Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Proc Natl Acad Sci U S A ; 115(1): 168-173, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29255041

RESUMEN

Circulating influenza viruses evade neutralization in their human hosts by acquiring escape mutations at epitopes of prevalent antibodies. A goal for next-generation influenza vaccines is to reduce escape likelihood by selectively eliciting antibodies recognizing conserved surfaces on the viral hemagglutinin (HA). The receptor-binding site (RBS) on the HA "head" and a region near the fusion peptide on the HA "stem" are two such sites. We describe here a human antibody clonal lineage, designated CL6649, members of which bind a third conserved site ("lateral patch") on the side of the H1-subtype, HA head. A crystal structure of HA with bound Fab6649 shows the conserved antibody footprint. The site was invariant in isolates from 1977 (seasonal) to 2012 (pdm2009); antibodies in CL6649 recognize HAs from the entire period. In 2013, human H1 viruses acquired mutations in this epitope that were retained in subsequent seasons, prompting modification of the H1 vaccine component in 2017. The mutations inhibit Fab6649 binding. We infer from the rapid spread of these mutations in circulating H1 influenza viruses that the previously subdominant, conserved lateral patch had become immunodominant for individuals with B-cell memory imprinted by earlier H1 exposure. We suggest that introduction of the pdm2009 H1 virus, to which most of the broadly prevalent, neutralizing antibodies did not bind, conferred a selective advantage in the immune systems of infected hosts to recall of memory B cells that recognized the lateral patch, the principal exposed epitope that did not change when pdm2009 displaced previous seasonal H1 viruses.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Epítopos/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Memoria Inmunológica , Vacunas contra la Influenza/inmunología , Epítopos/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética
2.
Anal Chem ; 92(17): 11879-11887, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32867501

RESUMEN

Avian influenza viruses, such as A(H5N1) and A(H7N9), are primary public health concerns due to their pandemic potential. Influenza vaccines represent the most effective response to this threat especially with timely provision. The current pandemic response timelines require a substantial period for strain-specific reference antigen and sera preparation for use with single-radial immunodiffusion (SRID), the accepted vaccine potency assay. To address this time lag, the isotope dilution mass spectrometry (IDMS) method was developed to quantify the absolute hemagglutinin (HA, the main influenza antigen) amount in the vaccine without the need for purified, inactivated, and calibrated virus reference antigens. However, an additional challenge in determining potency is to differentiate between vaccine antigens in their most potent form from other less potent, stressed antigen forms. The limited trypsin digestion (LTD) method has been developed and does not require strain-specific full-length reference antigens or antibodies; instead, stressed HA is selectively degraded, leaving the more potent form to be measured. LTD, followed by precipitation and IDMS, allows for efficient differentiation between potent and significantly less potent HA for vaccine release and potency testing across the vaccine's shelf life. In this study, we tested the LTD-IDMS assay on A(H5N1) vaccine material that had been stressed by low pH, heat, and multiple freeze-thaw cycles. The results showed that the LTD-IDMS method effectively quantified the potent HA in A(H5N1) vaccine material with results comparable to SRID. As such, it shows great promise to complement and potentially replace SRID in a pandemic when strain-specific reagents may not be readily available.


Asunto(s)
Hemaglutininas/metabolismo , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/inmunología , Espectrometría de Masas/métodos , Humanos
3.
Proc Natl Acad Sci U S A ; 112(6): 1767-72, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25624487

RESUMEN

Human cytomegalovirus (HCMV) is a major cause of morbidity and mortality in transplant patients and the leading viral cause of birth defects after congenital infection. The glycoprotein complexes gH/gL/gO and gH/gL/UL128/UL130/UL131A (Pentamer) are key targets of the human humoral response against HCMV and are required for HCMV entry into fibroblasts and endothelial/epithelial cells, respectively. We expressed and characterized soluble forms of gH/gL, gH/gL/gO, and Pentamer. Mass spectrometry and mutagenesis analysis revealed that gL-Cys144 forms disulfide bonds with gO-Cys351 in gH/gL/gO and with UL128-Cys162 in the Pentamer. Notably, Pentamer harboring the UL128-Cys162Ser/gL-Cys144Ser mutations had impaired syncytia formation and reduced interference of HCMV entry into epithelial cells. Electron microscopy analysis showed that HCMV gH/gL resembles HSV gH/gL and that gO and UL128/UL130/UL131A bind to the same site at the gH/gL N terminus. These data are consistent with gH/gL/gO and Pentamer forming mutually exclusive cell entry complexes and reveal the overall location of gH/gL-, gH/gL/gO-, and Pentamer-specific neutralizing antibody binding sites. Our results provide, to our knowledge, the first structural view of gH/gL/gO and Pentamer supporting the development of vaccines and antibody therapeutics against HCMV.


Asunto(s)
Citomegalovirus/inmunología , Citomegalovirus/fisiología , Glicoproteínas de Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Anticuerpos Neutralizantes/inmunología , Sitios de Unión/genética , Western Blotting , Cromatografía de Afinidad , Secuencia Conservada/genética , Citomegalovirus/metabolismo , Disulfuros/metabolismo , Citometría de Flujo , Humanos , Procesamiento de Imagen Asistido por Computador , Espectrometría de Masas , Glicoproteínas de Membrana/química , Microscopía Electrónica , Complejos Multiproteicos/química , Mutagénesis , Mutagénesis Sitio-Dirigida , Mutación/genética , Unión Proteica , Proteínas del Envoltorio Viral/química
4.
PLoS Pathog ; 11(10): e1005230, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26485028

RESUMEN

Human Cytomegalovirus (HCMV) is a major cause of morbidity and mortality in transplant patients and in fetuses following congenital infection. The glycoprotein complexes gH/gL/gO and gH/gL/UL128/UL130/UL131A (Pentamer) are required for HCMV entry in fibroblasts and endothelial/epithelial cells, respectively, and are targeted by potently neutralizing antibodies in the infected host. Using purified soluble forms of gH/gL/gO and Pentamer as well as a panel of naturally elicited human monoclonal antibodies, we determined the location of key neutralizing epitopes on the gH/gL/gO and Pentamer surfaces. Mass Spectrometry (MS) coupled to Chemical Crosslinking or to Hydrogen Deuterium Exchange was used to define residues that are either in proximity or part of neutralizing epitopes on the glycoprotein complexes. We also determined the molecular architecture of the gH/gL/gO- and Pentamer-antibody complexes by Electron Microscopy (EM) and 3D reconstructions. The EM analysis revealed that the Pentamer specific neutralizing antibodies bind to two opposite surfaces of the complex, suggesting that they may neutralize infection by different mechanisms. Together, our data identify the location of neutralizing antibodies binding sites on the gH/gL/gO and Pentamer complexes and provide a framework for the development of antibodies and vaccines against HCMV.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Antígenos Virales/inmunología , Citomegalovirus/inmunología , Epítopos de Linfocito B/inmunología , Proteínas Virales de Fusión/inmunología , Anticuerpos Monoclonales/inmunología , Sitios de Unión , Línea Celular , Cromatografía Liquida , Ensayo de Inmunoadsorción Enzimática , Humanos , Resonancia por Plasmón de Superficie , Espectrometría de Masas en Tándem , Transfección , Internalización del Virus
5.
Proc Natl Acad Sci U S A ; 110(1): 264-9, 2013 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-23175789

RESUMEN

Affinity maturation refines a naive B-cell response by selecting mutations in antibody variable domains that enhance antigen binding. We describe a B-cell lineage expressing broadly neutralizing influenza virus antibodies derived from a subject immunized with the 2007 trivalent vaccine. The lineage comprises three mature antibodies, the unmutated common ancestor, and a common intermediate. Their heavy-chain complementarity determining region inserts into the conserved receptor-binding pocket of influenza HA. We show by analysis of structures, binding kinetics and long time-scale molecular dynamics simulations that antibody evolution in this lineage has rigidified the initially flexible heavy-chain complementarity determining region by two nearly independent pathways and that this preconfiguration accounts for most of the affinity gain. The results advance our understanding of strategies for developing more broadly effective influenza vaccines.


Asunto(s)
Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales/genética , Linfocitos B/inmunología , Sitios de Unión de Anticuerpos/genética , Vacunas contra la Influenza/inmunología , Modelos Moleculares , Orthomyxoviridae/inmunología , Secuencia de Aminoácidos , Anticuerpos Neutralizantes/química , Cristalografía por Rayos X , Evolución Molecular , Fragmentos Fab de Inmunoglobulinas/química , Región Variable de Inmunoglobulina/genética , Simulación de Dinámica Molecular , Datos de Secuencia Molecular
6.
EMBO J ; 30(2): 408-16, 2011 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-21157433

RESUMEN

Non-enveloped viruses of different types have evolved distinct mechanisms for penetrating a cellular membrane during infection. Rotavirus penetration appears to occur by a process resembling enveloped-virus fusion: membrane distortion linked to conformational changes in a viral protein. Evidence for such a mechanism comes from crystallographic analyses of fragments of VP4, the rotavirus-penetration protein, and infectivity analyses of structure-based VP4 mutants. We describe here the structure of an infectious rotavirus particle determined by electron cryomicroscopy (cryoEM) and single-particle analysis at about 4.3 Å resolution. The cryoEM image reconstruction permits a nearly complete trace of the VP4 polypeptide chain, including the positions of most side chains. It shows how the two subfragments of VP4 (VP8(*) and VP5(*)) retain their association after proteolytic cleavage, reveals multiple structural roles for the ß-barrel domain of VP5(*), and specifies interactions of VP4 with other capsid proteins. The virion model allows us to integrate structural and functional information into a coherent mechanism for rotavirus entry.


Asunto(s)
Proteínas de la Cápside/genética , Modelos Moleculares , Rotavirus/ultraestructura , Virión/ultraestructura , Internalización del Virus , Membrana Celular/metabolismo , Microscopía por Crioelectrón , Cristalografía , Estructura Secundaria de Proteína , Rotavirus/metabolismo
7.
Proc Natl Acad Sci U S A ; 108(23): 9619-24, 2011 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-21586636

RESUMEN

Respiratory syncytial virus (RSV), the main cause of infant bronchiolitis, remains a major unmet vaccine need despite more than 40 years of vaccine research. Vaccine candidates based on a chief RSV neutralization antigen, the fusion (F) glycoprotein, have foundered due to problems with stability, purity, reproducibility, and potency. Crystal structures of related parainfluenza F glycoproteins have revealed a large conformational change between the prefusion and postfusion states, suggesting that postfusion F antigens might not efficiently elicit neutralizing antibodies. We have generated a homogeneous, stable, and reproducible postfusion RSV F immunogen that elicits high titers of neutralizing antibodies in immunized animals. The 3.2-Å X-ray crystal structure of this substantially complete RSV F reveals important differences from homology-based structural models. Specifically, the RSV F crystal structure demonstrates the exposure of key neutralizing antibody binding sites on the surface of the postfusion RSV F trimer. This unanticipated structural feature explains the engineered RSV F antigen's efficiency as an immunogen. This work illustrates how structural-based antigen design can guide the rational optimization of candidate vaccine antigens.


Asunto(s)
Anticuerpos Antivirales/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Proteínas Virales de Fusión/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales Humanizados , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Sitios de Unión de Anticuerpos , Dicroismo Circular , Cristalografía por Rayos X , Humanos , Inmunización , Lactante , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica , Modelos Moleculares , Datos de Secuencia Molecular , Palivizumab , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/metabolismo , Homología de Secuencia de Aminoácido , Sigmodontinae , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/ultraestructura
8.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 9): 1685-96, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23999292

RESUMEN

Quinolinate synthase (QS) catalyzes the condensation of iminoaspartate and dihydroxyacetone phosphate to form quinolinate, the universal precursor for the de novo biosynthesis of nicotinamide adenine dinucleotide. QS has been difficult to characterize owing either to instability or lack of activity when it is overexpressed and purified. Here, the structure of QS from Pyrococcus furiosus has been determined at 2.8 Šresolution. The structure is a homodimer consisting of three domains per protomer. Each domain shows the same topology with a four-stranded parallel ß-sheet flanked by four α-helices, suggesting that the domains are the result of gene triplication. Biochemical studies of QS indicate that the enzyme requires a [4Fe-4S] cluster, which is lacking in this crystal structure, for full activity. The organization of domains in the protomer is distinctly different from that of a monomeric structure of QS from P. horikoshii [Sakuraba et al. (2005), J. Biol. Chem. 280, 26645-26648]. The domain arrangement in P. furiosus QS may be related to protection of cysteine side chains, which are required to chelate the [4Fe-4S] cluster, prior to cluster assembly.


Asunto(s)
Proteínas Hierro-Azufre/química , Modelos Químicos , Complejos Multienzimáticos/química , Pyrococcus furiosus/enzimología , Secuencia de Aminoácidos , Vías Biosintéticas/genética , Dominio Catalítico , Secuencia Conservada , Cristalografía por Rayos X , Duplicación de Gen/genética , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Datos de Secuencia Molecular , Complejos Multienzimáticos/metabolismo , Multimerización de Proteína/genética , Estructura Terciaria de Proteína/genética , Pyrococcus horikoshii/enzimología , Pyrococcus horikoshii/genética , Especificidad por Sustrato
9.
PLoS Pathog ; 7(9): e1002277, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21980294

RESUMEN

Binding of herpes simplex virus (HSV) glycoprotein D (gD) to a cell surface receptor is required to trigger membrane fusion during entry into host cells. Nectin-1 is a cell adhesion molecule and the main HSV receptor in neurons and epithelial cells. We report the structure of gD bound to nectin-1 determined by x-ray crystallography to 4.0 Å resolution. The structure reveals that the nectin-1 binding site on gD differs from the binding site of the HVEM receptor. A surface on the first Ig-domain of nectin-1, which mediates homophilic interactions of Ig-like cell adhesion molecules, buries an area composed by residues from both the gD N- and C-terminal extensions. Phenylalanine 129, at the tip of the loop connecting ß-strands F and G of nectin-1, protrudes into a groove on gD, which is otherwise occupied by C-terminal residues in the unliganded gD and by N-terminal residues in the gD/HVEM complex. Notably, mutation of Phe129 to alanine prevents nectin-1 binding to gD and HSV entry. Together these data are consistent with previous studies showing that gD disrupts the normal nectin-1 homophilic interactions. Furthermore, the structure of the complex supports a model in which gD-receptor binding triggers HSV entry through receptor-mediated displacement of the gD C-terminal region.


Asunto(s)
Moléculas de Adhesión Celular/química , Herpesvirus Humano 1/química , Receptores Virales/química , Proteínas del Envoltorio Viral/química , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Herpesvirus Humano 1/fisiología , Humanos , Nectinas , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores Virales/genética , Receptores Virales/metabolismo , Relación Estructura-Actividad , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
10.
EMBO Rep ; 12(12): 1300-5, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22081141

RESUMEN

The protein PCSK9 (proprotein convertase subtilisin/kexin type 9) is a key regulator of low-density lipoprotein receptor (LDLR) levels and cardiovascular health. We have determined the crystal structure of LDLR bound to PCSK9 at neutral pH. The structure shows LDLR in a new extended conformation. The PCSK9 C-terminal domain is solvent exposed, enabling cofactor binding, whereas the catalytic domain and prodomain interact with LDLR epidermal growth factor(A) and ß-propeller domains, respectively. Thus, PCSK9 seems to hold LDLR in an extended conformation and to interfere with conformational rearrangements required for LDLR recycling.


Asunto(s)
Proproteína Convertasas/química , Receptores de LDL/química , Receptores de LDL/metabolismo , Serina Endopeptidasas/química , Regulación hacia Abajo , Humanos , Concentración de Iones de Hidrógeno , Modelos Moleculares , Proproteína Convertasa 9 , Proproteína Convertasas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteolisis , Serina Endopeptidasas/metabolismo , Resonancia por Plasmón de Superficie
12.
NPJ Vaccines ; 8(1): 150, 2023 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-37794051

RESUMEN

Currently licensed influenza vaccines focus immune responses on viral hemagglutinin (HA), while the other major surface glycoprotein neuraminidase (NA) is not tightly controlled in inactivated vaccine formulations despite evidence that anti-NA antibodies reduce clinical disease. We utilized a bicistronic self-amplifying mRNA (sa-mRNA) platform encoding both HA and NA from four seasonal influenza strains, creating a quadrivalent influenza vaccine. sa-mRNA vaccines encoding an NA component induced the production of NA-inhibiting antibodies and CD4+ T-cell responses in both monovalent and quadrivalent formulations. Including NA in the vaccine enabled cross-neutralization against antigenically drifted strains and provided greater protection than HA alone upon A(H3N2) challenge in ferrets. These results demonstrate that next-generation bicistronic sa-mRNA vaccines expressing HA and NA induce potent antibodies against both viral coat proteins, as well as vaccine-specific cell-mediated immunity. When formulated as a quadrivalent seasonal influenza vaccine, the sa-mRNA platform provides an opportunity to increase the breadth of protection through cross-neutralizing anti-NA antibodies.

13.
Proc Natl Acad Sci U S A ; 106(26): 10644-8, 2009 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-19487668

RESUMEN

Rotaviruses, major causes of childhood gastroenteritis, are nonenveloped, icosahedral particles with double-strand RNA genomes. By the use of electron cryomicroscopy and single-particle reconstruction, we have visualized a rotavirus particle comprising the inner capsid coated with the trimeric outer-layer protein, VP7, at a resolution (4 A) comparable with that of X-ray crystallography. We have traced the VP7 polypeptide chain, including parts not seen in its X-ray crystal structure. The 3 well-ordered, 30-residue, N-terminal "arms" of each VP7 trimer grip the underlying trimer of VP6, an inner-capsid protein. Structural differences between free and particle-bound VP7 and between free and VP7-coated inner capsids may regulate mRNA transcription and release. The Ca(2+)-stabilized VP7 intratrimer contact region, which presents important neutralizing epitopes, is unaltered upon capsid binding.


Asunto(s)
Microscopía por Crioelectrón/métodos , Rotavirus/ultraestructura , Virión/ultraestructura , Antígenos Virales/química , Antígenos Virales/metabolismo , Antígenos Virales/ultraestructura , Calcio/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/ultraestructura , Cristalografía por Rayos X , Modelos Biológicos , Modelos Moleculares , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Rotavirus/metabolismo , Proteínas del Núcleo Viral/química , Proteínas del Núcleo Viral/metabolismo , Proteínas del Núcleo Viral/ultraestructura , Virión/metabolismo
14.
Mol Ther Methods Clin Dev ; 25: 225-235, 2022 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-35345593

RESUMEN

The spike (S) protein of SARS-CoV-2 plays a crucial role in cell entry, and the nucleocapsid (N) protein is highly conserved among human coronavirus homologs. For potentially broad effectiveness against both original virus and emerging variants, we developed Alphavirus-based self-amplifying mRNA (sa-mRNA) SARS-CoV-2 vaccines: an sa-mRNA S encoding a full-length S protein stabilized in a prefusion conformation and an sa-mRNA S-N co-expressing S and N proteins for the original virus. We show that these sa-mRNA SARS-CoV-2 vaccines raised potent neutralizing antibody responses in mice against not only the original virus but also the Alpha, Beta, Gamma, and Delta variants. sa-mRNA S vaccines against the Alpha and Beta variants also raised robust cross-reactive neutralizing antibody responses against their homologous viruses and heterologous variants. sa-mRNA S and sa-mRNA S-N vaccines elicited Th1-dominant, antigen-specific CD4+ T cell responses to S and N proteins and robust and broad CD8+ T cell responses to S protein. Hamsters immunized with either vaccine were fully protected from lung infection and showed significant reduction of viral load in upper respiratory tract. Our findings demonstrate that sa-mRNA SARS-CoV-2 vaccines are potent in animal models with potential to be highly effective against SARS-CoV-2 infection in humans.

15.
Mol Ther Methods Clin Dev ; 27: 195-205, 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36320414

RESUMEN

Vaccines are the primary intervention against influenza. Currently licensed inactivated vaccines focus immunity on viral hemagglutinin (HA). Self-amplifying mRNA (sa-mRNA) vaccines offer an opportunity to generate immunity to multiple viral proteins, including additional neuraminidase (NA). This evaluation of a bicistronic approach for sa-mRNA vaccine development compared subgenomic promoter and internal ribosome entry site strategies and found consistent and balanced expression of both HA and NA proteins in transfected cells. In mice, sa-mRNA bicistronic A/H5N1 vaccines raised potent anti-HA and anti-NA neutralizing antibody responses and HA- or NA-specific CD4+ and CD8+ T cell responses. The addition of NA also boosted the cross-neutralizing response to heterologous A/H1N1. Similar immunogenicity results were obtained for bicistronic seasonal A/H3N2 and B/Yamagata vaccines. In ferrets, sa-mRNA bicistronic A/H1N1 vaccine fully protected lung from infection by homologous virus and showed significant reduction of viral load in upper respiratory tract, warranting further evaluation of sa-mRNA bicistronic vaccine in humans.

16.
J Virol ; 84(7): 3270-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20053749

RESUMEN

The native envelope (Env) spike on the surface of human immunodeficiency virus type 1 (HIV-1) is trimeric, and thus trimeric Env vaccine immunogens are currently being explored in preclinical immunogenicity studies. Key challenges have included the production and purification of biochemically homogeneous and stable trimers and the evaluation of these immunogens utilizing standardized virus panels for neutralization assays. Here we report the binding and neutralizing antibody (NAb) responses elicited by clade A (92UG037.8) and clade C (CZA97.012) Env gp140 trimer immunogens in guinea pigs. These trimers have been selected and engineered for optimal biochemical stability and have defined antigenic properties. Purified gp140 trimers with Ribi adjuvant elicited potent, cross-clade NAb responses against tier 1 viruses as well as detectable but low-titer NAb responses against select tier 2 viruses from clades A, B, and C. In particular, the clade C trimer elicited NAbs that neutralized 27%, 20%, and 47% of tier 2 viruses from clades A, B, and C, respectively. Heterologous DNA prime, protein boost as well as DNA prime, recombinant adenovirus boost regimens expressing these antigens, however, did not result in an increased magnitude or breadth of NAb responses in this system. These data demonstrate the immunogenicity of stable, homogeneous clade A and clade C gp140 trimers and exemplify the utility of standardized tier 1 and tier 2 virus panels for assessing the NAb responses of candidate HIV-1 Env immunogens.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Neutralizantes/sangre , Anticuerpos Anti-VIH/sangre , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Secuencia de Aminoácidos , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Cobayas , Inmunización , Datos de Secuencia Molecular , Productos del Gen env del Virus de la Inmunodeficiencia Humana/química
17.
Vaccine ; 39(38): 5351-5357, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34393015

RESUMEN

Vaccination is the most effective approach to reduce the substantial morbidity and mortality caused by influenza infection. Vaccine efficacy is highly sensitive to antigenic changes causing differences between circulating and vaccine viruses. Adjuvants such as MF59 increase antibody-mediated cross-reactive immunity and therefore may provide broader seasonal protection. A recent clinical trial showed that an MF59-adjuvanted vaccine was more efficacious than a nonadjuvanted comparator in subjects < 2 years of age, although not in those ≥ 2 years, during influenza seasons in which the predominant circulating virus was an A/H3N2 strain that was antigenically different from the vaccine virus. This finding suggested that the increased efficacy of the adjuvanted vaccine in younger subjects may be mediated by strain cross-reactive antibodies. A subset of the trial population, representing subjects with distinct age and/or immunological history, was tested for antibody responses to the vaccine A/H3N2 strain as well as A/H3N2 drifted strains antigenically matching the viruses circulating during the trial seasons. The neutralizing tests showed that, compared with nonadjuvanted vaccine, the adjuvanted vaccine improved not only the neutralizing antibody response to the vaccine strain but also the cross-reactive antibody response to the drifted strains in subjects with lower preexisting antibody titers, regardless of their age or vaccine history. The results demonstrated an immunological benefit and suggested a potential efficacy benefit by adjuvanted vaccine in subjects with lower preexisting antibody responses.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Anticuerpos Antivirales , Formación de Anticuerpos , Niño , Preescolar , Humanos , Subtipo H3N2 del Virus de la Influenza A , Gripe Humana/prevención & control , Polisorbatos , Escualeno
18.
Vaccines (Basel) ; 9(8)2021 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-34451986

RESUMEN

Children with sickle cell disease (SCD) suffer life-threatening transient aplastic crisis (TAC) when infected with parvovirus B19. In utero, infection of healthy fetuses may result in anemia, hydrops, and death. Unfortunately, although promising vaccine candidates exist, no product has yet been licensed. One barrier to vaccine development has been the lack of a cost-effective, standardized parvovirus B19 neutralization assay. To fill this void, we evaluated the unique region of VP1 (VP1u), which contains prominent targets of neutralizing antibodies. We discovered an antigenic cross-reactivity between VP1 and VP2 that, at first, thwarted the development of a surrogate neutralization assay. We overcame the cross-reactivity by designing a mutated VP1u (VP1uAT) fragment. A new VP1uAT ELISA yielded results well correlated with neutralization (Spearman's correlation coefficient = 0.581; p = 0.001), superior to results from a standard clinical diagnostic ELISA or an ELISA with virus-like particles. Virus-specific antibodies from children with TAC, measured by the VP1uAT and neutralization assays, but not other assays, gradually increased from days 0 to 120 post-hospitalization. We propose that this novel and technically simple VP1uAT ELISA might now serve as a surrogate for the neutralization assay to support rapid development of a parvovirus B19 vaccine.

19.
Biochemistry ; 49(16): 3499-509, 2010 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-20364833

RESUMEN

Uridine phosphorylase is a key enzyme in the pyrimidine salvage pathway. This enzyme catalyzes the reversible phosphorolysis of uridine to uracil and ribose 1-phosphate (or 2'-deoxyuridine to 2'-deoxyribose 1-phosphate). Here we report the structure of hexameric Escherichia coli uridine phosphorylase treated with 5-fluorouridine and sulfate and dimeric bovine uridine phosphorylase treated with 5-fluoro-2'-deoxyuridine or uridine, plus sulfate. In each case the electron density shows three separate species corresponding to the pyrimidine base, sulfate, and a ribosyl species, which can be modeled as a glycal. In the structures of the glycal complexes, the fluorouracil O2 atom is appropriately positioned to act as the base required for glycal formation via deprotonation at C2'. Crystals of bovine uridine phosphorylase treated with 2'-deoxyuridine and sulfate show intact nucleoside. NMR time course studies demonstrate that uridine phosphorylase can catalyze the hydrolysis of the fluorinated nucleosides in the absence of phosphate or sulfate, without the release of intermediates or enzyme inactivation. These results add a previously unencountered mechanistic motif to the body of information on glycal formation by enzymes catalyzing the cleavage of glycosyl bonds.


Asunto(s)
Uridina Fosforilasa/química , Dominio Catalítico , Clonación Molecular , Cristalización , Escherichia coli/enzimología , Floxuridina/farmacología , Humanos , Cinética , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sulfatos/farmacología , Ultracentrifugación , Uridina/farmacología , Uridina Fosforilasa/genética , Uridina Fosforilasa/metabolismo
20.
Vaccine ; 36(21): 3010-3017, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29680201

RESUMEN

Influenza vaccines are the most effective intervention to prevent the substantial public health burden of seasonal and pandemic influenza. The capability of hemagglutinin (HA), the main antigen in inactivated influenza vaccines (IIVs), to elicit functional neutralizing antibodies determines IIV effectiveness. When HA is subjected to environmental stress during manufacturing or while stored prior to administration, such as low pH and temperature excursions, the HA immunological activity can be affected. Single-radial immunodiffusion (SRID), the standard in vitro potency assay for IIVs, is believed to specifically detect immunologically active HA and has been applied to evaluate HA stability against stress. Here we report that transient low pH treatment and freeze/thaw cycles with HA in PBS abolish SRID-quantified in vitro potency for all HAs of multiple influenza strains. Raised temperature substantially decreases in vitro potency with more extensive HA structural changes. Chemical stress and mechanical stress moderately change SRID in vitro potency values in a strain-dependent manner. Trypsin digestion, which selectively degrades stressed HA, followed by RP-HPLC quantification as a candidate alternative in vitro potency assay yields results comparable to SRID. Mouse immunogenicity studies confirm that HA stressed by transient low pH treatment does not elicit functional antibodies in vivo, nor does it have a measureable SRID value. However, HA stressed by raised temperature elicits high titers of functional antibodies in vivo despite substantial loss of SRID in vitro potency. This discrepancy between SRID in vitro potency and vaccine immunogenicity suggests that SRID may not reliably indicate IIV potency under all conditions. Further efforts to develop alternate potency assays that can better predict in vivo immunogenicity should continue along with additional studies exploring HA conformation, SRID values and consequent immunogenicity.


Asunto(s)
Almacenaje de Medicamentos/métodos , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Tecnología Farmacéutica/métodos , Potencia de la Vacuna , Animales , Anticuerpos Antivirales/sangre , Femenino , Congelación , Concentración de Iones de Hidrógeno , Vacunas contra la Influenza/aislamiento & purificación , Vacunas contra la Influenza/efectos de la radiación , Ratones Endogámicos BALB C , Temperatura , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Vacunas de Productos Inactivados/aislamiento & purificación , Vacunas de Productos Inactivados/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA