154216

RESUMEN

Chronic lymphocytic leukemia (CLL) is the most common leukemia in the Western hemisphere, but its pathogenesis is still poorly understood. Constitutive tyrosine phosphorylation (p) of signal transducer and activator of transcription (STAT) 3 occurs in several solid tumors and hematologic malignancies. In CLL, however, STAT3 is constitutively phosphorylated on serine 727, not tyrosine 705, residues. Because the biologic significance of serine pSTAT3 in CLL is not known, we studied peripheral blood cells of 106 patients with CLL and found that, although tyrosine pSTAT3 was inducible, serine pSTAT3 was constitutive in all patients studied, regardless of blood count, disease stage, or treatment status. In addition, we demonstrated that constitutive serine pSTAT3 translocates to the nucleus by the karyopherin-beta nucleocytoplasmic system and binds DNA. Dephosphorylation of inducible tyrosine pSTAT3 did not affect STAT3-DNA binding, suggesting that constitutive serine pSTAT3 binds DNA. Furthermore, infection of CLL cells with lentiviral STAT3-small hairpin RNA reduced the expression of several STAT3-regulated survival and proliferation genes and induced apoptosis, suggesting that constitutive serine pSTAT3 initiates transcription in CLL cells. Taken together, our data suggest that constitutive phosphorylation of STAT3 on serine 727 residues is a hallmark of CLL and that STAT3 be considered a therapeutic target in this disease.


Asunto(s)
Núcleo Celular/metabolismo , ADN de Neoplasias/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Proteínas de Neoplasias/metabolismo , Factor de Transcripción STAT3/metabolismo , Transcripción Genética , Transporte Activo de Núcleo Celular/genética , Anciano , Anciano de 80 o más Años , Apoptosis/genética , Línea Celular Tumoral , Núcleo Celular/genética , Proliferación Celular , Supervivencia Celular/genética , ADN de Neoplasias/genética , Femenino , Humanos , Lentivirus , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/terapia , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Fosforilación/genética , Factor de Transcripción STAT3/genética , Serina/genética , Serina/metabolismo
14.
J Biol Chem ; 284(48): 33703-12, 2009 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-19812041

RESUMEN

TEM-1 beta-lactamase is the most common plasmid-encoded beta-lactamase in Gram-negative bacteria and is a model class A enzyme. The active site of class A beta-lactamases share several conserved residues including Ser(70), Glu(166), and Asn(170) that coordinate a hydrolytic water involved in deacylation. Unlike Ser(70) and Glu(166), the functional significance of residue Asn(170) is not well understood even though it forms hydrogen bonds with both Glu(166) and the hydrolytic water. The goal of this study was to examine the importance of Asn(170) for catalysis and substrate specificity of beta-lactam antibiotic hydrolysis. The codon for position 170 was randomized to create a library containing all 20 possible amino acids. The random library was introduced into Escherichia coli, and functional clones were selected on agar plates containing ampicillin. DNA sequencing of the functional clones revealed that only asparagine (wild type) and glycine at this position are consistent with wild-type function. The determination of kinetic parameters for several substrates revealed that the N170G mutant is very efficient at hydrolyzing substrates that contain a primary amine in the antibiotic R-group that would be close to the Asn(170) side chain in the acyl-intermediate. In addition, the x-ray structure of the N170G enzyme indicated that the position of an active site water important for deacylation is altered compared with the wild-type enzyme. Taken together, the results suggest the N170G TEM-1 enzyme hydrolyzes ampicillin efficiently because of substrate-assisted catalysis where the primary amine of the ampicillin R-group positions the hydrolytic water and allows for efficient deacylation.


Asunto(s)
Sustitución de Aminoácidos , Mutación , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Secuencia de Aminoácidos , Ampicilina/farmacología , Antibacterianos/farmacología , Asparagina/química , Asparagina/genética , Asparagina/metabolismo , Catálisis , Dominio Catalítico/genética , Cefalexina/química , Cefalexina/metabolismo , Cefalotina/química , Cefalotina/metabolismo , Cristalografía por Rayos X , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Ácido Glutámico/química , Ácido Glutámico/genética , Ácido Glutámico/metabolismo , Cinética , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Especificidad por Sustrato , beta-Lactamasas/química
15.
Curr Opin Struct Biol ; 44: 211-218, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28591681

RESUMEN

Recognition and binding to host glycans present on cellular surfaces is an initial and critical step in viral entry. Diverse families of host glycans such as histo-blood group antigens, sialoglycans and glycosaminoglycans are recognized by viruses. Glycan binding determines virus-host specificity, tissue tropism, pathogenesis and potential for interspecies transmission. Viruses including noroviruses, rotaviruses, enteroviruses, influenza, and papillomaviruses have evolved novel strategies to bind specific glycans often in a strain-specific manner. Structural studies have been instrumental in elucidating the molecular determinants of these virus-glycan interactions, aiding in developing vaccines and antivirals targeting this key interaction. Our review focuses on these key structural aspects of virus-glycan interactions, particularly highlighting the different strain-specific strategies employed by viruses to bind host glycans.


Asunto(s)
Interacciones Huésped-Patógeno , Polisacáridos/metabolismo , Fenómenos Fisiológicos de los Virus , Virus/metabolismo , Animales , Humanos
16.
Clin Vaccine Immunol ; 23(2): 181-3, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26656120

RESUMEN

The performance of an assay to detect antibodies to a norovirus nonstructural fusion protein, designated VPR and consisting of three proteins (GI.1 virus protein genome-linked [VPg], a virus protease, and an RNA-dependent RNA polymerase), was evaluated. The assay sensitivity and specificity were 74.5% and >95%, respectively, for identifying GI.1 norovirus infection among persons who received either a monovalent GI.1 norovirus virus-like particle (VLP) vaccine or placebo by the intranasal route followed by an oral live GI.1 norovirus challenge.


Asunto(s)
Anticuerpos Antivirales/sangre , Infecciones por Caliciviridae/inmunología , Norovirus/inmunología , Proteínas no Estructurales Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Administración Intranasal , Anticuerpos Antivirales/inmunología , Infecciones por Caliciviridae/diagnóstico , Infecciones por Caliciviridae/virología , Heces/virología , Humanos , Inmunoensayo , Norovirus/química , Sensibilidad y Especificidad , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/inmunología , Proteínas Virales de Fusión/inmunología
17.
Curr Opin Virol ; 18: 117-25, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27318434

RESUMEN

Human noroviruses are major causative agents of sporadic and epidemic gastroenteritis both in children and adults. Currently there are no licensed therapeutic intervention measures either in terms of vaccines or drugs available for these highly contagious human pathogens. Genetic and antigenic diversity of these viruses, rapid emergence of new strains, and their ability to infect a broad population by using polymorphic histo-blood group antigens for cell attachment, pose significant challenges for the development of effective antiviral agents. Despite these impediments, there is progress in the design and development of therapeutic agents. These include capsid-based candidate vaccines, and potential antivirals either in the form of glycomimetics or designer antibodies that block HBGA binding, as well as those that target essential non-structural proteins such as the viral protease and RNA-dependent RNA polymerase. In addition to these classical approaches, recent studies suggest the possibility of interferons and targeting host cell factors as viable approaches to counter norovirus infection. This review provides a brief overview of this progress.


Asunto(s)
Infecciones por Caliciviridae/tratamiento farmacológico , Gastroenteritis/tratamiento farmacológico , Norovirus/efectos de los fármacos , Anticuerpos/genética , Anticuerpos/uso terapéutico , Infecciones por Caliciviridae/epidemiología , Infecciones por Caliciviridae/virología , Proteínas de la Cápside/metabolismo , Gastroenteritis/virología , Humanos , Interferones/uso terapéutico , Norovirus/genética , Norovirus/inmunología , Péptido Hidrolasas/inmunología , Polisacáridos/metabolismo , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/metabolismo , Vacunas Virales , Acoplamiento Viral/efectos de los fármacos
18.
Curr Opin Virol ; 7: 119-27, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25073118

RESUMEN

A critical event in the life cycle of a virus is its initial attachment to host cells. This involves recognition by the viruses of specific receptors on the cell surface, including glycans. Viruses typically exhibit strain-dependent variations in recognizing specific glycan receptors, a feature that contributes significantly to cell tropism, host specificity, host adaptation and interspecies transmission. Examples include influenza viruses, noroviruses, rotaviruses, and parvoviruses. Both rotaviruses and noroviruses are well known gastroenteric pathogens that are of significant global health concern. While rotaviruses, in the family Reoviridae, are the major causative agents of life-threatening diarrhea in children, noroviruses, which belong to the Caliciviridae family, cause epidemic and sporadic cases of acute gastroenteritis across all age groups. Both exhibit enormous genotypic and serotypic diversity. Consistent with this diversity each exhibits strain-dependent variations in the types of glycans they recognize for cell attachment. This chapter reviews the current status of the structural biology of such strain-dependent glycan specificities in these two families of viruses.


Asunto(s)
Gastroenteritis/metabolismo , Norovirus/metabolismo , Polisacáridos/química , Polisacáridos/metabolismo , Receptores Virales/química , Receptores Virales/metabolismo , Rotavirus/metabolismo , Animales , Gastroenteritis/virología , Humanos , Norovirus/genética , Rotavirus/genética , Especificidad de la Especie
20.
Biol Chem ; 386(1): 85-93, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15843151

RESUMEN

Calpains are non-lysosomal, Ca 2+ -dependent cysteine proteases, which are ubiquitously distributed across cell types and vertebrate species. The rules that govern calpain specificity have not yet been determined. To elucidate the cleavage pattern of calpains, we carried out calpain-induced proteolytic studies on the insulin-like growth factor binding proteins IGFBP-4 and -5. Proteolysis of IGFBPs is well characterized in numerous reports. Our results show that calpain cleavage sites are in the non-conserved unstructured regions of the IGFBPs. Compilation of the calpain-induced proteolytic cleavage sites in several proteins reported in the literature, together with our present study, has not revealed clear preferences for amino acid sequences. We therefore conclude that calpains seem not to recognize amino acid sequences, but instead cleave with low sequence specificity at unstructured or solvent-exposed fragments that connect folded, stable domains of target proteins.


Asunto(s)
Calpaína/química , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/química , Secuencia de Aminoácidos , Clonación Molecular , Regulación de la Expresión Génica , Humanos , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/química , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/química , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Datos de Secuencia Molecular , Pliegue de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Especificidad por Sustrato/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA