Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.439
Filtrar
1.
J Med Virol ; 96(5): e29657, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38727035

RESUMEN

The H1N1pdm09 virus has been a persistent threat to public health since the 2009 pandemic. Particularly, since the relaxation of COVID-19 pandemic mitigation measures, the influenza virus and SARS-CoV-2 have been concurrently prevalent worldwide. To determine the antigenic evolution pattern of H1N1pdm09 and develop preventive countermeasures, we collected influenza sequence data and immunological data to establish a new antigenic evolution analysis framework. A machine learning model (XGBoost, accuracy = 0.86, area under the receiver operating characteristic curve = 0.89) was constructed using epitopes, physicochemical properties, receptor binding sites, and glycosylation sites as features to predict the antigenic similarity relationships between influenza strains. An antigenic correlation network was constructed, and the Markov clustering algorithm was used to identify antigenic clusters. Subsequently, the antigenic evolution pattern of H1N1pdm09 was analyzed at the global and regional scales across three continents. We found that H1N1pdm09 evolved into around five antigenic clusters between 2009 and 2023 and that their antigenic evolution trajectories were characterized by cocirculation of multiple clusters, low-level persistence of former dominant clusters, and local heterogeneity of cluster circulations. Furthermore, compared with the seasonal H1N1 virus, the potential cluster-transition determining sites of H1N1pdm09 were restricted to epitopes Sa and Sb. This study demonstrated the effectiveness of machine learning methods for characterizing antigenic evolution of viruses, developed a specific model to rapidly identify H1N1pdm09 antigenic variants, and elucidated their evolutionary patterns. Our findings may provide valuable support for the implementation of effective surveillance strategies and targeted prevention efforts to mitigate the impact of H1N1pdm09.


Asunto(s)
Antígenos Virales , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Humanos , Gripe Humana/epidemiología , Gripe Humana/prevención & control , Gripe Humana/virología , Gripe Humana/inmunología , Antígenos Virales/genética , Antígenos Virales/inmunología , Aprendizaje Automático , Evolución Molecular , Epítopos/genética , Epítopos/inmunología , COVID-19/epidemiología , COVID-19/prevención & control , COVID-19/virología , COVID-19/inmunología , Pandemias/prevención & control , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , SARS-CoV-2/genética , SARS-CoV-2/inmunología
2.
Nat Commun ; 15(1): 3833, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714654

RESUMEN

Antigenic characterization of circulating influenza A virus (IAV) isolates is routinely assessed by using the hemagglutination inhibition (HI) assays for surveillance purposes. It is also used to determine the need for annual influenza vaccine updates as well as for pandemic preparedness. Performing antigenic characterization of IAV on a global scale is confronted with high costs, animal availability, and other practical challenges. Here we present a machine learning model that accurately predicts (normalized) outputs of HI assays involving circulating human IAV H3N2 viruses, using their hemagglutinin subunit 1 (HA1) sequences and associated metadata. Each season, the model learns an updated nonlinear mapping of genetic to antigenic changes using data from past seasons only. The model accurately distinguishes antigenic variants from non-variants and adaptively characterizes seasonal dynamics of HA1 sites having the strongest influence on antigenic change. Antigenic predictions produced by the model can aid influenza surveillance, public health management, and vaccine strain selection activities.


Asunto(s)
Antígenos Virales , Glicoproteínas Hemaglutininas del Virus de la Influenza , Subtipo H3N2 del Virus de la Influenza A , Gripe Humana , Aprendizaje Automático , Estaciones del Año , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/genética , Humanos , Gripe Humana/inmunología , Gripe Humana/virología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Antígenos Virales/inmunología , Antígenos Virales/genética , Pruebas de Inhibición de Hemaglutinación , Variación Antigénica/genética , Vacunas contra la Influenza/inmunología
3.
Sci Rep ; 14(1): 12184, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38806597

RESUMEN

Catalytic antibodies possess a dual function that enables both antigen recognition and degradation. However, their time-consuming preparation is a significant drawback. This study developed a new method for quickly converting mice monoclonal antibodies into catalytic antibodies using site-directed mutagenesis. Three mice type monoclonal antibodies targeting hemagglutinin molecule of influenza A virus could be transformed into the catalytic antibodies by deleting Pro95 in CDR-3 of the light chain. No catalytic activity was observed for monoclonal antibodies and light chains. In contrast, the Pro95-deleted light chains exhibited a catalytic activity to cleave the antigenic peptide including the portion of conserved region of hemagglutinin molecule. The affinity of the Pro95-deleted light chains to the antigen increased approximately 100-fold compared to the wild-type light chains. In the mutants, three residues (Asp1, Ser92, and His93) come closer to the appropriate position to create the catalytic site and contributing to the enhancement of both catalytic function and immunoreactivity. Notably, the Pro95-deleted catalytic light chains could suppress influenza virus infection in vitro assay, whereas the parent antibody and the light chain did not. This strategy offers a rapid and efficient way to create catalytic antibodies from existing antibodies, accelerating the development for various applications in diagnostic and therapeutic applications.


Asunto(s)
Anticuerpos Catalíticos , Anticuerpos Monoclonales , Animales , Ratones , Anticuerpos Monoclonales/inmunología , Anticuerpos Catalíticos/metabolismo , Anticuerpos Catalíticos/inmunología , Anticuerpos Catalíticos/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Mutagénesis Sitio-Dirigida , Virus de la Influenza A/inmunología , Dominio Catalítico , Humanos , Cadenas Ligeras de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/inmunología , Cadenas Ligeras de Inmunoglobulina/metabolismo , Anticuerpos Antivirales/inmunología , Ratones Endogámicos BALB C
4.
Sci Transl Med ; 16(745): eadj4685, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38691617

RESUMEN

Current seasonal influenza virus vaccines induce responses primarily against immunodominant but highly plastic epitopes in the globular head of the hemagglutinin (HA) glycoprotein. Because of viral antigenic drift at these sites, vaccines need to be updated and readministered annually. To increase the breadth of influenza vaccine-mediated protection, we developed an antigenically complex mixture of recombinant HAs designed to redirect immune responses to more conserved domains of the protein. Vaccine-induced antibodies were disproportionally redistributed to the more conserved stalk of the HA without hindering, and in some cases improving, antibody responses against the head domain. These improved responses led to increased protection against homologous and heterologous viral challenges in both mice and ferrets compared with conventional vaccine approaches. Thus, antigenically complex protein mixtures can at least partially overcome HA head domain antigenic immunodominance and may represent a step toward a more universal influenza vaccine.


Asunto(s)
Hurones , Glicoproteínas Hemaglutininas del Virus de la Influenza , Vacunas contra la Influenza , Vacunación , Animales , Vacunas contra la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/inmunología , Ratones , Anticuerpos Antivirales/inmunología , Humanos , Gripe Humana/prevención & control , Gripe Humana/inmunología , Antígenos Virales/inmunología , Femenino , Ratones Endogámicos BALB C
5.
Proc Natl Acad Sci U S A ; 121(22): e2310677121, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38753503

RESUMEN

Seasonal and pandemic-associated influenza strains cause highly contagious viral respiratory infections that can lead to severe illness and excess mortality. Here, we report on the optimization of our small-molecule inhibitor F0045(S) targeting the influenza hemagglutinin (HA) stem with our Sulfur-Fluoride Exchange (SuFEx) click chemistry-based high-throughput medicinal chemistry (HTMC) strategy. A combination of SuFEx- and amide-based lead molecule diversification and structure-guided design led to identification and validation of ultrapotent influenza fusion inhibitors with subnanomolar EC50 cellular antiviral activity against several influenza A group 1 strains. X-ray structures of six of these compounds with HA indicate that the appended moieties occupy additional pockets on the HA surface and increase the binding interaction, where the accumulation of several polar interactions also contributes to the improved affinity. The compounds here represent the most potent HA small-molecule inhibitors to date. Our divergent HTMC platform is therefore a powerful, rapid, and cost-effective approach to develop bioactive chemical probes and drug-like candidates against viral targets.


Asunto(s)
Antivirales , Glicoproteínas Hemaglutininas del Virus de la Influenza , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Humanos , Antivirales/farmacología , Antivirales/química , Química Farmacéutica/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Cristalografía por Rayos X/métodos , Química Clic/métodos , Animales , Virus de la Influenza A/efectos de los fármacos , Células de Riñón Canino Madin Darby , Inhibidores de Proteínas Virales de Fusión/farmacología , Inhibidores de Proteínas Virales de Fusión/química , Perros
6.
Acta Biochim Pol ; 71: 12289, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38721309

RESUMEN

The aim of the study was to determine the level of anti-hemagglutinin antibodies in the serum of patients during the 2021/2022 epidemic season in Poland. A total of 700 sera samples were tested, divided according to the age of the patients into 7 age groups: 0-4 years of age, 5-9 years of age, 10-14 years of age, 15-25 years of age, 26-44 years of age, 45-64 years of age and ≥65 years of age, 100 samples were collected from each age group. Anti-hemagglutinin antibody levels was determined using the haemagglutination inhibition assay (OZHA). The results obtained confirm the presence of anti-hemagglutinin antibodies for the antigens A/Victoria/2570/2019 (H1N1) pdm09, A/Cambodia/e0826360/2020 (H3N2), B/Washington/02/2019 and B/Phuket/3073/2013 recommended by World Health Organization (WHO) for the 2021/2022 epidemic season. The analysis of the results shows differences in the levels of individual anti-hemagglutinin antibodies in the considered age groups. In view of very low percentage of the vaccinated population in Poland, which was 6.90% in the 2021/2022 epidemic season, the results obtained in the study would have to be interpreted as the immune system response in patients after a previous influenza virus infection.


Asunto(s)
Anticuerpos Antivirales , Glicoproteínas Hemaglutininas del Virus de la Influenza , Subtipo H1N1 del Virus de la Influenza A , Subtipo H3N2 del Virus de la Influenza A , Gripe Humana , Humanos , Polonia/epidemiología , Adulto , Persona de Mediana Edad , Adolescente , Gripe Humana/inmunología , Gripe Humana/epidemiología , Gripe Humana/sangre , Gripe Humana/virología , Niño , Anciano , Preescolar , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Adulto Joven , Lactante , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Masculino , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Femenino , Recién Nacido , Pruebas de Inhibición de Hemaglutinación , Virus de la Influenza B/inmunología , Estaciones del Año , Epidemias , Prevalencia
7.
Front Immunol ; 15: 1352022, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38698856

RESUMEN

The complement system is an innate immune mechanism against microbial infections. It involves a cascade of effector molecules that is activated via classical, lectin and alternative pathways. Consequently, many pathogens bind to or incorporate in their structures host negative regulators of the complement pathways as an evasion mechanism. Factor H (FH) is a negative regulator of the complement alternative pathway that protects "self" cells of the host from non-specific complement attack. FH has been shown to bind viruses including human influenza A viruses (IAVs). In addition to its involvement in the regulation of complement activation, FH has also been shown to perform a range of functions on its own including its direct interaction with pathogens. Here, we show that human FH can bind directly to IAVs of both human and avian origin, and the interaction is mediated via the IAV surface glycoprotein haemagglutinin (HA). HA bound to common pathogen binding footprints on the FH structure, complement control protein modules, CCP 5-7 and CCP 15-20. The FH binding to H1 and H3 showed that the interaction overlapped with the receptor binding site of both HAs, but the footprint was more extensive for the H3 HA than the H1 HA. The HA - FH interaction impeded the initial entry of H1N1 and H3N2 IAV strains but its impact on viral multicycle replication in human lung cells was strain-specific. The H3N2 virus binding to cells was significantly inhibited by preincubation with FH, whereas there was no alteration in replicative rate and progeny virus release for human H1N1, or avian H9N2 and H5N3 IAV strains. We have mapped the interaction between FH and IAV, the in vivo significance of which for the virus or host is yet to be elucidated.


Asunto(s)
Factor H de Complemento , Glicoproteínas Hemaglutininas del Virus de la Influenza , Virus de la Influenza A , Gripe Humana , Unión Proteica , Humanos , Factor H de Complemento/metabolismo , Factor H de Complemento/inmunología , Animales , Gripe Humana/inmunología , Gripe Humana/virología , Gripe Humana/metabolismo , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Sitios de Unión , Gripe Aviar/virología , Gripe Aviar/inmunología , Gripe Aviar/metabolismo , Aves/virología , Interacciones Huésped-Patógeno/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H9N2 del Virus de la Influenza A/inmunología
8.
Nat Commun ; 15(1): 4350, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38782954

RESUMEN

mRNA lipid nanoparticle (LNP) vaccines would be useful during an influenza virus pandemic since they can be produced rapidly and do not require the generation of egg-adapted vaccine seed stocks. Highly pathogenic avian influenza viruses from H5 clade 2.3.4.4b are circulating at unprecedently high levels in wild and domestic birds and have the potential to adapt to humans. Here, we generate an mRNA lipid nanoparticle (LNP) vaccine encoding the hemagglutinin (HA) glycoprotein from a clade 2.3.4.4b H5 isolate. The H5 mRNA-LNP vaccine elicits strong T cell and antibody responses in female mice, including neutralizing antibodies and broadly-reactive anti-HA stalk antibodies. The H5 mRNA-LNP vaccine elicits antibodies at similar levels compared to whole inactivated vaccines in female mice with and without prior H1N1 exposures. Finally, we find that the H5 mRNA-LNP vaccine is immunogenic in male ferrets and prevents morbidity and mortality of animals following 2.3.4.4b H5N1 challenge. Together, our data demonstrate that a monovalent mRNA-LNP vaccine expressing 2.3.4.4b H5 is immunogenic and protective in pre-clinical animal models.


Asunto(s)
Anticuerpos Antivirales , Hurones , Glicoproteínas Hemaglutininas del Virus de la Influenza , Subtipo H5N1 del Virus de la Influenza A , Vacunas contra la Influenza , Nanopartículas , Infecciones por Orthomyxoviridae , Vacunas de ARNm , Animales , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Femenino , Ratones , Nanopartículas/química , Masculino , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/genética , Anticuerpos Antivirales/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Vacunas de ARNm/inmunología , Anticuerpos Neutralizantes/inmunología , Ratones Endogámicos BALB C , Gripe Aviar/prevención & control , Gripe Aviar/inmunología , Gripe Aviar/virología , Humanos , ARN Mensajero/genética , ARN Mensajero/inmunología , ARN Mensajero/metabolismo , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/genética , Aves/virología , Lípidos/química , Liposomas
9.
Eur J Med Chem ; 272: 116469, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38704939

RESUMEN

Accurate diagnosis and effective antiviral treatments are urgently needed for the prevention and control of flu caused by influenza viruses. In this study, a novel oleanic acid (OA) functionalized gold nanorod OA-AuNP was prepared through a convenient ligand-exchange reaction. As hemagglutinin (HA) on the viral surface binds strongly to the multiple OA molecules on the surface of the nanoparticle, the prepared OA-AuNP was found to exhibit potent antiviral activity against a wide range of influenza A virus strains. Furthermore, the change in color resulting from the specific binding between HA and OA and the resultant aggregation of the OA-AuNP can be visually observed or measured by UV-vis spectra with a detection limit of 2 and 0.18 hemagglutination units (HAU), respectively, which is comparable to the commercially available influenza colloid gold rapid diagnostic kits. These findings demonstrate the potential of the OA-AuNP for the development of novel multivalent antiviral conjugates and the diagnosis of influenza virus.


Asunto(s)
Antivirales , Oro , Nanotubos , Oro/química , Nanotubos/química , Antivirales/farmacología , Antivirales/química , Virus de la Influenza A/efectos de los fármacos , Humanos , Nanopartículas del Metal/química , Estructura Molecular , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Pruebas de Sensibilidad Microbiana , Perros , Animales , Relación Dosis-Respuesta a Droga , Relación Estructura-Actividad
10.
Viruses ; 16(5)2024 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-38793574

RESUMEN

Influenza viruses are constantly evolving and are therefore monitored worldwide in the hope to reduce the burden of disease by annual updates to vaccine recommendations. We conducted genomic sequencing of 110 influenza A and 30 influenza B viruses from specimens collected between October 2023 and February 2024 in Arizona, USA. We identified mutations in the hemagglutinin (HA) antigenic sites as well as the neuraminidase (NA) gene in our samples. We also found no unique HA and NA mutations in vaccinated yet influenza-infected individuals. Real-time genomic sequencing surveillance is important to ensure influenza vaccine effectiveness.


Asunto(s)
Genoma Viral , Virus de la Influenza A , Virus de la Influenza B , Gripe Humana , Mutación , Neuraminidasa , Arizona/epidemiología , Humanos , Gripe Humana/epidemiología , Gripe Humana/virología , Neuraminidasa/genética , Virus de la Influenza B/genética , Virus de la Influenza A/genética , Virus de la Influenza A/clasificación , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Genómica/métodos , Filogenia , Adulto , Monitoreo Epidemiológico , Niño , Adolescente , Persona de Mediana Edad , Masculino , Femenino , Preescolar , Anciano , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/genética , Adulto Joven , Secuenciación Completa del Genoma
11.
Nat Commun ; 15(1): 4505, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802413

RESUMEN

Avian influenza A virus H7N9 causes severe human infections with >30% fatality. Currently, there is no H7N9-specific prevention or treatment for humans. Here, from a 2013 H7N9 convalescent case in Hong Kong, we isolate four hemagglutinin (HA)-reactive monoclonal antibodies (mAbs), with three directed to the globular head domain (HA1) and one to the stalk domain (HA2). Two clonally related HA1-directed mAbs, H7.HK1 and H7.HK2, potently neutralize H7N9 and protect female mice from lethal H7N9/AH1 challenge. Cryo-EM structures reveal that H7.HK1 and H7.HK2 bind to a ß14-centered surface and disrupt the 220-loop that makes hydrophobic contacts with sialic acid on an adjacent protomer, thereby blocking viral entry. Sequence analysis indicates the lateral patch targeted by H7.HK1 and H7.HK2 to be conserved among influenza subtypes. Both H7.HK1 and H7.HK2 retain HA1 binding and neutralization capacity to later H7N9 isolates from 2016-2017, consistent with structural data showing that the antigenic mutations during this timeframe occur at their epitope peripheries. The HA2-directed mAb H7.HK4 lacks neutralizing activity but when used in combination with H7.HK2 moderately augments female mouse protection. Overall, our data reveal antibodies to a conserved lateral HA1 supersite that confer neutralization, and when combined with a HA2-directed non-neutralizing mAb, augment protection.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Glicoproteínas Hemaglutininas del Virus de la Influenza , Subtipo H7N9 del Virus de la Influenza A , Gripe Humana , Subtipo H7N9 del Virus de la Influenza A/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Humanos , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Femenino , Gripe Humana/inmunología , Gripe Humana/virología , Gripe Humana/prevención & control , Ratones , Anticuerpos Antivirales/inmunología , Anticuerpos Monoclonales/inmunología , Ratones Endogámicos BALB C , Microscopía por Crioelectrón , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Epítopos/inmunología
12.
Virus Res ; 345: 199402, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38772446

RESUMEN

H1N1 influenza virus is a significant global public health concern. Monoclonal antibodies (mAbs) targeting specific viral proteins such as hemagglutinin (HA) have become an important therapeutic strategy, offering highly specific targeting to block viral transmission and infection. This study focused on the development of mAbs targeting HA of the A/Victoria/2570/2019 (H1N1pdm09, VIC-19) strain by utilizing hybridoma technology to produce two mAbs with high binding capacity. Notably, mAb 2B2 has demonstrated a strong affinity for HA proteins in recent H1N1 influenza vaccine strains. In vitro assessments showed that both mAbs exhibited broad-spectrum hemagglutination inhibition and potent neutralizing effects against various vaccine strains of H1N1pdm09 viruses. 2B2 was also effective in animal models, offering both preventive and therapeutic protection against infections caused by recent H1N1 strains, highlighting its potential for clinical application. By individually co-cultivating each of the aforementioned mAbs with the virus in chicken embryos, four amino acid substitution sites in HA (H138Q, G140R, A141E/V, and D187E) were identified in escape mutants, three in the antigenic site Ca2, and one in Sb. The identification of such mutations is pivotal, as it compels further investigation into how these alterations could undermine the binding efficacy and neutralization capacity of antibodies, thereby impacting the design and optimization of mAb therapies and influenza vaccines. This research highlights the necessity for continuous exploration into the dynamic interaction between viral evolution and antibody response, which is vital for the formulation of robust therapeutic and preventive strategies against influenza.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Glicoproteínas Hemaglutininas del Virus de la Influenza , Subtipo H1N1 del Virus de la Influenza A , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae , Animales , Subtipo H1N1 del Virus de la Influenza A/inmunología , Anticuerpos Monoclonales/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Anticuerpos Antivirales/inmunología , Ratones , Anticuerpos Neutralizantes/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Pruebas de Inhibición de Hemaglutinación , Humanos , Embrión de Pollo , Femenino , Gripe Humana/inmunología , Gripe Humana/virología , Gripe Humana/prevención & control
13.
Vaccine ; 42(15): 3505-3513, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38714444

RESUMEN

It is necessary to develop universal vaccines that act broadly and continuously to combat regular seasonal epidemics of influenza and rare pandemics. The aim of this study was to find the optimal dose regimen for the efficacy and safety of a mixture of previously developed recombinant adenovirus-based vaccines that expressed influenza nucleoprotein, hemagglutinin, and ectodomain of matrix protein 2 (rAd/NP and rAd/HA-M2e). The vaccine efficacy and safety were measured in the immunized mice with the mixture of rAd/NP and rAd/HA-M2e intranasally or intramuscularly. The minimum dose that would be efficacious in a single intranasal administration of the vaccine mixture and cross-protective efficacy against various influenza strains were examined. In addition, the immune responses that may affect the cross-protective efficacy were measured. We found that intranasal administration is an optimal route for 107 pfu of vaccine mixture, which is effective against pre-existing immunity against adenovirus. In a study to find the minimum dose with vaccine efficacy, the 106 pfu of vaccine mixture showed higher antibody titers to the nucleoprotein than did the same dose of rAd/NP alone in the serum of immunized mice. The 106 pfu of vaccine mixture overcame the morbidity and mortality of mice against the lethal dose of pH1N1, H3N2, and H5N1 influenza infections. No noticeable side effects were observed in single and repeated toxicity studies. We found that the mucosal administration of adenovirus-based universal influenza vaccine has both efficacy and safety, and can provide cross-protection against various influenza infections even at doses lower than those previously known to be effective.


Asunto(s)
Adenoviridae , Administración Intranasal , Anticuerpos Antivirales , Protección Cruzada , Glicoproteínas Hemaglutininas del Virus de la Influenza , Vacunas contra la Influenza , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae , Proteínas de la Matriz Viral , Animales , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Proteínas de la Matriz Viral/inmunología , Proteínas de la Matriz Viral/genética , Adenoviridae/genética , Adenoviridae/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Ratones , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/inmunología , Femenino , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/genética , Eficacia de las Vacunas , Nucleoproteínas/inmunología , Nucleoproteínas/genética , Proteínas del Núcleo Viral/inmunología , Proteínas del Núcleo Viral/genética , Inyecciones Intramusculares , Proteínas Viroporinas
14.
Vaccine ; 42(14): 3365-3373, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38627145

RESUMEN

The head domain of the hemagglutinin of influenza viruses plays a dominant role in the antibody response due to the presence of immunodominant antigenic sites that are the main targets of host neutralizing antibodies. For the H1 hemagglutinin, five major antigenic sites defined as Sa, Sb, Ca1, Ca2, and Cb have been described. Although previous studies have focused on defining the hierarchy of the antigenic sites of the hemagglutinin in different human cohorts, it is still unclear if the immunodominance profile of the antigenic sites might change with the antibody levels of individuals or if other demographic factors (such as exposure history, sex, or age) could also influence the importance of the antigenic sites. The major antigenic sites of influenza viruses hemagglutinins are responsible for eliciting most of the hemagglutination inhibition antibodies in the host. To determine the antibody prevalence towards each major antigenic site, we evaluated the hemagglutination inhibition against a panel of mutant H1 viruses, each one lacking one of the "classic" antigenic sites. Our results showed that the individuals from the Stop Flu NYU cohort had an immunodominant response towards the sites Sb and Ca2 of H1 hemagglutinin. A simple logistic regression analysis of the immunodominance profiles and the hemagglutination inhibition titers displayed by each donor revealed that individuals with high hemagglutination inhibition titers against the wild-type influenza virus exhibited higher probabilities of displaying an immunodominance profile dominated by Sb, followed by Ca2 (Sb > Ca2 profile), while individuals with low hemagglutination inhibition titers presented a higher chance of displaying an immunodominance profile in which Sb and Ca2 presented the same level of immunodominance (Sb = Ca2 profile). Finally, while age exhibited an influence on the immunodominance of the antigenic sites, biological sex was not related to displaying a specific immunodominance profile.


Asunto(s)
Anticuerpos Antivirales , Pruebas de Inhibición de Hemaglutinación , Glicoproteínas Hemaglutininas del Virus de la Influenza , Epítopos Inmunodominantes , Gripe Humana , Humanos , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Femenino , Masculino , Adulto , Epítopos Inmunodominantes/inmunología , Persona de Mediana Edad , Gripe Humana/inmunología , Gripe Humana/prevención & control , Adulto Joven , Factores de Edad , Factores Sexuales , Adolescente , Estudios de Cohortes , Anciano , Antígenos Virales/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre
15.
Appl Microbiol Biotechnol ; 108(1): 307, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38656587

RESUMEN

Surface plasmon resonance (SPR)-based biosensors have emerged as a powerful platform for bioprocess monitoring due to their ability to detect biointeractions in real time, without the need for labeling. Paramount for the development of a robust detection platform is the immobilization of a ligand with high specificity and affinity for the in-solution species of interest. Following the 2009 H1N1 pandemic, much effort has been made toward the development of quality control platforms for influenza A vaccine productions, many of which have employed SPR for detection. Due to the rapid antigenic drift of influenza's principal surface protein, hemagglutinin, antibodies used for immunoassays need to be produced seasonally. The production of these antibodies represents a 6-8-week delay in immunoassay and, thus, vaccine availability. This review focuses on SPR-based assays that do not rely on anti-HA antibodies for the detection, characterization, and quantification of influenza A in bioproductions and biological samples. KEY POINTS: • The single radial immunodiffusion assay (SRID) has been the gold standard for the quantification of influenza vaccines since 1979. Due to antigenic drift of influenza's hemagglutinin protein, new antibody reagents for the SRID assay must be produced each year, requiring 6-8 weeks. The resulting delay in immunoassay availability is a major bottleneck in the influenza vaccine pipeline. This review highlights ligand options for the detection and quantification of influenza viruses using surface plasmon resonance biosensors.


Asunto(s)
Vacunas contra la Influenza , Control de Calidad , Resonancia por Plasmón de Superficie , Resonancia por Plasmón de Superficie/métodos , Vacunas contra la Influenza/inmunología , Humanos , Anticuerpos Antivirales/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Gripe Humana/diagnóstico , Gripe Humana/prevención & control , Gripe Humana/inmunología , Inmunoensayo/métodos , Inmunoensayo/normas , Técnicas Biosensibles/métodos , Virus de la Influenza A/inmunología
16.
Immunity ; 57(5): 1141-1159.e11, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38670113

RESUMEN

Broadly neutralizing antibodies (bnAbs) targeting the hemagglutinin (HA) stem of influenza A viruses (IAVs) tend to be effective against either group 1 or group 2 viral diversity. In rarer cases, intergroup protective bnAbs can be generated by human antibody paratopes that accommodate the conserved glycan differences between the group 1 and group 2 stems. We applied germline-engaging nanoparticle immunogens to elicit a class of cross-group bnAbs from physiological precursor frequency within a humanized mouse model. Cross-group protection depended on the presence of the human bnAb precursors within the B cell repertoire, and the vaccine-expanded antibodies enriched for an N55T substitution in the CDRH2 loop, a hallmark of the bnAb class. Structurally, this single mutation introduced a flexible fulcrum to accommodate glycosylation differences and could alone enable cross-group protection. Thus, broad IAV immunity can be expanded from the germline repertoire via minimal antigenic input and an exceptionally simple antibody development pathway.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Virus de la Influenza A , Vacunas contra la Influenza , Infecciones por Orthomyxoviridae , Vacunación , Animales , Ratones , Humanos , Anticuerpos Antivirales/inmunología , Vacunas contra la Influenza/inmunología , Virus de la Influenza A/inmunología , Anticuerpos Neutralizantes/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Sustitución de Aminoácidos , Linfocitos B/inmunología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Anticuerpos ampliamente neutralizantes/inmunología
17.
Vaccine ; 42(15): 3410-3419, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38641498

RESUMEN

The application of recombinant herpesvirus of turkey, expressing the H9 hemagglutinin gene from low pathogenic avian influenza virus (LPAIV) H9N2 and the avian orthoavulavirus-1 (AOAV-1) (commonly known as Newcastle Disease virus (NDV)) fusion protein (F) as an rHVT-H9-F vaccine, is an alternative to currently used classical vaccines. This study investigated H9- and ND-specific humoral and mucosal responses, H9-specific cell-mediated immunity, and protection conferred by the rHVT-H9-F vaccine in specific pathogen-free (SPF) chickens. Vaccination elicited systemic NDV F- and AIV H9-specific antibody response but also local antibodies in eye wash fluid and oropharyngeal swabs. The ex vivo H9-specific stimulation of splenic and pulmonary T cells in the vaccinated group demonstrated the ability of vaccination to induce systemic and local cellular responses. The clinical protection against a challenge using a LPAIV H9N2 strain of the G1 lineage isolated in Morocco in 2016 was associated with a shorter duration of shedding along with reduced viral genome load in the upper respiratory tract and reduced cloacal shedding compared to unvaccinated controls.


Asunto(s)
Anticuerpos Antivirales , Pollos , Subtipo H9N2 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Aviar , Esparcimiento de Virus , Animales , Subtipo H9N2 del Virus de la Influenza A/inmunología , Subtipo H9N2 del Virus de la Influenza A/genética , Pollos/inmunología , Gripe Aviar/prevención & control , Gripe Aviar/inmunología , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Esparcimiento de Virus/inmunología , Organismos Libres de Patógenos Específicos , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/genética , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Inmunidad Celular , Herpesvirus Meleágrido 1/inmunología , Herpesvirus Meleágrido 1/genética , Vacunación/métodos , Inmunidad Humoral , Vectores Genéticos/inmunología , Inmunogenicidad Vacunal , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/administración & dosificación , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética
18.
Biochem Biophys Res Commun ; 711: 149919, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38608435

RESUMEN

Subunit vaccines are among the most useful vaccine modalities; however, their low immunogenicity necessitates the addition of adjuvants. Although adjuvants improve immune responses induced by vaccines, they often cause adverse reactions. To address this, we developed an adjuvant-free subunit vaccine platform that uses pre-existing antibodies generated from past infections or vaccinations as carriers for the delivery of vaccine antigens. Although we have confirmed the usefulness of this platform for nasal vaccines, its suitability as a parenterally injectable vaccine remains uncertain. Here, we verified the potential of our vaccine platform to harness pre-existing immunity for parenterally injectable vaccines. We generated RBD-HA by combining the receptor binding domain (RBD) derived from SARS-CoV-2 as a vaccine antigen with hemagglutinin (HA) sourced from influenza viruses to serve as the carrier protein. We revealed that subcutaneous vaccination with RBD-HA effectively triggered strong RBD-specific IgG responses in mice previously infected with the influenza A virus, even in the absence of adjuvants, and conferred protection to mice against SARS-CoV-2 upon challenge. Furthermore, we revealed that vaccination with RBD-HA did not induce an inflammatory response, such as inflammatory cytokine production, swelling, and recruitment of inflammatory immune cells, whereas conventional vaccines combined with adjuvants induced these adverse reactions. In addition, we demonstrated the remarkable versatility of this platform using a vaccine antigen derived from Streptococcus pneumoniae. These findings indicate the potential of this adjuvant-free vaccine platform to enhance the efficacy of parenterally injectable subunit vaccines and reduce adverse reactions.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Inmunoglobulina G , Ratones Endogámicos BALB C , SARS-CoV-2 , Animales , Inmunoglobulina G/inmunología , Inmunoglobulina G/sangre , Ratones , SARS-CoV-2/inmunología , COVID-19/prevención & control , COVID-19/inmunología , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/administración & dosificación , Humanos , Anticuerpos Antivirales/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación
19.
Int J Biol Macromol ; 267(Pt 2): 131458, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38593899

RESUMEN

Avian influenza virus (AIV) H7N9 diseases have been recently reported, raising concerns about a potential pandemic. Thus, there is an urgent need for effective therapeutics for AIV H7N9 infections. Herein, camelid immunization and yeast two-hybrid techniques were used to identify potent neutralizing nanobodies (Nbs) targeting the H7 subtype hemagglutinin. First, we evaluated the binding specificity and hemagglutination inhibition activity of the screened Nbs against the H7 subtype hemagglutinin. Nb-Z77, with high hemagglutination inhibition activity was selected from the screened Nbs to optimize the yeast expression conditions and construct oligomeric forms of Nb-Z77 using various ligation methods. The oligomers Nb-Z77-DiGS, Nb-Z77-TriGS, Nb-Z77-Fc and Nb-Z77-Foldon were successfully constructed and expressed. Nb-Z77-DiGS and Nb-Z77-Foldon exhibited considerably greater activity than did Nb-Z77 against H7 subtype hemagglutinin, with median effective concentrations of 384.7 and 27.33 pM and binding affinity values of 213 and 5.21 pM, respectively. Nb-Z77-DiGS and Nb-Z77-Foldon completely inhibited the hemagglutination activity of the inactivated virus H7-Re1 at the lowest concentration of 0.938 µg/mL. This study screened a strain of Nb with high hemagglutination inhibition activity and enhanced its antiviral activity through oligomerization, which may have great potential for developing effective agents for the prevention, diagnosis, and treatment of AIV H7 subtype infection.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza , Anticuerpos de Dominio Único , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/química , Animales , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H7N9 del Virus de la Influenza A/inmunología , Humanos , Pruebas de Inhibición de Hemaglutinación , Gripe Aviar/inmunología , Gripe Aviar/virología , Gripe Aviar/prevención & control , Anticuerpos Antivirales/inmunología , Anticuerpos Neutralizantes/inmunología
20.
mBio ; 15(5): e0074124, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38587427

RESUMEN

Outbreaks of acute respiratory viral diseases, such as influenza and COVID-19 caused by influenza A virus (IAV) and SARS-CoV-2, pose a serious threat to global public health, economic security, and social stability. This calls for the development of broad-spectrum antivirals to prevent or treat infection or co-infection of IAV and SARS-CoV-2. Hemagglutinin (HA) on IAV and spike (S) protein on SARS-CoV-2, which contain various types of glycans, play crucial roles in mediating viral entry into host cells. Therefore, they are key targets for the development of carbohydrate-binding protein-based antivirals. This study demonstrated that griffithsin (GRFT) and the GRFT-based bivalent entry inhibitor GL25E (GRFT-L25-EK1) showed broad-spectrum antiviral effects against IAV infection in vitro by binding to HA in a carbohydrate-dependent manner and effectively protected mice from lethal IAV infection. Although both GRFT and GL25E could inhibit infection of SARS-CoV-2 Omicron variants, GL25E proved to be significantly more effective than GRFT and EK1 alone. Furthermore, GL25E effectively inhibited in vitro co-infection of IAV and SARS-CoV-2 and demonstrated good druggability, including favorable safety and stability profiles. These findings suggest that GL25E is a promising candidate for further development as a broad-spectrum antiviral drug for the prevention and treatment of infection or co-infection from IAV and SARS-CoV-2.IMPORTANCEInfluenza and COVID-19 are highly contagious respiratory illnesses caused by the influenza A virus (IAV) and SARS-CoV-2, respectively. IAV and SARS-CoV-2 co-infection exacerbates damage to lung tissue and leads to more severe clinical symptoms, thus calling for the development of broad-spectrum antivirals for combating IAV and SARS-CoV-2 infection or co-infection. Here we found that griffithsin (GRFT), a carbohydrate-binding protein, and GL25E, a recombinant protein consisting of GRFT, a 25 amino acid linker, and EK1, a broad-spectrum coronavirus inhibitor, could effectively inhibit IAV and SARS-CoV-2 infection and co-infection by targeting glycans on HA of IAV and spike (S) protein of SARS-CoV-2. GL25E is more effective than GRFT because GL25E can also interact with the HR1 domain in SARS-CoV-2 S protein. Furthermore, GL25E possesses favorable safety and stability profiles, suggesting that it is a promising candidate for development as a drug to prevent and treat IAV and SARS-CoV-2 infection or co-infection.


Asunto(s)
Antivirales , COVID-19 , Coinfección , Virus de la Influenza A , Lectinas de Plantas , SARS-CoV-2 , Internalización del Virus , Animales , Antivirales/farmacología , Virus de la Influenza A/efectos de los fármacos , Ratones , SARS-CoV-2/efectos de los fármacos , Humanos , Internalización del Virus/efectos de los fármacos , Coinfección/tratamiento farmacológico , Coinfección/virología , Lectinas de Plantas/farmacología , COVID-19/virología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/virología , Tratamiento Farmacológico de COVID-19 , Perros , Ratones Endogámicos BALB C , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Células de Riñón Canino Madin Darby
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA