Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.660
Filtrar
1.
ACS Nano ; 18(20): 12905-12916, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38721835

RESUMO

For most frequent respiratory viruses, there is an urgent need for a universal influenza vaccine to provide cross-protection against intra- and heterosubtypes. We previously developed an Escherichia coli fusion protein expressed extracellular domain of matrix 2 (M2e) and nucleoprotein, named NM2e, and then combined it with an aluminum adjuvant, forming a universal vaccine. Although NM2e has demonstrated a protective effect against the influenza virus in mice to some extent, further improvement is still needed for the induction of immune responses ensuring adequate cross-protection against influenza. Herein, we fabricated a cationic solid lipid nanoadjuvant using poly(lactic acid) (PLA) and dimethyl-dioctadecyl-ammonium bromide (DDAB) and loaded NM2e to generate an NM2e@DDAB/PLA nanovaccine (Nv). In vitro experiments suggested that bone marrow-derived dendritic cells incubated with Nv exhibited ∼4-fold higher antigen (Ag) uptake than NM2e at 16 h along with efficient activation by NM2e@DDAB/PLA Nv. In vivo experiments revealed that Ag of the Nv group stayed in lymph nodes (LNs) for more than 14 days after initial immunization and DCs in LNs were evidently activated and matured. Furthermore, the Nv primed T and B cells for robust humoral and cellular immune responses after immunization. It also induced a ratio of IgG2a/IgG1 higher than that of NM2e to a considerable extent. Moreover, NM2e@DDAB/PLA Nv quickly restored body weight and improved survival of homo- and heterosubtype influenza challenged mice, and the cross-protection efficiency was over 90%. Collectively, our study demonstrated that NM2e@DDAB/PLA Nv could offer notable protection against homo- and heterosubtype influenza virus challenges, offering the potential for the development of a universal influenza vaccine.


Assuntos
Adjuvantes Imunológicos , Vacinas contra Influenza , Poliésteres , Compostos de Amônio Quaternário , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/química , Vacinas contra Influenza/administração & dosagem , Animais , Camundongos , Poliésteres/química , Adjuvantes Imunológicos/química , Adjuvantes Imunológicos/farmacologia , Compostos de Amônio Quaternário/química , Feminino , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/imunologia , Nanopartículas/química , Proteção Cruzada/imunologia , Adjuvantes de Vacinas/química , Proteínas da Matriz Viral/imunologia
2.
Front Immunol ; 15: 1381508, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38690272

RESUMO

Seasonal influenza remains a serious global health problem, leading to high mortality rates among the elderly and individuals with comorbidities. Vaccination is generally accepted as the most effective strategy for influenza prevention. While current influenza vaccines are effective, they still have limitations, including narrow specificity for certain serological variants, which may result in a mismatch between vaccine antigens and circulating strains. Additionally, the rapid variability of the virus poses challenges in providing extended protection beyond a single season. Therefore, mRNA technology is particularly promising for influenza prevention, as it enables the rapid development of multivalent vaccines and allows for quick updates of their antigenic composition. mRNA vaccines have already proven successful in preventing COVID-19 by eliciting rapid cellular and humoral immune responses. In this study, we present the development of a trivalent mRNA vaccine candidate, evaluate its immunogenicity using the hemagglutination inhibition assay, ELISA, and assess its efficacy in animals. We demonstrate the higher immunogenicity of the mRNA vaccine candidate compared to the inactivated split influenza vaccine and its enhanced ability to generate a cross-specific humoral immune response. These findings highlight the potential mRNA technology in overcoming current limitations of influenza vaccines and hold promise for ensuring greater efficacy in preventing seasonal influenza outbreaks.


Assuntos
Anticorpos Antivirais , Reações Cruzadas , Imunidade Humoral , Vacinas contra Influenza , Vacinas de mRNA , Vacinas contra Influenza/imunologia , Animais , Vacinas de mRNA/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Humanos , Reações Cruzadas/imunologia , Camundongos , Influenza Humana/prevenção & controle , Influenza Humana/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Feminino , Estações do Ano , Imunogenicidade da Vacina , SARS-CoV-2/imunologia , SARS-CoV-2/genética , Camundongos Endogâmicos BALB C , Vírus da Influenza A Subtipo H1N1/imunologia , COVID-19/prevenção & controle , COVID-19/imunologia , Vacinação
3.
Sci Transl Med ; 16(745): eadj4685, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38691617

RESUMO

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.


Assuntos
Furões , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Vacinas contra Influenza , Vacinação , Animais , Vacinas contra Influenza/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/imunologia , Camundongos , Anticorpos Antivirais/imunologia , Humanos , Influenza Humana/prevenção & controle , Influenza Humana/imunologia , Antígenos Virais/imunologia , Feminino , Camundongos Endogâmicos BALB C
4.
Front Immunol ; 15: 1370564, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38711520

RESUMO

There are considerable avenues through which currently licensed influenza vaccines could be optimized. We tested influenza vaccination in a mouse model with two adjuvants: Sendai virus-derived defective interfering (SDI) RNA, a RIG-I agonist; and an amphiphilic imidazoquinoline (IMDQ-PEG-Chol), a TLR7/8 agonist. The negatively charged SDI RNA was formulated into lipid nanoparticles (LNPs) facilitating direct delivery of SDI RNA to the cytosol, where RIG-I sensing induces inflammatory and type I interferon responses. We previously tested SDI RNA and IMDQ-PEG-Chol as standalone and combination adjuvants for influenza and SARS-CoV-2 vaccines. Here, we tested two different ionizable lipids, K-Ac7-Dsa and S-Ac7-Dog, for LNP formulations. The LNPs were incorporated with SDI RNA to determine its potential as a combination adjuvant with IMDQ-PEG-Chol by evaluating the host immune response to vaccination and infection in immunized BALB/c mice. Adjuvanticity of IMDQ-PEG-Chol with and without empty or SDI-loaded LNPs was validated with quadrivalent inactivated influenza vaccine (QIV), showing robust induction of antibody titers and T-cell responses. Depending on the adjuvant combination and LNP formulation, humoral and cellular vaccine responses could be tailored towards type 1 or type 2 host responses with specific cytokine profiles that correlated with the protective responses to viral infection. The extent of protection conferred by different vaccine/LNP/adjuvant combinations was tested by challenging mice with a vaccine-matched strain of influenza A virus A/Singapore/gp1908/2015 IVR-180 (H1N1). Groups that received either LNP formulated with SDI or IMDQ-PEG-Chol, or both, showed very low levels of viral replication in their lungs at 5 days post-infection (DPI). These studies provide evidence that the combination of vaccines with LNPs and/or adjuvants promote antigen-specific cellular responses that can contribute to protection upon infection. Interestingly, we observed differences in humoral and cellular responses to vaccination between different groups receiving K-Ac7-Dsa or S-Ac7-Dog lipids in LNP formulations. The differences were also reflected in inflammatory responses in lungs of vaccinated animals to infection, depending on LNP formulations. Therefore, this study suggests that the composition of the LNPs, particularly the ionizable lipid, plays an important role in inducing inflammatory responses in vivo, which is important for vaccine safety and to prevent adverse effects upon viral exposure.


Assuntos
Adjuvantes Imunológicos , Vacinas contra Influenza , Lipossomos , Camundongos Endogâmicos BALB C , Nanopartículas , Infecções por Orthomyxoviridae , Animais , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Camundongos , Adjuvantes Imunológicos/administração & dosagem , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/imunologia , Feminino , Lipídeos , Vacinação/métodos , Adjuvantes de Vacinas , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Modelos Animais de Doenças , Vírus Sendai/imunologia , Influenza Humana/prevenção & controle , Influenza Humana/imunologia
5.
Vaccine ; 42(15): 3505-3513, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38714444

RESUMO

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.


Assuntos
Adenoviridae , Administração Intranasal , Anticorpos Antivirais , Proteção Cruzada , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Vacinas contra Influenza , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae , Proteínas da Matriz Viral , Animais , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/genética , Proteínas da Matriz Viral/imunologia , Proteínas da Matriz Viral/genética , Adenoviridae/genética , Adenoviridae/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Camundongos , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/imunologia , Feminino , Vírus da Influenza A Subtipo H3N2/imunologia , Vírus da Influenza A Subtipo H3N2/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N1/genética , Eficácia de Vacinas , Nucleoproteínas/imunologia , Nucleoproteínas/genética , Proteínas do Core Viral/imunologia , Proteínas do Core Viral/genética , Injeções Intramusculares , Proteínas Viroporinas
6.
Antiviral Res ; 225: 105877, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38561077

RESUMO

The conventional inactivated split seasonal influenza vaccine offers low efficacy, particularly in the elderly and against antigenic variants. Here, to improve the efficacy of seasonal vaccination for the elderly population, we tested whether supplementing seasonal bivalent (H1N1 + H3N2) split (S) vaccine with M2 ectodomain repeat and multi-subtype consensus neuraminidase (NA) proteins (N1 NA + N2 NA + flu B NA) on a virus-like particle (NA-M2e) would induce enhanced cross-protection against different influenza viruses in aged mice. Immunization with split vaccine plus NA-M2e (S + NA-M2e) increased vaccine-specific IgG antibodies towards T-helper type 1 responses and hemagglutination inhibition titers. Aged mice with NA-M2e supplemented vaccination were protected against homologous and heterologous viruses at higher efficacies, as evidenced by preventing weight loss, lowering lung viral loads, inducing broadly cross-protective humoral immunity, and IFN-γ+ CD4 and CD8 T cell responses than those with seasonal vaccine. Overall, this study supports a new strategy of NA-M2e supplemented vaccination to enhance protection against homologous and antigenically different viruses in the elderly.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Idoso , Humanos , Camundongos , Animais , Infecções por Orthomyxoviridae/prevenção & controle , Neuraminidase , Vírus da Influenza A Subtipo H3N2 , Estações do Ano , Anticorpos Antivirais , Proteção Cruzada , Camundongos Endogâmicos BALB C
7.
Int J Biol Macromol ; 266(Pt 2): 131289, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38570002

RESUMO

Intranasal vaccination offers crucial protection against influenza virus pandemics. However, antigens, especially subunit antigens, often fail to induce effective immune responses without the help of immune adjuvants. Our research has demonstrated that a polyelectrolyte complex, composed of curdlan sulfate/O-(2-hydroxyl) propyl-3-trimethyl ammonium chitosan chloride (CS/O-HTCC), effectively triggers both mucosal and systemic immune responses when administrated intranasal. In this study, stable nanoparticles formed by curdlan-O-HTCC conjugate (CO NP) were prepared and characterized. Furthermore, the efficacy of CO NP was evaluated as a mucosal adjuvant in an intranasal influenza H1N1 subunit vaccine. The results revealed that CO NP exhibits uniform and spherical morphology, with a size of 190.53 ± 4.22 nm, and notably, it remains stable in PBS at 4 °C for up to 6 weeks. Biological evaluation demonstrated that CO NP stimulates the activation of antigen-presenting cells (APCs), including macrophages and dendritic cells (DCs), both in vitro and in vivo. Furthermore, intranasal administration of CO NP effectively elicits cellular and humoral immune responses, notably enhancing mucosal immunity. Thus, CO NP emerges as a promising mucosal adjuvant for influenza subunit vaccines.


Assuntos
Adjuvantes Imunológicos , Administração Intranasal , Quitosana , Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza , Nanopartículas , Vacinas de Subunidades Antigênicas , beta-Glucanas , Vírus da Influenza A Subtipo H1N1/imunologia , Quitosana/química , Nanopartículas/química , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/química , Vacinas contra Influenza/administração & dosagem , beta-Glucanas/química , beta-Glucanas/farmacologia , beta-Glucanas/administração & dosagem , Animais , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/química , Adjuvantes Imunológicos/administração & dosagem , Camundongos , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Imunidade nas Mucosas/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Feminino , Células Dendríticas/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/imunologia
8.
Open Vet J ; 14(1): 350-359, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38633177

RESUMO

Background: Equine influenza (EI) is a transmissible viral respiratory sickness of the Equidae family. Two viruses, H7N7 and H3N8 caused EI; however, H7N7 has not been detected for decades. H3N8 has circulated and bifurcated into Eurasian and American lineages. The latter subsequently diversified into Kentucky, South America, and Florida sub-lineages. Florida clade 1 (FC1) and Florida clade 2 (FC2) strains are the only circulating EI viruses (EIVs) in the meantime. Immunization is considered the major means for the prevention and control of EI infection. Using disparate technologies and platforms, several vaccines have been developed and commercialized. According to the recommendations of the World Organization for Animal Health (WOAH), all commercial vaccines shall comprise representatives of both FC1 and FC2 strains. Unfortunately, most of the commercially available vaccines were not updated to incorporate a representative of FC2 strains. Aim: The purpose of this research was to develop a new EI vaccine candidate that incorporates the hemagglutinin (HA) antigen from the currently circulating FC2. Methods: In this study, we report the expression of the full-length recombinant HA gene of FC2 in the baculovirus expression system. Results: The HA recombinant protein has been proven to maintain its biological characteristics by hemadsorption (HAD) and hemagglutination tests. Moreover, using a reference-specific serum, the specificity of the HA has been confirmed through the implementation of immunoperoxidase and western immunoblotting assays. Conclusion: In conclusion, we report the expression of specific biologically active recombinant HA of FC2, which would act as a foundation for the generation of an updated EI subunit or virus vector vaccine candidates.


Assuntos
Vírus da Influenza A Subtipo H3N8 , Vírus da Influenza A Subtipo H7N7 , Infecções por Orthomyxoviridae , Vacinas , Cavalos , Animais , Hemaglutininas , Vírus da Influenza A Subtipo H3N8/genética , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/veterinária , Baculoviridae
9.
J Agric Food Chem ; 72(17): 9782-9794, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38597360

RESUMO

Uncontrolled inflammation contributes significantly to the mortality in acute respiratory infections. Our previous research has demonstrated that maize bran feruloylated oligosaccharides (FOs) possess notable anti-inflammatory properties linked to the NF-kB pathway regulation. In this study, we clarified that the oral administration of FOs moderately inhibited H1N1 virus infection and reduced lung inflammation in influenza-infected mice by decreasing a wide spectrum of cytokines (IFN-α, IFN-ß, IL-6, IL-10, and IL-23) in the lungs. The mechanism involves FOs suppressing the transduction of the RIG-I/MAVS/TRAF3 signaling pathway, subsequently lowering the expression of NF-κB. In silico analysis suggests that FOs have a greater binding affinity for the RIG-I/MAVS signaling complex. This indicates that FOs have potential as promising targets for immune modulation. Moreover, in MAVS knockout mice, we confirmed that the anti-inflammatory function of FOs against influenza depends on MAVS. Comprehensive analysis using 16S rRNA gene sequencing and metabolite profiling techniques showed that FOs have the potential to restore immunity by modulating the gut microbiota. In conclusion, our study demonstrates that FOs are effective anti-inflammatory phytochemicals in inhibiting lung inflammation caused by influenza. This suggests that FOs could serve as a potential nutritional strategy for preventing the H1N1 virus infection and associated lung inflammation.


Assuntos
Proteína DEAD-box 58 , Vírus da Influenza A Subtipo H1N1 , Influenza Humana , Camundongos Knockout , Oligossacarídeos , Infecções por Orthomyxoviridae , Transdução de Sinais , Fator 3 Associado a Receptor de TNF , Animais , Camundongos , Oligossacarídeos/administração & dosagem , Oligossacarídeos/química , Oligossacarídeos/farmacologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/metabolismo , Vírus da Influenza A Subtipo H1N1/imunologia , Humanos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Influenza Humana/metabolismo , Fator 3 Associado a Receptor de TNF/genética , Fator 3 Associado a Receptor de TNF/metabolismo , Fator 3 Associado a Receptor de TNF/imunologia , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/metabolismo , Proteína DEAD-box 58/imunologia , Pneumonia/imunologia , Pneumonia/prevenção & controle , Pneumonia/metabolismo , Pneumonia/virologia , Camundongos Endogâmicos C57BL , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/virologia , Citocinas/metabolismo , Citocinas/imunologia , Citocinas/genética , Feminino , NF-kappa B/imunologia , NF-kappa B/genética , NF-kappa B/metabolismo , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/farmacologia
10.
J Med Virol ; 96(4): e29605, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38634474

RESUMO

Interferon lambda (IFNλ), classified as a type III IFN, is a representative cytokine that plays an important role in innate immunity along with type I IFN. IFNλ can elicit antiviral states by inducing peculiar sets of IFN-stimulated genes (ISGs). In this study, an adenoviral vector expression system with a tetracycline operator system was used to express human IFNλ4 in cells and mice. The formation of recombinant adenovirus (rAd-huIFNλ4) was confirmed using immunohistochemistry assays and transmission electron microscopy. Its purity was verified by quantifying host cell DNA and host cell proteins, as well as by confirming the absence of the replication-competent adenovirus. The transduction of rAd-huIFNλ4 induced ISGs and inhibited four subtypes of the influenza virus in both mouse-derived (LA-4) and human-derived cells (A549). The antiviral state was confirmed in BALB/c mice following intranasal inoculation with 109 PFU of rAd-huIFNλ4, which led to the inhibition of four subtypes of the influenza virus in mouse lungs, with reduced inflammatory lesions. These results imply that human IFNλ4 could induce antiviral status by modulating ISG expression in mice.


Assuntos
Antivirais , Influenza Humana , Interferon lambda , Orthomyxoviridae , Animais , Humanos , Camundongos , Antivirais/farmacologia , Imunidade Inata , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Interferon lambda/metabolismo , Interferon lambda/farmacologia , Interferon Tipo I/genética , Interferons/metabolismo , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Vetores Genéticos
12.
Immunity ; 57(5): 1141-1159.e11, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38670113

RESUMO

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.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Vírus da Influenza A , Vacinas contra Influenza , Infecções por Orthomyxoviridae , Vacinação , Animais , Camundongos , Humanos , Anticorpos Antivirais/imunologia , Vacinas contra Influenza/imunologia , Vírus da Influenza A/imunologia , Anticorpos Neutralizantes/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Substituição de Aminoácidos , Linfócitos B/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Anticorpos Amplamente Neutralizantes/imunologia
13.
Virology ; 595: 110097, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38685171

RESUMO

Current influenza vaccine is not effective in providing cross-protection against variants. We evaluated the immunogenicity and efficacy of multi-subtype neuraminidase (NA) and M2 ectodomain virus-like particle (m-cNA-M2e VLP) and chimeric M2e-H3 stalk protein vaccines (M2e-H3 stalk) in ferrets. Our results showed that ferrets with recombinant m-cNA-M2e VLP or M2e-H3 stalk vaccination induced multi-vaccine antigen specific IgG antibodies (M2e, H3 stalk, NA), NA inhibition, antibody-secreting cells, and IFN-γ secreting cell responses. Ferrets immunized with either m-cNA-M2e VLP or M2e-H3 stalk vaccine were protected from H1N1 and H3N2 influenza viruses by lowering viral titers in nasal washes, trachea, and lungs after challenge. Vaccinated ferret antisera conferred broad humoral immunity in naïve mice. Our findings provide evidence that immunity to M2e and HA-stalk or M2e plus multi-subtype NA proteins induces cross-protection in ferrets.


Assuntos
Anticorpos Antivirais , Proteção Cruzada , Furões , Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A Subtipo H3N2 , Vacinas contra Influenza , Neuraminidase , Infecções por Orthomyxoviridae , Vacinas de Partículas Semelhantes a Vírus , Animais , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Proteção Cruzada/imunologia , Anticorpos Antivirais/imunologia , Neuraminidase/imunologia , Neuraminidase/genética , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Camundongos , Proteínas da Matriz Viral/imunologia , Proteínas da Matriz Viral/genética , Feminino , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Proteínas Viroporinas , Proteínas Virais
14.
Carbohydr Polym ; 336: 122080, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38670772

RESUMO

Traditional Chinese medicine polysaccharides have numerous biological activities with broad applications in the biomedical industries. However, a clear understanding of the pharmacological activities of compound polysaccharides with multi-component structures remain challenging. This study aimed to investigate the immune boosting effect of compound polysaccharides on the influenza vaccine and assess the preliminary structure-activity relationship. The compound polysaccharide (CP) was isolated from the combined Chinese herbs lentinan, pachymaran and tremellan, and purified by gradient ethanol precipitation to obtain its subcomponents of CP-20, CP-40, CP-60, and CP-80 with decreasing molecular weights. These polysaccharides were mainly composed of glucans with different linkage patterns, including α-(1 â†’ 3)-glucan, α-(1 â†’ 4)-glucan and ß-(1 â†’ 6)-glucan. A significant improvement was observed in the survival of mice vaccinated with inactivated (IAV) vaccine and the isolated polysaccharides as adjuvants. A reduction in the pulmonary virus titer and weight loss were also observed. Moreover, CP-40 and CP-60, as well as the original CP, significantly enhanced the serum anti-IAV antibody titers and interleukin IL-2, IL-5, and IL-6 concentrations. These preliminary results indicate the immune boosting effect of the compound polysaccharides is highly relevant to the specific structural properties of the subcomponent, and CP-40 is worthy of further exploration as a glycan adjuvant for the IAV vaccine.


Assuntos
Adjuvantes Imunológicos , Vacinas contra Influenza , Camundongos Endogâmicos BALB C , Polissacarídeos , Vacinas de Produtos Inativados , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/química , Vacinas contra Influenza/farmacologia , Animais , Vacinas de Produtos Inativados/imunologia , Camundongos , Polissacarídeos/farmacologia , Polissacarídeos/química , Polissacarídeos/isolamento & purificação , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/química , Feminino , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/imunologia , Citocinas/metabolismo
15.
ACS Infect Dis ; 10(5): 1552-1560, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38623820

RESUMO

Tyrosine cross-linking has recently been used to produce nanoclusters (NCs) from peptides to enhance their immunogenicity. In this study, NCs were generated using the ectodomain of the ion channel Matrix 2 (M2e) protein, a conserved influenza surface antigen. The NCs were administered via intranasal (IN) or intramuscular (IM) routes in a mouse model in a prime-boost regimen in the presence of the adjuvant CpG. After boost, a significant increase in anti-M2e IgG and its subtypes was observed in the serum and lungs of mice vaccinated through the IM and IN routes; however, significant enhancement in anti-M2e IgA in lungs was observed only in the IN group. Analysis of cytokine concentrations in stimulated splenocyte cultures indicated a Th1/Th17-biased response. Mice were challenged with a lethal dose of A/California/07/2009 (H1N1pdm), A/Puerto Rico/08/1934 (H1N1), or A/Hong Kong/08/1968 (H3N2) strains. Mice that received M2e NCs + CpG were significantly protected against these strains and showed decreased lung viral titers compared with the naive mice and M2e NC-alone groups. The IN-vaccinated group showed superior protection against the H3N2 strain as compared to the IM group. This research extends our earlier efforts involving the tyrosine-based cross-linking method and highlights the potential of this technology in enhancing the immunogenicity of short peptide immunogens.


Assuntos
Anticorpos Antivirais , Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza , Infecções por Orthomyxoviridae , Tirosina , Animais , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Camundongos , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/imunologia , Tirosina/química , Tirosina/farmacologia , Vírus da Influenza A Subtipo H1N1/imunologia , Feminino , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Proteínas da Matriz Viral/imunologia , Proteínas da Matriz Viral/genética , Camundongos Endogâmicos BALB C , Vírus da Influenza A Subtipo H3N2/imunologia , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Pulmão/virologia , Pulmão/imunologia , Administração Intranasal , Injeções Intramusculares , Citocinas , Proteção Cruzada , Proteínas Viroporinas
16.
Vaccine ; 42(11): 2770-2780, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38508930

RESUMO

The COVID-19 pandemic has highlighted the need for mucosal vaccines as breakthrough infections, short-lived immune responses and emergence of new variants have challenged the efficacy provided by the first generation of vaccines against SARS-CoV-2 viruses. M2SR SARS-CoV-2, an M2-deleted single-replication influenza virus vector modified to encode the SARS-CoV-2 receptor binding domain, was evaluated following intranasal delivery in a hamster challenge model for protection against Wuhan SARS-CoV-2. An adjuvanted inactivated SARS-CoV-2 whole virus vaccine administered intramuscularly was also evaluated. The intranasal M2SR SARS-CoV-2 was more effective than the intramuscular adjuvanted inactivated whole virus vaccine in providing protection against SARS-CoV-2 challenge. M2SR SARS-CoV-2 elicited neutralizing serum antibodies against Wuhan and Omicron SARS-CoV-2 viruses in addition to cross-reactive mucosal antibodies. Furthermore, M2SR SARS-CoV-2 generated serum HAI and mucosal antibody responses against influenza similar to an H3N2 M2SR influenza vaccine. The intranasal dual influenza/COVID M2SR SARS-CoV-2 vaccine has the potential to provide protection against both influenza and COVID.


Assuntos
COVID-19 , Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Humanos , Cricetinae , Influenza Humana/prevenção & controle , Vacinas contra COVID-19 , SARS-CoV-2 , Infecções por Orthomyxoviridae/prevenção & controle , Vírus da Influenza A Subtipo H3N2 , Pandemias/prevenção & controle , Anticorpos Antivirais , COVID-19/prevenção & controle , Vacinação , Anticorpos Neutralizantes , Adjuvantes Imunológicos
17.
Front Immunol ; 15: 1322879, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38482020

RESUMO

Orf virus (ORFV) is a large DNA virus that can harbor and efficiently deliver viral antigens in swine. Here we used ORFV as a vector platform to deliver chimeric hemagglutinins (HA) of Influenza A virus of swine (IAV-S). Vaccine development against IAV-S faces limitations posed by strain-specific immunity and the antigenic diversity of the IAV-S strains circulating in the field. A promising alternative aiming at re-directing immune responses on conserved epitopes of the stalk segment of the hemagglutinin (HA2) has recently emerged. Sequential immunization with chimeric HAs comprising the same stalk but distinct exotic head domains can potentially induce cross-reactive immune responses against conserved epitopes of the HA2 while breaking the immunodominance of the head domain (HA1). Here, we generated two recombinant ORFVs expressing chimeric HAs encoding the stalk region of a contemporary H1N1 IAV-S strain and exotic heads derived from either H6 or H8 subtypes, ORFVΔ121cH6/1 and ORFVΔ121cH8/1, respectively. The resulting recombinant viruses were able to express the heterologous protein in vitro. Further, the immunogenicity and cross-protection of these vaccine candidates were assessed in swine after sequential intramuscular immunization with OV-cH6/1 and OV-cH8/1, and subsequent challenge with divergent IAV-S strains. Humoral responses showed that vaccinated piglets presented increasing IgG responses in sera. Additionally, cross-reactive IgG and IgA antibody responses elicited by immunization were detected in sera and bronchoalveolar lavage (BAL), respectively, by ELISA against different viral clades and a diverse range of contemporary H1N1 IAV-S strains, indicating induction of humoral and mucosal immunity in vaccinated animals. Importantly, viral shedding was reduced in nasal swabs from vaccinated piglets after intranasal challenge with either Oh07 (gamma clade) or Ca09 (npdm clade) IAV-S strains. These results demonstrated the efficiency of ORFV-based vectors in delivering chimeric IAV-S HA-based vaccine candidates and underline the potential use of chimeric-HAs for prevention and control of influenza in swine.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza , Vírus do Orf , Infecções por Orthomyxoviridae , Animais , Suínos , Hemaglutininas/genética , Infecções por Orthomyxoviridae/prevenção & controle , Vírus da Influenza A Subtipo H1N1/genética , Anticorpos Antivirais , Imunoglobulina G , Epitopos
18.
Vet Immunol Immunopathol ; 271: 110743, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38522410

RESUMO

Equine influenza is a contagious respiratory disease caused by H3N8 type A influenza virus. Vaccination against equine influenza is conducted regularly; however, infection still occurs globally because of the short immunity duration and suboptimal efficacy of current vaccines. Hence the objective of this study was to investigate whether an adjuvant combination can improve immune responses to equine influenza virus (EIV) vaccines. Seventy-two mice were immunized with an EIV vaccine only or with monophosphoryl lipid A (MPL), polyinosinic-polycytidylic acid (Poly I:C), or MPL + Poly I:C. Prime immunization was followed by boost immunization after 2 weeks. Mice were euthanized at 4, 8, and 32 weeks post-prime immunization, respectively. Sera were collected to determine humoral response. Bone marrow, spleen, and lung samples were harvested to determine memory cell responses, antigen-specific T-cell proliferation, and lung viral titers. MPL + Poly I:C resulted in the highest IgG, IgG1, and IgG2a antibodies and hemagglutination inhibition titers among the groups and sustained their levels until 32 weeks post-prime immunization. The combination enhanced memory B cell responses in the bone marrow and spleen. At 8 weeks post-prime immunization, the combination induced higher CD8+ central memory T cell frequencies in the lungs and CD8+ central memory T cells in the spleen. In addition, the combination group exhibited enhanced antigen-specific T cell proliferation, except for CD4+ T cells in the lungs. Our results demonstrated improved immune responses when using MPL + Poly I:C in EIV vaccines by inducing enhanced humoral responses, memory cell responses, and antigen-specific T cell proliferation.


Assuntos
Adjuvantes Imunológicos , Vírus da Influenza A Subtipo H3N8 , Vacinas contra Influenza , Lipídeo A , Lipídeo A/análogos & derivados , Infecções por Orthomyxoviridae , Poli I-C , Animais , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Poli I-C/farmacologia , Poli I-C/administração & dosagem , Lipídeo A/farmacologia , Lipídeo A/administração & dosagem , Lipídeo A/imunologia , Camundongos , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/veterinária , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/farmacologia , Feminino , Vírus da Influenza A Subtipo H3N8/imunologia , Anticorpos Antivirais/sangue , Cavalos/imunologia , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/prevenção & controle , Doenças dos Cavalos/virologia , Imunoglobulina G/sangue , Memória Imunológica
19.
J Control Release ; 368: 275-289, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38382812

RESUMO

Virus like particles (VLPs) have been well recognized as one of the most important vaccine platforms due to their structural similarity to natural viruses to induce effective humoral and cellular immune responses. Nevertheless, lack of viral nucleic acids in VLPs usually leads the vaccine candidates less efficient in provoking innate immune against viral infection. Here, we constructed a biomimetic dual antigen hybrid influenza nanovaccines THM-HA@Mn with robust immunogenicity via in situ synthesizing a stimulator of interferon genes (STING) agonist Mn3O4 inside the cavity of a recombinant Hepatitis B core antigen VLP (HBc VLP) having fused SpyTag and influenza M2e antigen peptides (Tag-HBc-M2e, THM for short), followed by conjugating a recombinant hemagglutinin (rHA) antigen on the surface of the nanoparticles through SpyTag/SpyCatcher ligating. Such inside Mn3O4 immunostimulator-outside rHA antigen design, together with the chimeric M2e antigen on the HBc skeleton, enabled the synthesized hybrid nanovaccines THM-HA@Mn to well imitate the spatial distribution of M2e/HA antigens and immunostimulant in natural influenza virus. In vitro cellular experiments indicated that compared with the THM-HA antigen without Mn3O4 and a mixture vaccine consisting of THM-HA + MnOx, the THM-HA@Mn hybrid nanovaccines showed the highest efficacies in dendritic cells uptake and in promoting BMDC maturation, as well as inducing expression of TNF-α and type I interferon IFN-ß. The THM-HA@Mn also displayed the most sustained antigen release at the injection site, the highest efficacies in promoting the DC maturation in lymph nodes and germinal center B cells activation in the spleen of the immunized mice. The co-delivery of immunostimulant and antigens enabled the THM-HA@Mn nanovaccines to induce the highest systemic antigen-specific antibody responses and cellular immunogenicity in mice. Together with the excellent colloid dispersion stability, low cytotoxicity, as well as good biosafety, the synthetic hybrid nanovaccines presented in this study offers a promising strategy to design VLP-based vaccine with robust natural and adaptive immunogenicity against emerging viral pathogens.


Assuntos
Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Vacinas de Partículas Semelhantes a Vírus , Animais , Camundongos , Humanos , Influenza Humana/prevenção & controle , Vacinas de Partículas Semelhantes a Vírus/genética , Imunidade Celular , Adjuvantes Imunológicos , Camundongos Endogâmicos BALB C , Anticorpos Antivirais , Infecções por Orthomyxoviridae/prevenção & controle
20.
Nat Commun ; 15(1): 850, 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38346952

RESUMO

Conserved epitopes shared between virus subtypes are often subdominant, making it difficult to induce broadly reactive antibodies by immunization. Here, we generate a plasmid DNA mix vaccine that encodes protein heterodimers with sixteen different influenza A virus hemagglutinins (HA) representing all HA subtypes except H1 (group 1) and H7 (group 2). Each single heterodimer expresses two different HA subtypes and is targeted to MHC class II on antigen presenting cells (APC). Female mice immunized with the plasmid mix produce antibodies not only against the 16 HA subtypes, but also against non-included H1 and H7. We demonstrate that individual antibody molecules cross-react between different HAs. Furthermore, the mix vaccine induces T cell responses to conserved HA epitopes. Immunized mice are partially protected against H1 viruses. The results show that application of valency-based immuno-selection to diversified antigens can be used to direct antibody responses towards conserved (subdominant) epitopes on viral antigens.


Assuntos
Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Feminino , Camundongos , Animais , Humanos , Influenza Humana/prevenção & controle , Hemaglutininas , Anticorpos Antivirais , Epitopos , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Infecções por Orthomyxoviridae/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA