Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
NPJ Vaccines ; 6(1): 7, 2021 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-33420102

RESUMEN

HCV vaccine development is stymied by the high genetic diversity of the virus and the variability of the envelope glycoproteins. One strategy to overcome this is to identify conserved, functionally important regions-such as the epitopes of broadly neutralizing antibodies (bNAbs)-and use these as a basis for structure-based vaccine design. Here, we report an anti-idiotype approach that has generated an antibody that mimics a highly conserved neutralizing epitope on HCV E2. Crucially, a mutagenesis screen was used to identify the antibody, designated B2.1 A, whose binding characteristics to the bNAb AP33 closely resemble those of the original antigen. Protein crystallography confirmed that B2.1 A is a structural mimic of the AP33 epitope. When used as an immunogen B2.1 A induced antibodies that recognized the same epitope and E2 residues as AP33 and most importantly protected against HCV challenge in a mouse model.

3.
Biochemistry ; 57(26): 3797-3806, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29812914

RESUMEN

The thermoacidophilic archaea Picrophilus torridus and Sulfolobus solfataricus catabolize glucose via a nonphosphorylative Entner-Doudoroff pathway and a branched Entner-Doudoroff pathway, respectively. Key enzymes for these Entner-Doudoroff pathways are the aldolases, 2-keto-3-deoxygluconate aldolase (KDG-aldolase) and 2-keto-3-deoxy-6-phosphogluconate aldolase [KD(P)G-aldolase]. KDG-aldolase from P. torridus (Pt-KDG-aldolase) is highly specific for the nonphosphorylated substrate, 2-keto-3-deoxygluconate (KDG), whereas KD(P)G-aldolase from S. solfataricus [Ss-KD(P)G-aldolase] is an enzyme that catalyzes the cleavage of both KDG and 2-keto-3-deoxy-6-phosphogluconate (KDPG), with a preference for KDPG. The structural basis for the high specificity of Pt-KDG-aldolase for KDG as compared to the more promiscuous Ss-KD(P)G-aldolase has not been analyzed before. In this work, we report the elucidation of the structure of Ss-KD(P)G-aldolase in complex with KDPG at 2.35 Å and that of KDG-aldolase from P. torridus at 2.50 Å resolution. By superimposition of the active sites of the two enzymes, and subsequent site-directed mutagenesis studies, a network of four amino acids, namely, Arg106, Tyr132, Arg237, and Ser241, was identified in Ss-KD(P)G-aldolase that interact with the negatively charged phosphate group of KDPG, thereby increasing the affinity of the enzyme for KDPG. This KDPG-binding network is absent in Pt-KDG-aldolase, which explains the low catalytic efficiency of KDPG cleavage.


Asunto(s)
Aldehído-Liasas/química , Proteínas Arqueales/química , Gluconatos/química , Sulfolobus solfataricus/enzimología , Thermoplasmales/enzimología , Modelos Moleculares , Dominios Proteicos , Relación Estructura-Actividad
4.
Oncotarget ; 7(50): 83017-83030, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27806324

RESUMEN

PRIMA-1Met is the methylated PRIMA-1 (p53 reactivation and induction of massive apoptosis) and could restore tumor suppressor function of mutant p53 and induce p53 dependent apoptosis in cancer cells harboring mutant p53. However, p53 independent activity of PRIMA-1Met remains elusive. Here we reported that PRIMA-1Met attenuated colorectal cancer cell growth irrespective of p53 status. Kinase profiling revealed that mitogen-activated or extracellular signal-related protein kinase (MEK) might be a potential target of PRIMA-1Met. Pull-down binding and ATP competitive assay showed that PRIMA-1Met directly bound MEK in vitro and in cells. Furthermore, the direct binding sites of PRIMA-1Met were explored by using a computational docking model. Treatment of colorectal cancer cells with PRIMA-1Met inhibited p53-independent phosphorylation of MEK, which in turn impaired anchorage-independent cell growth in vitro. Moreover, PRIMA-1Met suppressed colorectal cancer growth in xenograft mouse model by inhibiting MEK1 activity.Taken together, our findings demonstrate a novel p53-independent activity of PRIMA-1Met to inhibit MEK and suppress colorectal cancer growth.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Quinuclidinas/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antineoplásicos/metabolismo , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Relación Dosis-Respuesta a Droga , Células HCT116 , Células HT29 , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones Desnudos , Simulación del Acoplamiento Molecular , Fosforilación , Unión Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Quinuclidinas/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Biol Chem ; 290(46): 27736-48, 2015 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-26370075

RESUMEN

Streptococcus pneumoniae is an important human pathogen that causes a range of disease states. Sialidases are important bacterial virulence factors. There are three pneumococcal sialidases: NanA, NanB, and NanC. NanC is an unusual sialidase in that its primary reaction product is 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (Neu5Ac2en, also known as DANA), a nonspecific hydrolytic sialidase inhibitor. The production of Neu5Ac2en from α2-3-linked sialosides by the catalytic domain is confirmed within a crystal structure. A covalent complex with 3-fluoro-ß-N-acetylneuraminic acid is also presented, suggesting a common mechanism with other sialidases up to the final step of product formation. A conformation change in an active site hydrophobic loop on ligand binding constricts the entrance to the active site. In addition, the distance between the catalytic acid/base (Asp-315) and the ligand anomeric carbon is unusually short. These features facilitate a novel sialidase reaction in which the final step of product formation is direct abstraction of the C3 proton by the active site aspartic acid, forming Neu5Ac2en. NanC also possesses a carbohydrate-binding module, which is shown to bind α2-3- and α2-6-linked sialosides, as well as N-acetylneuraminic acid, which is captured in the crystal structure following hydration of Neu5Ac2en by NanC. Overall, the pneumococcal sialidases show remarkable mechanistic diversity while maintaining a common structural scaffold.


Asunto(s)
Proteínas Bacterianas/química , Inhibidores Enzimáticos/síntesis química , Neuraminidasa/química , Streptococcus pneumoniae/enzimología , Azúcares Ácidos/síntesis química , Proteínas Bacterianas/ultraestructura , Dominio Catalítico , Cristalografía por Rayos X , Neuraminidasa/ultraestructura
7.
BMC Struct Biol ; 15: 15, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26289431

RESUMEN

BACKGROUND: Streptococcus pneumoniae Neuraminidase A (NanA) is a multi-domain protein anchored to the bacterial surface. Upstream of the catalytic domain of NanA is a domain that conforms to the sialic acid-recognising CBM40 family of the CAZY (carbohydrate-active enzymes) database. This domain has been identified to play a critical role in allowing the bacterium to promote adhesion and invasion of human brain microvascular endothelial cells, and hence may play a key role in promoting bacterial meningitis. In addition, the CBM40 domain has also been reported to activate host chemokines and neutrophil recruitment during infection. RESULTS: Crystal structures of both apo- and holo- forms of the NanA CBM40 domain (residues 121 to 305), have been determined to 1.8 Å resolution. The domain shares the fold of other CBM40 domains that are associated with sialidases. When in complex with α2,3- or α2,6-sialyllactose, the domain is shown to interact only with the terminal sialic acid. Significantly, a deep acidic pocket adjacent to the sialic acid-binding site is identified, which is occupied by a lysine from a symmetry-related molecule in the crystal. This pocket is adjacent to a region that is predicted to be involved in protein-protein interactions. CONCLUSIONS: The structural data provide the details of linkage-independent sialyllactose binding by NanA CBM40 and reveal striking surface features that may hold the key to recognition of binding partners on the host cell surface. The structure also suggests that small molecules or sialic acid analogues could be developed to fill the acidic pocket and hence provide a new therapeutic avenue against meningitis caused by S. pneumoniae.


Asunto(s)
Proteínas Bacterianas/química , Neuraminidasa/química , Streptococcus pneumoniae/enzimología , Factores de Virulencia/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Lactosa/análogos & derivados , Lactosa/metabolismo , Modelos Moleculares , Neuraminidasa/metabolismo , Unión Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , Ácidos Siálicos/metabolismo , Streptococcus pneumoniae/química , Factores de Virulencia/metabolismo
8.
Nat Commun ; 6: 7624, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26154892

RESUMEN

The gastrointestinal mucus layer is colonized by a dense community of microbes catabolizing dietary and host carbohydrates during their expansion in the gut. Alterations in mucosal carbohydrate availability impact on the composition of microbial species. Ruminococcus gnavus is a commensal anaerobe present in the gastrointestinal tract of >90% of humans and overrepresented in inflammatory bowel diseases (IBD). Using a combination of genomics, enzymology and crystallography, we show that the mucin-degrader R. gnavus ATCC 29149 strain produces an intramolecular trans-sialidase (IT-sialidase) that cleaves off terminal α2-3-linked sialic acid from glycoproteins, releasing 2,7-anhydro-Neu5Ac instead of sialic acid. Evidence of IT-sialidases in human metagenomes indicates that this enzyme occurs in healthy subjects but is more prevalent in IBD metagenomes. Our results uncover a previously unrecognized enzymatic activity in the gut microbiota, which may contribute to the adaptation of intestinal bacteria to the mucosal environment in health and disease.


Asunto(s)
Adaptación Fisiológica/fisiología , Regulación Bacteriana de la Expresión Génica/fisiología , Glicoproteínas/metabolismo , Mucosa Intestinal/microbiología , Neuraminidasa/metabolismo , Ruminococcus/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Glicoproteínas/genética , Humanos , Mucinas/metabolismo , Neuraminidasa/genética , Ruminococcus/genética , Ruminococcus/metabolismo
9.
Antimicrob Agents Chemother ; 59(3): 1495-504, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25534734

RESUMEN

Compounds that target the cellular factors essential for influenza virus replication represent an innovative approach to antiviral therapy. Sp2CBMTD is a genetically engineered multivalent protein that masks sialic acid-containing cellular receptors on the respiratory epithelium, which are recognized by influenza viruses. Here, we evaluated the antiviral potential of Sp2CBMTD against lethal infection in mice with an emerging A/Anhui/1/2013 (H7N9) influenza virus and addressed the mechanistic basis of its activity in vivo. Sp2CBMTD was administered to mice intranasally as a single or repeated dose (0.1, 1, 10, or 100 µg) before (day -7, -3, and/or -1) or after (6 or 24 h) H7N9 virus inoculation. A single Sp2CBMTD dose (10 or 100 µg) protected 80% to 100% of the mice when administered 7 days before the H7N9 lethal challenge. Repeated Sp2CBMTD administration conferred the highest protection, resulting in 100% survival of the mice even at the lowest dose tested (0.1 µg). When treatment began 24 h after exposure to the H7N9 virus, a single administration of 100 µg of Sp2CBMTD protected 40% of the mice from death. The administration of Sp2CBMTD induced the pulmonary expression of proinflammatory mediators (interleukin-6 [IL-6], IL-1ß, RANTES, monocyte chemotactic protein-1 [MCP-1], macrophage inflammatory protein-1α [MIP-1α], and inducible protein [IP-10]) and recruited neutrophils to the respiratory tract before H7N9 virus infection, which resulted in less pronounced inflammation and rapid virus clearance from mouse lungs. Sp2CBMTD administration did not affect the virus-specific adaptive immune response, which was sufficient to protect against reinfection with a higher dose of homologous H7N9 virus or heterologous H5N1 virus. Thus, Sp2CBMTD was effective in preventing H7N9 infections in a lethal mouse model and holds promise as a prophylaxis option against zoonotic influenza viruses.


Asunto(s)
Antivirales/uso terapéutico , Proteínas Portadoras/uso terapéutico , Subtipo H7N9 del Virus de la Influenza A , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Receptores de Superficie Celular/fisiología , Ácidos Siálicos/metabolismo , Animales , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Femenino , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/inmunología , Replicación Viral
10.
Proc Natl Acad Sci U S A ; 111(17): 6401-6, 2014 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-24733924

RESUMEN

There is a need for new approaches for the control of influenza given the burden caused by annual seasonal outbreaks, the emergence of viruses with pandemic potential, and the development of resistance to current antiviral drugs. We show that multivalent biologics, engineered using carbohydrate-binding modules specific for sialic acid, mask the cell-surface receptor recognized by the influenza virus and protect mice from a lethal challenge with 2009 pandemic H1N1 influenza virus. The most promising biologic protects mice when given as a single 1-µg intranasal dose 7 d in advance of viral challenge. There also is sufficient virus replication to establish an immune response, potentially protecting the animal from future exposure to the virus. Furthermore, the biologics appear to stimulate inflammatory mediators, and this stimulation may contribute to their protective ability. Our results suggest that this host-targeted approach could provide a front-line prophylactic that has the potential to protect against any current and future influenza virus and possibly against other respiratory pathogens that use sialic acid as a receptor.


Asunto(s)
Gripe Humana/metabolismo , Gripe Humana/prevención & control , Ingeniería de Proteínas , Receptores Virales/metabolismo , Animales , Peso Corporal , Quimiocinas/metabolismo , Perros , Humanos , Mediadores de Inflamación/metabolismo , Subtipo H1N1 del Virus de la Influenza A/fisiología , Pulmón/patología , Pulmón/virología , Células de Riñón Canino Madin Darby , Ratones , Ácido N-Acetilneuramínico/metabolismo , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Receptores de Superficie Celular/metabolismo , Análisis de Supervivencia
11.
Transplantation ; 97(10): 1043-8, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24503760

RESUMEN

BACKGROUND: The strategy of evaluating every donation opportunity warrants an investigation into the financial feasibility of this practice. The purpose of this investigation is to measure resource utilization required for procurement of transplantable organs in an organ procurement organization (OPO). METHODS: Donors were stratified into those that met OPTN-defined eligible death criteria (ED donors, n=589) and those that did not (NED donors, n=703). Variable direct costs and time utilization by OPO staff for organ procurement were measured and amortized per organ transplanted using permutation methods and statistical bootstrapping/resampling approaches. RESULTS: More organs per donor were procured (3.66±1.2 vs. 2.34±0.8, P<0.0001) and transplanted (3.51±1.2 vs. 2.08±0.8, P<0.0001) in ED donors compared with NED donors. The variable direct costs were significantly lower in the NED donors ($29,879.4±11590.1 vs. $19,019.6±7599.60, P<0.0001). In contrast, the amortized variable direct costs per organ transplanted were significantly higher in the NED donors ($8,414.5±138.29 vs. $9,272.04±344.56, P<0.0001). The ED donors where thoracic organ procurement occurred were 67% more expensive than in abdominal-only organ procurement. The total time allocated per donor was significantly shorter in the NED donors (91.2±44.9 hr vs. 86.8±78.6 hr, P=0.01). In contrast, the amortized time per organ transplanted was significantly longer in the NED donors (23.1±0.8 hr vs. 36.9±3.2 hr, P<0.001). DISCUSSION: The variable direct costs and time allocated per organ transplanted is significantly higher in donors that do not meet the eligible death criteria.


Asunto(s)
Muerte Encefálica , Trasplante de Órganos/estadística & datos numéricos , Donantes de Tejidos/provisión & distribución , Obtención de Tejidos y Órganos/organización & administración , Anciano , Análisis Costo-Beneficio , Toma de Decisiones , Estudios de Factibilidad , Femenino , Humanos , Masculino , Trasplante de Órganos/economía , Estados Unidos
12.
Biochemistry ; 52(51): 9177-86, 2013 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-24295366

RESUMEN

Aspergillus fumigatus is an airborne fungal pathogen. We previously cloned and characterized an exo-sialidase from A. fumigatus and showed that it preferred 2-keto-3-deoxynononic acid (KDN) as a substrate to N-acetylneuraminic acid (Neu5Ac). The purpose of this study was to investigate the structure-function relationships of critical catalytic site residues. Site-directed mutagenesis was used to create three mutant recombinant enzymes: the catalytic nucleophile (Y358H), the general acid/base catalyst (D84A), and an enlargement of the binding pocket to attempt to accommodate the N-acetyl group of Neu5Ac (R171L). Crystal structures for all enzymes were determined. The D84A mutation had an effect in decreasing the activity of AfKDNase that was stronger than that of the same mutation in the structurally similar sialidase from the bacterium Micromonospora viridifaciens. These data suggest that the catalytic acid is more important in the reaction of AfKDNase and that catalysis is less dependent on nucleophilic or electrostatic stabilization of the developing positive charge at the transition state for hydrolysis. Removal of the catalytic nucleophile (Y358H) significantly lowered the activity of the enzyme, but this mutant remained a retaining glycosidase as demonstrated by nuclear magnetic resonance spectroscopic analysis. This is a novel finding that has not been shown with other sialidases. Kinetic activity measured at pH 5.2 revealed that R171L had higher activity on a Neu5Ac-based substrate than wild-type KDNase; hence, leucine in place of arginine in the binding pocket improved catalysis toward Neu5Ac substrates. Hence, whether a sialidase is primarily a KDNase or a neuraminidase is due in part to the presence of an amino acid that creates a steric clash with the N-acetyl group.


Asunto(s)
Aspergillus fumigatus/enzimología , Proteínas Fúngicas/metabolismo , Glicósido Hidrolasas/metabolismo , Micromonospora/enzimología , Modelos Moleculares , Proteínas Mutantes/metabolismo , Neuraminidasa/metabolismo , Sustitución de Aminoácidos , Biocatálisis , Dominio Catalítico , Cristalografía por Rayos X , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Glicósido Hidrolasas/química , Glicósido Hidrolasas/genética , Concentración de Iones de Hidrógeno , Hidrólisis , Cetoácidos/química , Cetoácidos/metabolismo , Cinética , Conformación Molecular , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/metabolismo , Neuraminidasa/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
13.
Prog Transplant ; 23(3): 290-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23996950

RESUMEN

CONTEXT: The Organ Donor Breakthrough Collaborative recommended high-leverage changes including "master effective requesting. OBJECTIVE: To measure who approaches decedent families to request organ donation and to determine whether trained specialists will solicit authorization at equal frequency regardless of donor characteristics. METHODS: Retrospective analysis of data from 2006 to 2009 in an organ center's donor database. Decedents were stratified into 2 groups: those that met the Organ Procurement and Transplantation Network's eligible death criteria (ED donors) and those that did not (not eligible death [NED] donors). RESULTS: Of decedents whose families were approached for authorization, 46% were ED donors and 54% were NED donors. Trained specialists solicited authorization from 76% of the total population but were more likely to solicit authorization from ED donors than NED donors (86% vs 68%, P<.001). Trained specialists were more likely to solicit authorization from donors whose cause of death was overrepresented in ED donors and donors less than 50 years old. Trained specialists were more likely than others to obtain authorization from families of all donors. Multivariable modeling demonstrated that having a trained specialist approach the decedent's family was associated with the highest odds of obtaining authorization. CONCLUSIONS: Trained specialists approached most families of decedents for authorization, but disproportionately approached fewer families of NED donors. Having a trained specialist approach the decedent family has the strongest impact on obtaining donor authorization. These data suggest that fewer resources are allocated to NED donors, which may adversely affect the supply of deceased donor organs.


Asunto(s)
Familia/psicología , Relaciones Profesional-Familia , Especialización , Obtención de Tejidos y Órganos , Adulto , Alabama , Causas de Muerte , Distribución de Chi-Cuadrado , Toma de Decisiones , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
14.
BMC Biochem ; 13: 19, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-23031230

RESUMEN

BACKGROUNDS: Streptococcus pneumoniae expresses three distinct sialidases, NanA, NanB, and NanC, that are believed to be key virulence factors and thus, potential important drug targets. We previously reported that the three enzymes release different products from sialosides, but could share a common catalytic mechanism before the final step of product formation. However, the kinetic investigations of the three sialidases have not been systematically done thus far, due to the lack of an easy and steady measurement of sialidase reaction rate. RESULTS: In this work, we present further kinetic characterization of pneumococcal sialidases by using a direct spectrophotometric method with the chromogenic substrate p-nitrophenyl-N-acetylneuraminic acid (p-NP-Neu5Ac). Using our assay, the measured kinetic parameters of the three purified pneumococcal sialidase, NanA, NanB and NanC, were obtained and were in perfect agreement with the previously published data. The major advantage of this alternative method resides in the direct measurement of the released product, allowing to readily determine of initial reaction rates and record complete hydrolysis time courses. CONCLUSION: We developed an accurate, fast and sensitive spectrophotometric method to investigate the kinetics of sialidase-catalyzed reactions. This fast, sensitive, inexpensive and accurate method could benefit the study of the kinetics and inhibition of sialidases in general.


Asunto(s)
Neuraminidasa/antagonistas & inhibidores , Biocatálisis , Tampones (Química) , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Ácido N-Acetilneuramínico/metabolismo , Neuraminidasa/genética , Neuraminidasa/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrofotometría Ultravioleta , Streptococcus pneumoniae/enzimología , Especificidad por Sustrato
15.
Chembiochem ; 13(16): 2374-83, 2012 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-23070966

RESUMEN

The major human pathogen Streptococcus pneumoniae plays a key role in several disease states including septicaemia, meningitis and community-acquired pneumonia. Although vaccines against S. pneumoniae are available as prophylactics, there remains a need to identify and characterise novel chemical entities that can treat the diseases caused by this pathogen. S. pneumoniae expresses three sialidases, enzymes that cleave sialic acid from carbohydrate-based surface molecules. Two of these enzymes, NanA and NanB, have been implicated in the pathogenesis of S. pneumoniae and are considered to be validated drug targets. Here we report our studies on the synthesis and structural characterisation of novel NanB-selective inhibitors that are inspired by the ß-amino-sulfonic acid family of buffers.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Himecromona/análogos & derivados , Neuraminidasa/antagonistas & inhibidores , Streptococcus pneumoniae/enzimología , Ácidos Sulfónicos/farmacología , Cristalografía por Rayos X , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Himecromona/síntesis química , Himecromona/química , Himecromona/farmacología , Modelos Moleculares , Estructura Molecular , Neuraminidasa/química , Neuraminidasa/metabolismo , Relación Estructura-Actividad , Ácidos Sulfónicos/síntesis química , Ácidos Sulfónicos/química
16.
J Virol ; 86(23): 12923-32, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22993159

RESUMEN

The E2 envelope glycoprotein of hepatitis C virus (HCV) binds to the host entry factor CD81 and is the principal target for neutralizing antibodies (NAbs). Most NAbs recognize hypervariable region 1 on E2, which undergoes frequent mutation, thereby allowing the virus to evade neutralization. Consequently, there is great interest in NAbs that target conserved epitopes. One such NAb is AP33, a mouse monoclonal antibody that recognizes a conserved, linear epitope on E2 and potently neutralizes a broad range of HCV genotypes. In this study, the X-ray structure of AP33 Fab in complex with an epitope peptide spanning residues 412 to 423 of HCV E2 was determined to 1.8 Å. In the complex, the peptide adopts a ß-hairpin conformation and docks into a deep binding pocket on the antibody. The major determinants of antibody recognition are E2 residues L413, N415, G418, and W420. The structure is compared to the recently described HCV1 Fab in complex with the same epitope. Interestingly, the antigen-binding sites of HCV1 and AP33 are completely different, whereas the peptide conformation is very similar in the two structures. Mutagenesis of the peptide-binding residues on AP33 confirmed that these residues are also critical for AP33 recognition of whole E2, confirming that the peptide-bound structure truly represents AP33 interaction with the intact glycoprotein. The slightly conformation-sensitive character of the AP33-E2 interaction was explored by cross-competition analysis and alanine-scanning mutagenesis. The structural details of this neutralizing epitope provide a starting point for the design of an immunogen capable of eliciting AP33-like antibodies.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Hepatitis C/prevención & control , Modelos Moleculares , Tetraspanina 28/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas contra Hepatitis Viral/inmunología , Proteínas Virales/inmunología , Animales , Anticuerpos Neutralizantes/química , Cristalografía por Rayos X , Epítopos/genética , Ratones , Mutagénesis , Tetraspanina 28/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/metabolismo
17.
Glycobiology ; 22(2): 174-80, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21846691

RESUMEN

The hemagglutinin-neuraminidase (HN) glycoprotein is utilized by human parainfluenza viruses for binding to the host cell. By the use of glycan array assays, we demonstrate that, in addition to the first catalytic-binding site, the HN of human parainfluenza virus type 1 has a second site for binding covered by N-linked glycan. Our data suggest that attachment of the first site to sialic acid (SA)-linked receptors triggers exposure of the second site. We found that both sites bind to α2-3-linked SAs with a preference for a sialyl-Lewis(x) motif. Binding to α2-3-linked SAs with a sulfated sialyl-Lewis motif as well as to α2-8-linked SAs was unique for the second binding site. Neither site recognizes α2-6-linked oligosaccharides.


Asunto(s)
Inhibidores Enzimáticos/metabolismo , Proteína HN/química , Proteína HN/metabolismo , Mutación/genética , Neuraminidasa/química , Virus de la Parainfluenza 1 Humana/metabolismo , Receptores de Superficie Celular/metabolismo , Azidas/química , Azidas/metabolismo , Sitios de Unión , Inhibidores Enzimáticos/química , Proteína HN/genética , Ácidos Hexurónicos/química , Ácidos Hexurónicos/metabolismo , Humanos , Ácido N-Acetilneuramínico/análogos & derivados , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/metabolismo , Neuraminidasa/antagonistas & inhibidores , Oligosacáridos/química , Oligosacáridos/metabolismo , Virus de la Parainfluenza 1 Humana/genética , Unión Proteica , Receptores de Superficie Celular/química , Antígeno Sialil Lewis X
18.
J Mol Biol ; 409(4): 496-503, 2011 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-21514303

RESUMEN

The human pathogen Streptococcus pneumoniae is the major cause of bacterial meningitis, respiratory tract infection, septicemia, and otitis media. The bacterium expresses neuraminidase (NA) proteins that contribute to pathogenesis by cleaving sialic acids from host glycoconjugates, thereby enhancing biofilm formation and colonization. Recent in vivo experiments have shown that antiviral compounds, widely used in clinics and designed to inhibit influenza NA, significantly reduce biofilm formation and nasopharyngeal colonization of S. pneumoniae in mice. Here, we present the structural basis for the beneficial effect of these compounds against pneumococcal infection. Crystal structures of pneumococcal NanA in complex with zanamivir and oseltamivir carboxylate are discussed, correlated with measured inhibitory constants K(i), and compared with the binding modes of the inhibitors in the viral enzyme. Inhibitor structures show for the first time how clinically approved anti-influenza compounds interact with an NA of the human pathogen S. pneumoniae and give a rational explanation for their antibacterial effects.


Asunto(s)
Antivirales/farmacología , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/química , Oseltamivir/análogos & derivados , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/enzimología , Zanamivir/farmacología , Animales , Ácidos Carboxílicos/química , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Datos de Secuencia Molecular , Estructura Molecular , Neuraminidasa/genética , Neuraminidasa/metabolismo , Oseltamivir/farmacología , Conformación Proteica
19.
J Biol Chem ; 286(12): 10783-92, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21247893

RESUMEN

Aspergillus fumigatus is a filamentous fungus that can cause severe respiratory disease in immunocompromised individuals. A putative sialidase from A. fumigatus was recently cloned and shown to be relatively poor in cleaving N-acetylneuraminic acid (Neu5Ac) in comparison with bacterial sialidases. Here we present the first crystal structure of a fungal sialidase. When the apo structure was compared with bacterial sialidase structures, the active site of the Aspergillus enzyme suggested that Neu5Ac would be a poor substrate because of a smaller pocket that normally accommodates the acetamido group of Neu5Ac in sialidases. A sialic acid with a hydroxyl in place of an acetamido group is 2-keto-3-deoxynononic acid (KDN). We show that KDN is the preferred substrate for the A. fumigatus sialidase and that A. fumigatus can utilize KDN as a sole carbon source. A 1.45-Å resolution crystal structure of the enzyme in complex with KDN reveals KDN in the active site in a boat conformation and nearby a second binding site occupied by KDN in a chair conformation, suggesting that polyKDN may be a natural substrate. The enzyme is not inhibited by the sialidase transition state analog 2-deoxy-2,3-dehydro-N-acetylneuraminic acid (Neu5Ac2en) but is inhibited by the related 2,3-didehydro-2,3-dideoxy-D-glycero-D-galacto-nonulosonic acid that we show bound to the enzyme in a 1.84-Å resolution crystal structure. Using a fluorinated KDN substrate, we present a 1.5-Å resolution structure of a covalently bound catalytic intermediate. The A. fumigatus sialidase is therefore a KDNase with a similar catalytic mechanism to Neu5Ac exosialidases, and this study represents the first structure of a KDNase.


Asunto(s)
Aspergillus fumigatus/enzimología , Proteínas Fúngicas/química , Glicósido Hidrolasas/química , Neuraminidasa/química , Sitios de Unión , Cristalografía por Rayos X , Proteínas Fúngicas/metabolismo , Glicósido Hidrolasas/metabolismo , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/metabolismo , Neuraminidasa/metabolismo , Estructura Terciaria de Proteína , Relación Estructura-Actividad
20.
J Am Chem Soc ; 133(6): 1718-21, 2011 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-21244006

RESUMEN

Streptococcus penumoniae is a major human pathogen responsible for respiratory tract infections, septicemia, and meningitis and continues to produce numerous cases of disease with relatively high mortalities. S. pneumoniae encodes up to three sialidases, NanA, NanB, and NanC, that have been implicated in pathogenesis and are potential drug targets. NanA has been shown to be a promiscuous sialidase, hydrolyzing the removal of Neu5Ac from a variety of glycoconjugates with retention of configuration at the anomeric center, as we confirm by NMR. NanB is an intramolecular trans-sialidase producing 2,7-anhydro-Neu5Ac selectively from α2,3-sialosides. Here, we show that the first product of NanC is 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (Neu5Ac2en) that can be slowly hydrated by the enzyme to Neu5Ac. We propose that the three pneumococcal sialidases share a common catalytic mechanism up to the final product formation step, and speculate on the roles of the enzymes in the lifecycle of the bacterium.


Asunto(s)
Neuraminidasa/metabolismo , Streptococcus pneumoniae/enzimología , Biocatálisis , Dominio Catalítico , Cinética , Modelos Moleculares , Neuraminidasa/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA