Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.233
Filtrar
1.
Age Ageing ; 53(Supplement_2): ii70-ii79, 2024 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-38745493

RESUMO

This systematic review evaluated the impact of oral probiotics on the immune response to vaccination in older people. A literature search was performed in three electronic databases up to January 2023. Randomised controlled trials (RCTs) conducted in older people (age ≥ 60 years) investigating oral probiotics and vaccine response outcomes were included. Characteristics and outcome data of the included studies were extracted and analysed and study quality was assessed using the Cochrane Risk of Bias Tool for randomised trials. Ten RCTs involving 1,560 participants, reported in 9 papers, were included. Nine studies involved the seasonal influenza vaccine and one a COVID-19 vaccine. All studies used lactobacilli, some in combination with bifidobacteria. Studies reported outcomes including anti-vaccine antibody titres or concentrations, seroconversion and seroprotection. When comparing antibody titres, seroprotection rate and seroconversion rate between probiotic and placebo groups expressed as a response ratio, the weighted mean values were 1.29, 1.16 and 2.00, respectively. Meta-analysis showed that probiotics increase seroconversion rates to all three strains of the seasonal influenza vaccine: odds ratio (95% confidence interval) 2.74 (1.31, 5.70; P = 0.007) for the H1N1 strain; 1.90 (1.04, 3.44; P = 0.04) for the H3N2 strain; 1.72 (1.05, 2.80; P = 0.03) for the B strain. There was a low level of heterogeneity in these findings. Several studies were at high risk of bias due to missing outcome data. Lactobacilli may improve the vaccine response, but further research is needed to be more certain of this.


Assuntos
Vacinas contra Influenza , Probióticos , Ensaios Clínicos Controlados Aleatórios como Assunto , Humanos , Probióticos/uso terapêutico , Probióticos/administração & dosagem , Idoso , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Administração Oral , Vacinas contra COVID-19/administração & dosagem , Vacinas contra COVID-19/imunologia , Vacinação/métodos , Pessoa de Meia-Idade , COVID-19/prevenção & controle , COVID-19/imunologia , Influenza Humana/prevenção & controle , Influenza Humana/imunologia , SARS-CoV-2/imunologia
2.
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
3.
Nat Commun ; 15(1): 3666, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38693120

RESUMO

Respiratory viral infection increases host susceptibility to secondary bacterial infections, yet the precise dynamics within airway epithelia remain elusive. Here, we elucidate the pivotal role of CD47 in the airway epithelium during bacterial super-infection. We demonstrated that upon influenza virus infection, CD47 expression was upregulated and localized on the apical surface of ciliated cells within primary human nasal or bronchial epithelial cells. This induced CD47 exposure provided attachment sites for Staphylococcus aureus, thereby compromising the epithelial barrier integrity. Through bacterial adhesion assays and in vitro pull-down assays, we identified fibronectin-binding proteins (FnBP) of S. aureus as a key component that binds to CD47. Furthermore, we found that ciliated cell-specific CD47 deficiency or neutralizing antibody-mediated CD47 inactivation enhanced in vivo survival rates. These findings suggest that interfering with the interaction between airway epithelial CD47 and pathogenic bacterial FnBP holds promise for alleviating the adverse effects of super-infection.


Assuntos
Antígeno CD47 , Células Epiteliais , Infecções Estafilocócicas , Staphylococcus aureus , Superinfecção , Antígeno CD47/metabolismo , Antígeno CD47/genética , Humanos , Animais , Superinfecção/microbiologia , Camundongos , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/virologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Influenza Humana/metabolismo , Influenza Humana/imunologia , Influenza Humana/virologia , Aderência Bacteriana , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia , Mucosa Respiratória/virologia , Camundongos Endogâmicos C57BL , Brônquios/metabolismo , Brônquios/citologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Camundongos Knockout , Vírus da Influenza A Subtipo H1N1
4.
J Med Virol ; 96(5): e29657, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38727035

RESUMO

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.


Assuntos
Antígenos Virais , Vírus da Influenza A Subtipo H1N1 , Influenza Humana , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Humanos , Influenza Humana/epidemiologia , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Influenza Humana/imunologia , Antígenos Virais/genética , Antígenos Virais/imunologia , Aprendizado de Máquina , Evolução Molecular , Epitopos/genética , Epitopos/imunologia , COVID-19/epidemiologia , COVID-19/prevenção & controle , COVID-19/virologia , COVID-19/imunologia , Pandemias/prevenção & controle , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , SARS-CoV-2/genética , SARS-CoV-2/imunologia
5.
Nat Commun ; 15(1): 3833, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38714654

RESUMO

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.


Assuntos
Antígenos Virais , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Vírus da Influenza A Subtipo H3N2 , Influenza Humana , Aprendizado de Máquina , Estações do Ano , Vírus da Influenza A Subtipo H3N2/imunologia , Vírus da Influenza A Subtipo H3N2/genética , Humanos , Influenza Humana/imunologia , Influenza Humana/virologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Antígenos Virais/imunologia , Antígenos Virais/genética , Testes de Inibição da Hemaglutinação , Variação Antigênica/genética , Vacinas contra Influenza/imunologia
6.
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
7.
Front Immunol ; 15: 1352022, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38698856

RESUMO

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.


Assuntos
Fator H do Complemento , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Vírus da Influenza A , Influenza Humana , Ligação Proteica , Humanos , Fator H do Complemento/metabolismo , Fator H do Complemento/imunologia , Animais , Influenza Humana/imunologia , Influenza Humana/virologia , Influenza Humana/metabolismo , Vírus da Influenza A/imunologia , Vírus da Influenza A/fisiologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Sítios de Ligação , Influenza Aviária/virologia , Influenza Aviária/imunologia , Influenza Aviária/metabolismo , Aves/virologia , Interações Hospedeiro-Patógeno/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vírus da Influenza A Subtipo H9N2/imunologia
8.
Acta Biochim Pol ; 71: 12289, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38721309

RESUMO

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.


Assuntos
Anticorpos Antivirais , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A Subtipo H3N2 , Influenza Humana , Humanos , Polônia/epidemiologia , Adulto , Pessoa de Meia-Idade , Adolescente , Influenza Humana/imunologia , Influenza Humana/epidemiologia , Influenza Humana/sangue , Influenza Humana/virologia , Criança , Idoso , Pré-Escolar , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Adulto Jovem , Lactente , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Masculino , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Feminino , Recém-Nascido , Testes de Inibição da Hemaglutinação , Vírus da Influenza B/imunologia , Estações do Ano , Epidemias , Prevalência
9.
Immunity ; 57(5): 1160-1176.e7, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38697118

RESUMO

Multimodal single-cell profiling methods can capture immune cell variations unfolding over time at the molecular, cellular, and population levels. Transforming these data into biological insights remains challenging. Here, we introduce a framework to integrate variations at the human population and single-cell levels in vaccination responses. Comparing responses following AS03-adjuvanted versus unadjuvanted influenza vaccines with CITE-seq revealed AS03-specific early (day 1) response phenotypes, including a B cell signature of elevated germinal center competition. A correlated network of cell-type-specific transcriptional states defined the baseline immune status associated with high antibody responders to the unadjuvanted vaccine. Certain innate subsets in the network appeared "naturally adjuvanted," with transcriptional states resembling those induced uniquely by AS03-adjuvanted vaccination. Consistently, CD14+ monocytes from high responders at baseline had elevated phospho-signaling responses to lipopolysaccharide stimulation. Our findings link baseline immune setpoints to early vaccine responses, with positive implications for adjuvant development and immune response engineering.


Assuntos
Linfócitos B , Vacinas contra Influenza , Análise de Célula Única , Humanos , Vacinas contra Influenza/imunologia , Linfócitos B/imunologia , Centro Germinativo/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Vacinação , Anticorpos Antivirais/imunologia , Adjuvantes Imunológicos , Adjuvantes de Vacinas , Monócitos/imunologia , Polissorbatos , Esqualeno/imunologia , Imunidade Inata/imunologia
10.
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
11.
Immunity ; 57(5): 927-929, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38749392

RESUMO

Humans do not respond equally to vaccination. To investigate why, Mulè et al. developed a multimodal framework and found that high responders after unadjuvanted influenza vaccination exist in a naturally adjuvanted state, mimicking innate immunophenotypes following AS03-adjuvanted vaccination. This highlights biological factors that set apart high-antibody responders and how adjuvants can boost innate immune cues to improve humoral immunity.


Assuntos
Imunidade Inata , Vacinas contra Influenza , Humanos , Vacinas contra Influenza/imunologia , Imunidade Inata/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Vacinação , Adjuvantes Imunológicos , Imunidade Humoral , Adjuvantes de Vacinas , Anticorpos Antivirais/imunologia , Animais
12.
J Med Virol ; 96(5): e29678, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38751128

RESUMO

Death due to severe influenza is usually a fatal complication of a dysregulated immune response more than the acute virulence of an infectious agent. Although spleen tyrosine kinase (SYK) as a critical immune signaling molecule and therapeutic target plays roles in airway inflammation and acute lung injury, the role of SYK in influenza virus infection is not clear. Here, we investigated the antiviral and anti-inflammatory effects of SYK inhibitor R406 on influenza infection through a coculture model of human alveolar epithelial (A549) and macrophage (THP-1) cell lines and mouse model. The results showed that R406 treatment increased the viability of A549 and decreased the pathogenicity and mortality of lethal influenza virus in mice with influenza A infection, decreased levels of intracellular signaling molecules under the condition of inflammation during influenza virus infection. Combination therapy with oseltamivir further ameliorated histopathological damage in the lungs of mice and further delayed the initial time to death compared with R406 treatment alone. This study demonstrated that phosphorylation of SYK is involved in the pathogenesis of influenza, and R406 has antiviral and anti-inflammatory effects on the treatment of the disease, which may be realized through multiple pathways, including the already reported SYK/STAT/IFNs-mediated antiviral pathway, as well as TNF-α/SYK- and SYK/Akt-based immunomodulation pathway.


Assuntos
Anti-Inflamatórios , Antivirais , Modelos Animais de Doenças , Infecções por Orthomyxoviridae , Oxazinas , Quinase Syk , Animais , Humanos , Quinase Syk/antagonistas & inibidores , Camundongos , Antivirais/farmacologia , Antivirais/uso terapêutico , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/imunologia , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Oxazinas/farmacologia , Oxazinas/uso terapêutico , Piridinas/farmacologia , Piridinas/uso terapêutico , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Pulmão/patologia , Pulmão/virologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Células A549 , Vírus da Influenza A/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Oseltamivir/farmacologia , Oseltamivir/uso terapêutico , Influenza Humana/tratamento farmacológico , Influenza Humana/imunologia , Células THP-1 , Feminino , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico
13.
J Virol ; 98(5): e0041124, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38567952

RESUMO

Influenza A virus infection activates the NLRP3 inflammasome, a multiprotein signaling complex responsible for the proteolytic activation and release of the proinflammatory cytokine IL-1ß from monocytes and macrophages. Some influenza A virus (IAV) strains encode a short 90-amino acid peptide (PB1-F2) on an alternative open reading frame of segment 2, with immunomodulatory activity. We recently demonstrated that contemporary IAV PB1-F2 inhibits the activation of NLRP3, potentially by NEK7-dependent activation. PB1-F2 binds to NLRP3 with its C-terminal 50 amino acids, but the exact binding motif was unknown. On the NLRP3 side, the interface is formed through the leucine-rich-repeat (LRR) domain, potentially in conjunction with the pyrin domain. Here, we took advantage of PB1-F2 sequences from IAV strains with either weak or strong NLRP3 interaction. Sequence comparison and structure prediction using Alphafold2 identified a short four amino acid sequence motif (TQGS) in PB1-F2 that defines NLRP3-LRR binding. Conversion of this motif to that of the non-binding PB1-F2 suffices to lose inhibition of NLRP3 dependent IL-1ß release. The TQGS motif further alters the subcellular localization of PB1-F2 and its colocalization with NLRP3 LRR and pyrin domain. Structural predictions suggest the establishment of additional hydrogen bonds between the C-terminus of PB1-F2 and the LRR domain of NLRP3, with two hydrogen bonds connecting to threonine and glutamine of the TQGS motif. Phylogenetic data show that the identified NLRP3 interaction motif in PB1-F2 is widely conserved among recent IAV-infecting humans. Our data explain at a molecular level the specificity of NLRP3 inhibition by influenza A virus. IMPORTANCE: Influenza A virus infection is accompanied by a strong inflammatory response and high fever. The human immune system facilitates the swift clearance of the virus with this response. An essential signal protein in the proinflammatory host response is IL-1b. It is released from inflammatory macrophages, and its production and secretion depend on the function of NLRP3. We had previously shown that influenza A virus blocks NLRP3 activation by the expression of a viral inhibitor, PB1-F2. Here, we demonstrate how this short peptide binds to NLRP3 and provide evidence that a four amino acid stretch in PB1-F2 is necessary and sufficient to mediate this binding. Our data identify a new virus-host interface required to block one signaling path of the innate host response against influenza A virus.


Assuntos
Motivos de Aminoácidos , Vírus da Influenza A , Proteína 3 que Contém Domínio de Pirina da Família NLR , Ligação Proteica , Proteínas Virais , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteínas Virais/metabolismo , Proteínas Virais/genética , Proteínas Virais/química , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Células HEK293 , Influenza Humana/virologia , Influenza Humana/imunologia , Sequência de Aminoácidos
14.
EBioMedicine ; 103: 105103, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38574407

RESUMO

BACKGROUND: World Health Organisation (WHO) and USA Centers for Disease Control and Prevention (U.S. CDC) recommendations now allow simultaneous administration of COVID-19 and other vaccines. We compared antibody responses after coadministration of influenza and bivalent COVID-19 vaccines in the same (ipsilateral) arm vs. different (contralateral) arms. METHODS: Pre- and post-vaccination serum samples from individuals in the Prospective Assessment of COVID-19 in a Community (PACC) cohort were used to conduct haemaglutination inhibition (HI) assays with the viruses in the 2022-2023 seasonal influenza vaccine and focus reduction neutralisation tests (FRNT) using a BA.5 SARS-CoV-2 virus. The effect of ipsilateral vs. contralateral vaccination on immune responses was inferred in a model that accounted for higher variance in vaccine responses at lower pre-vaccination titers. FINDINGS: Ipsilateral vaccination did not cause higher influenza vaccine responses compared to contralateral vaccination. The response to SARS-CoV-2 was slightly increased in the ipsilateral group, but equivalence was not excluded. INTERPRETATION: Coadministration of influenza and bivalent COVID-19 vaccines in the same arm or different arms did not strongly influence the antibody response to either vaccine. FUNDING: This work was supported by the U.S. CDC (grant number: 75D30120C09259).


Assuntos
Anticorpos Antivirais , Vacinas contra COVID-19 , COVID-19 , Vacinas contra Influenza , Influenza Humana , SARS-CoV-2 , Humanos , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/administração & dosagem , COVID-19/prevenção & controle , COVID-19/imunologia , SARS-CoV-2/imunologia , Masculino , Feminino , Pessoa de Meia-Idade , Influenza Humana/prevenção & controle , Influenza Humana/imunologia , Adulto , Formação de Anticorpos/imunologia , Vacinação/métodos , Idoso , Estudos Prospectivos , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia
15.
J Virol ; 98(5): e0019824, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38591879

RESUMO

The involvement of secreted phospholipase A2s in respiratory diseases, such as asthma and respiratory viral infections, is well-established. However, the specific role of secreted phospholipase A2 group IIE (PLA2G2E) during influenza virus infection remains unexplored. Here, we investigated the role of PLA2G2E during H1N1 influenza virus infection using a targeted mouse model lacking Pla2g2e gene (Pla2g2e-/-). Our findings demonstrated that Pla2g2e-/- mice had significantly lower survival rates and higher viral loads in lungs compared to wild-type mice following influenza virus infection. While Pla2g2e-/- mice displayed comparable innate and humoral immune responses to influenza virus challenge, the animals showed impaired influenza-specific cellular immunity and reduced T cell-mediated cytotoxicity. This indicates that PLA2G2E is involved in regulating specific T cell responses during influenza virus infection. Furthermore, transgenic mice expressing the human PLA2G2E gene exhibited resistance to influenza virus infection along with enhanced influenza-specific cellular immunity and T cell-mediated cytotoxicity. Pla2g2e deficiency resulted in perturbation of lipid mediators in the lung and T cells, potentially contributing to its impact on the anti-influenza immune response. Taken together, these findings suggest that targeting PLA2G2E could hold potential as a therapeutic strategy for managing influenza virus infections.IMPORTANCEThe influenza virus is a highly transmissible respiratory pathogen that continues to pose a significant public health concern. It effectively evades humoral immune protection conferred by vaccines and natural infection due to its continuous viral evolution through the genetic processes of antigenic drift and shift. Recognition of conserved non-mutable viral epitopes by T cells may provide broad immunity against influenza virus. In this study, we have demonstrated that phospholipase A2 group IIE (PLA2G2E) plays a crucial role in protecting against influenza virus infection through the regulation of T cell responses, while not affecting innate and humoral immune responses. Targeting PLA2G2E could therefore represent a potential therapeutic strategy for managing influenza virus infection.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Pulmão , Infecções por Orthomyxoviridae , Animais , Camundongos , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Vírus da Influenza A Subtipo H1N1/imunologia , Pulmão/virologia , Pulmão/imunologia , Pulmão/patologia , Humanos , Fosfolipases A2 do Grupo II/genética , Fosfolipases A2 do Grupo II/imunologia , Linfócitos T/imunologia , Camundongos Knockout , Imunidade Celular , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Carga Viral , Modelos Animais de Doenças , Imunidade Humoral , Imunidade Inata , Influenza Humana/imunologia , Influenza Humana/virologia , Feminino
16.
Vaccine ; 42(14): 3365-3373, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38627145

RESUMO

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.


Assuntos
Anticorpos Antivirais , Testes de Inibição da Hemaglutinação , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Epitopos Imunodominantes , Influenza Humana , Humanos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Feminino , Masculino , Adulto , Epitopos Imunodominantes/imunologia , Pessoa de Meia-Idade , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Adulto Jovem , Fatores Etários , Fatores Sexuais , Adolescente , Estudos de Coortes , Idoso , Antígenos Virais/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue
17.
Vaccine ; 42(15): 3455-3460, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38658205

RESUMO

BACKGROUND: Previous research suggested that the inactivated influenza vaccine (IIV) may protect against SARS-CoV-2 infection or a severe course of COVID-19. These findings were however based on cohort studies, that are prone to confounding by indication. We examined the association between IIV and SARS-Cov-2 infection in a Dutch population using a test-negative design. METHODS: This test-negative case-control study was conducted in adults (≥60) who tested because of COVID-19 like symptoms at community SARS-CoV-2 testing locations in the Netherlands during the period of November 8th 2021-March 11th 2022. Information on receipt of IIV in October-November 2021 was routinely collected at each visit. Logistic regression was used to calculate unadjusted, partially (sex, age, education level) and fully adjusted (COVID-19 vaccination, IIV 2020) odds ratios (ORs) for receipt of IIV in SARS-CoV-2 positive versus negative subjects. Differential effects on SARS-CoV-2 risk by time since IIV were investigated by including an interaction term for calendar time: November 2021-January 2022 vs February-March 2022. RESULTS: In total, 1,832 participants were included in the main analysis, of whom 336 (18.3 %) had a positive SARS-CoV-2 test. No significant association between IIV and SARS-CoV-2 infection was found; fully adjusted OR of 1.07 (95 % CI: 0.78-1.49). The interaction term for time periods was not significant (1.04 [95 % CI: 0.51-2.15], p = 0.91). Results were robust in sensitivity analyses. CONCLUSIONS: While earlier observational studies suggested a protective non-specific effect of IIV and SARS-CoV-2 infections, this smaller, but well controlled test-negative design study does not suggest an effect, either positive or negative. Larger test-negative design studies, or alternative designs such as the self-controlled case series design are needed to confirm these findings and provide more definite answers on the topic.


Assuntos
COVID-19 , Vacinas contra Influenza , Influenza Humana , SARS-CoV-2 , Vacinas de Produtos Inativados , Humanos , COVID-19/prevenção & controle , COVID-19/imunologia , COVID-19/epidemiologia , Feminino , Masculino , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Pessoa de Meia-Idade , Vacinas de Produtos Inativados/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Idoso , Países Baixos/epidemiologia , Estudos de Casos e Controles , SARS-CoV-2/imunologia , Influenza Humana/prevenção & controle , Influenza Humana/imunologia , Idoso de 80 Anos ou mais , Vacinação/estatística & dados numéricos
18.
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
19.
Appl Microbiol Biotechnol ; 108(1): 307, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38656587

RESUMO

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.


Assuntos
Vacinas contra Influenza , Controle de Qualidade , Ressonância de Plasmônio de Superfície , Ressonância de Plasmônio de Superfície/métodos , Vacinas contra Influenza/imunologia , Humanos , Anticorpos Antivirais/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Influenza Humana/diagnóstico , Influenza Humana/prevenção & controle , Influenza Humana/imunologia , Imunoensaio/métodos , Imunoensaio/normas , Técnicas Biossensoriais/métodos , Vírus da Influenza A/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA