Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 298
Filter
1.
mSphere ; : e0028324, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39087764

ABSTRACT

In 2009, a novel swine-origin H1N1 virus emerged, causing a pandemic. The virus, known as H1N1pdm09, quickly displaced the circulating H1 lineage and became the dominant seasonal influenza A virus subtype infecting humans. Human-to-swine spillovers of the H1N1pdm09 have occurred frequently, and each occurrence has led to sustained transmission of the human-origin H1N1pdm09 within swine populations. In the present study, we developed a lipid nanoparticle-based DNA vaccine (LNP-DNA) containing the hemagglutinin gene of a swine-origin H1N1pdm09. In pigs, this LNP-DNA vaccine induced a robust antibody response after a single intramuscular immunization and protected the pigs against challenge infection with the homologous swine-origin H1N1pdm09 virus. In a mouse model, the LNP-DNA vaccine induced antibody and T-cell responses and protected mice against lethal challenge with a mouse-adapted human-origin H1N1pdm09 virus. These findings demonstrate the potential of the LNP-DNA vaccine to protect against both swine- and human-origin H1N1pdm09 viruses. IMPORTANCE: Swine influenza A virus (IAV) is widespread and causes significant economic losses to the swine industry. Moreover, bidirectional transmission of IAV between swine and humans commonly occurs. Once introduced into the swine population, human-origin IAV often reassorts with endemic swine IAV, resulting in reassortant viruses. Thus, it is imperative to develop a vaccine that is not only effective against IAV strains endemic in swine but also capable of preventing the spillover of human-origin IAV. In this study, we developed a lipid nanoparticle-encapsulated DNA plasmid vaccine (LNP-DNA) that demonstrates efficacy against both swine- and human-origin H1N1 viruses. The LNP-DNA vaccines are non-infectious and non-viable, meeting the criteria to serve as a vaccine platform for rapidly updating vaccines. Collectively, this LNP-DNA vaccine approach holds great potential for alleviating the impact of IAV on the swine industry and preventing the emergence of reassortant IAV strains.

2.
Viruses ; 16(7)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39066271

ABSTRACT

The unexpected emergence of oseltamivir-resistant A(H1N1) viruses in 2008 was facilitated in part by the establishment of permissive secondary neuraminidase (NA) substitutions that compensated for the fitness loss due to the NA-H275Y resistance substitution. These viruses were replaced in 2009 by oseltamivir-susceptible A(H1N1)pdm09 influenza viruses. Genetic analysis and screening of A(H1N1)pdm09 viruses circulating in Germany between 2009 and 2024 were conducted to identify any potentially synergistic or resistance-associated NA substitutions. Selected viruses were then subjected to further characterization in vitro. In the NA gene of circulating A(H1N1)pdm09 viruses, two secondary substitutions, NA-V241I and NA-N369K, were identified. These substitutions demonstrated a stable lineage in phylogenetic analysis since the 2010-2011 influenza season. The data indicate a slight increase in viral NA bearing two additional potentially synergistic substitutions, NA-I223V and NA-S247N, in the 2023-2024 season, which both result in a slight reduction in susceptibility to NA inhibitors. The accumulation of secondary synergistic substitutions in the NA of A(H1N1)pdm09 viruses increases the probability of the emergence of antiviral-resistant viruses. Therefore, it is crucial to closely monitor the evolution of circulating influenza viruses and to develop additional antiviral drugs against different target proteins.


Subject(s)
Antiviral Agents , Drug Resistance, Viral , Evolution, Molecular , Influenza A Virus, H1N1 Subtype , Influenza, Human , Mutation , Neuraminidase , Oseltamivir , Phylogeny , Viral Proteins , Neuraminidase/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/enzymology , Antiviral Agents/pharmacology , Drug Resistance, Viral/genetics , Humans , Influenza, Human/virology , Viral Proteins/genetics , Viral Proteins/metabolism , Oseltamivir/pharmacology , Germany , Amino Acid Substitution , Animals , Dogs
3.
J Med Virol ; 96(6): e29736, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38864349

ABSTRACT

Although a range of blood traits have been reported to be associated with influenza A(H1N1)pdm09 (H1N1pdm09) disease severity, their underlying causal relationships and biological mechanisms have remained unclear. This study aimed to investigate the causal relationship between blood traits and H1N1pdm09 using a two-sample Mendelian randomization analysis. Based on the data from our in-house genome-wide association study (GWAS) on H1N1pdm09 disease severity (Ncase [severe] = 70, Ncontrol [mild] = 95) and GWAS summaries of 44 blood traits from Biobank Japan (N = 12 303-143 658), we identified the potential causal effect of blood traits on severe H1N1pdm09. The inverse variance weighted method analysis revealed significant causal effects of lower aspartate aminotransferase (AST, ß = -3.212, p = 0.019), low-density-lipoprotein cholesterol (LDL-C, ß = -1.372, p = 0.045), and basophil counts (Baso, ß = -1.638, p = 0.047) on severe H1N1pdm09 disease. Additionally, polygenic risk score analysis further confirmed genetic overlap between these blood traits and severe H1N1pdm09 disease. This study provided evidence linking the lower level of AST, LDL-C, and lower count of Baso with severe H1N1pdm09 disease, potentially identifying new therapeutic targets for patients with severe influenza.


Subject(s)
Genome-Wide Association Study , Influenza A Virus, H1N1 Subtype , Influenza, Human , Mendelian Randomization Analysis , Humans , Influenza, Human/virology , Influenza, Human/genetics , Influenza, Human/epidemiology , Influenza A Virus, H1N1 Subtype/genetics , Japan/epidemiology , Genetic Predisposition to Disease , Severity of Illness Index , Polymorphism, Single Nucleotide , Aspartate Aminotransferases/blood , Cholesterol, LDL/blood , Asia, Eastern/epidemiology , Asian People/genetics , East Asian People
4.
Emerg Infect Dis ; 30(7): 1410-1415, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38916572

ABSTRACT

Since May 2023, a novel combination of neuraminidase mutations, I223V + S247N, has been detected in influenza A(H1N1)pdm09 viruses collected in countries spanning 5 continents, mostly in Europe (67/101). The viruses belong to 2 phylogenetically distinct groups and display ≈13-fold reduced inhibition by oseltamivir while retaining normal susceptibility to other antiviral drugs.


Subject(s)
Antiviral Agents , Drug Resistance, Viral , Influenza A Virus, H1N1 Subtype , Influenza, Human , Neuraminidase , Oseltamivir , Phylogeny , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Humans , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Influenza, Human/virology , Influenza, Human/drug therapy , Influenza, Human/epidemiology , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Drug Resistance, Viral/genetics , Mutation
5.
Sci Rep ; 14(1): 10660, 2024 05 09.
Article in English | MEDLINE | ID: mdl-38724525

ABSTRACT

Influenza Like Illness (ILI) and Severe Acute Respiratory Infection (SARI) cases are more prone to Influenza and SARS-CoV-2 infection. Accordingly, we genetically characterized Influenza and SARS-CoV-2 in 633 ILI and SARI cases by rRT-PCR and WGS. ILI and SARI cases showed H1N1pdm09 prevalence of 20.9% and 23.2% respectively. 135 (21.3%) H1N1pdm09 and 23 (3.6%) H3N2 and 5 coinfection (0.78%) of H1N1pdm09 and SARS-CoV-2 were detected. Phylogenetic analysis revealed H1N1pdm09 resemblance to clade 6B.1A.5a.2 and their genetic relatedness to InfA/Perth/34/2020, InfA/Victoria/88/2020 and InfA/Victoria/2570/2019. Pan 24 HA and 26 NA nonsynonymous mutations and novel HA (G6D, Y7F, Y78H, P212L, G339R, T508K and S523T) and NA (S229A) mutations were observed. S74R, N129D, N156K, S162N, K163Q and S164T alter HA Cb and Sa antibody recognizing site. Similarly, M19T, V13T substitution and multiple mutations in transmembrane and NA head domain drive antigenic drift. SARS-CoV-2 strains genetically characterized to Omicron BA.2.75 lineage containing thirty nonsynonymous spike mutations exhibited enhanced virulence and transmission rates. Coinfection although detected very minimal, the mutational changes in H1N1pdm09 and SARS-CoV-2 virus infected individuals could alter antibody receptor binding sites, allowing the viruses to escape immune response resulting in better adaptability and transmission. Thus continuous genomic surveillance is required to tackle any future outbreak.


Subject(s)
COVID-19 , Influenza A Virus, H1N1 Subtype , Influenza, Human , Phylogeny , SARS-CoV-2 , Humans , Influenza A Virus, H1N1 Subtype/genetics , SARS-CoV-2/genetics , Influenza, Human/virology , Influenza, Human/epidemiology , COVID-19/virology , COVID-19/epidemiology , Adult , Middle Aged , Male , Female , Adolescent , Young Adult , Genome, Viral/genetics , Aged , Coinfection/virology , Coinfection/epidemiology , Child , Child, Preschool , Severe Acute Respiratory Syndrome/virology , Severe Acute Respiratory Syndrome/epidemiology , Mutation , Infant
6.
J Med Virol ; 96(5): e29657, 2024 May.
Article in English | MEDLINE | ID: mdl-38727035

ABSTRACT

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.


Subject(s)
Antigens, Viral , Influenza A Virus, H1N1 Subtype , Influenza, Human , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Humans , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Influenza, Human/virology , Influenza, Human/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Machine Learning , Evolution, Molecular , Epitopes/genetics , Epitopes/immunology , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19/virology , COVID-19/immunology , Pandemics/prevention & control , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , SARS-CoV-2/genetics , SARS-CoV-2/immunology
7.
Rev Med Virol ; 34(3): e2533, 2024 May.
Article in English | MEDLINE | ID: mdl-38635404

ABSTRACT

Influenzavirus is among the most relevant candidates for a next pandemic. We review here the phylogeny of former influenza pandemics, and discuss candidate lineages. After briefly reviewing the other existing antiviral options, we discuss in detail the evidences supporting the efficacy of passive immunotherapies against influenzavirus, with a focus on convalescent plasma.


Subject(s)
Influenza A Virus, H7N9 Subtype , Influenza, Human , Humans , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Pandemics , Immunotherapy
8.
Virol J ; 21(1): 57, 2024 03 06.
Article in English | MEDLINE | ID: mdl-38448981

ABSTRACT

BACKGROUND: Non-pharmaceutical interventions implemented during the COVID-19 pandemic resulted in a marked reduction in influenza infections globally. The absence of influenza has raised concerns of waning immunity, and potentially more severe influenza seasons after the pandemic. METHODS: To evaluate immunity towards influenza post-COVID-19 pandemic we have assessed influenza A epidemics in Norway from October 2016 to June 2023 and measured antibodies against circulating strains of influenza A(H1N1)pdm09 and A(H3N2) in different age groups by hemagglutination inhibition (HAI) assays in a total of 3364 serum samples collected in 2019, 2021, 2022 and 2023. RESULTS: Influenza epidemics in Norway from October 2016 until June 2023 were predominately influenza As, with a mixture of A(H1N1)pdm09 and A(H3N2) subtype predominance. We did not observe higher numbers of infections during the influenza epidemics following the COVID-19 pandemic than in pre-COVID-19 seasons. Frequencies of protective HAI titers against A(H1N1)pdm09 and A(H3N2) viruses were reduced in sera collected in 2021 and 2022, compared to sera collected in 2019. The reduction could, however, largely be explained by antigenic drift of new virus strains, as protective HAI titers remained stable against the same strain from one season to the next. However, we observed the development of an immunity gap in the youngest children during the pandemic which resulted in a prominent reduction in HAI titers against A(H1N1)pdm09 in 2021 and 2022. The immunity gap was partially closed in sera collected in 2023 following the A(H1N1)pdm09-dominated influenza seasons of 2022/2023. During the 2022/2023 epidemic, drift variants of A(H1N1)pdm09 belonging to the 5a.2a.1 clade emerged, and pre-season HAI titers were significantly lower against this clade compared to the ancestral 5a.2 clade. CONCLUSION: The observed reduction in protective antibodies against A(H1N1)pdm09 and A(H3N2) viruses post COVID-19 is best explained by antigenic drift of emerging viruses, and not waning of antibody responses in the general population. However, the absence of influenza during the pandemic resulted in an immunity gap in the youngest children. While this immunity gap was partially closed following the 2022/2023 influenza season, children with elevated risk of severe infection should be prioritized for vaccination.


Subject(s)
COVID-19 , Influenza A Virus, H1N1 Subtype , Influenza, Human , Child , Humans , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Cross-Sectional Studies , Antigenic Drift and Shift , Influenza A Virus, H3N2 Subtype , COVID-19/epidemiology , Pandemics
9.
Vaccines (Basel) ; 12(3)2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38543939

ABSTRACT

The current influenza season started in Italy in October 2023, approaching the epidemic peak in late December (52nd week of the year). We aimed to explore the mid-term virologic surveillance data of the 2023/2024 influenza season (from 16 October 2023 to 7 January 2024) in Sicily, the fourth most populous Italian region. A test-negative design was used to estimate the effectiveness of seasonal influenza vaccine (VE) against A(H1N1)pdm09 virus, the predominant subtype in Sicily (96.2% of laboratory-confirmed influenza cases). Overall, 29.2% (n = 359/1230) of oropharyngeal swabs collected from patients with influenza-like illness (ILI) were positive for influenza. Among the laboratory-confirmed influenza cases, 12.5% (n = 45/359) were vaccinated against influenza, with higher prevalence of laboratory-confirmed diagnosis of influenza A among subjects vaccinated with quadrivalent inactivated standard dose (29.4%), live attenuated intranasal (25.1%), and quadrivalent inactivated high-dose (23.8%) influenza vaccines. Comparing influenza vaccination status for the 2023/2024 season among laboratory-confirmed influenza-positive and -negative samples, higher vaccination rates in influenza-negative samples (vs. positive) were observed in all age groups, except for 45-64 years old, regardless of sex and comorbidities. The overall adjusted VE (adj-VE) was 41.4% [95%CI: 10.5-61.6%], whereas the adj-VE was 37.9% [95%CI: -0.7-61.7%] among children 7 months-14 years old and 52.7% [95%CI: -38.0-83.8%] among the elderly (≥65 years old).

10.
Int J Med Microbiol ; 314: 151598, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38237287

ABSTRACT

Respiratory viral infections may have different impacts ranging from infection without symptoms to severe disease or even death though the reasons are not well characterized. A patient (age group 5-15 years) displaying symptoms of hemolytic uremic syndrome died one day after hospitalization. qPCR, next generation sequencing, virus isolation, antigenic characterization, resistance analysis was performed and virus replication kinetics in well-differentiated airway cells were determined. Autopsy revealed hemorrhagic pneumonia as major pathological manifestation. Lung samples harbored a large population of A(H1N1)pdm09 viruses with the polymorphism H456H/Y in PB1 polymerase. The H456H/Y viruses replicated much faster to high viral titers than upper respiratory tract viruses in vitro. H456H/Y-infected air-liquid interface cultures of differentiated airway epithelial cells did reflect a more pronounced loss of ciliated cells. A different pattern of virus quasispecies was found in the upper airway samples where substitution S263S/F (HA1) was observed. The data support the notion that viral quasispecies had evolved locally in the lung to support high replicative fitness. This change may have initiated further pathogenic processes leading to rapid dissemination of inflammatory mediators followed by development of hemorrhagic lung lesions and fatal outcome.


Subject(s)
Hemolytic-Uremic Syndrome , Influenza A Virus, H1N1 Subtype , Influenza, Human , Humans , Child, Preschool , Child , Adolescent , Epithelial Cells , Lung , Influenza, Human/epidemiology
11.
J Med Virol ; 96(2): e29427, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38288882

ABSTRACT

Influenza virus is known to cause mild to severe respiratory infections and is also prone to genetic mutations. Of all the mutations, neuraminidase (NA) gene mutations are a matter of concern, as most approved antivirals target this protein. During the 2020 influenza season, an emergence of mutation in the NA gene, affecting the binding of the World Health Organization (WHO)-recommended probes to the specific site of the NA gene, was reported by our group. As a result of this mutation, the WHO-recommended allelic discrimination real-time reverse transcriptase polymerase chain reaction (RT-PCR) assay was unable to detect wild-type (H275) or mutant oseltamivir-resistant (Y275) strains of influenza A(H1N1)pmd09 viruses. In the current study, the WHO-recommended probes were redesigned according to the mutation in the probe binding site. Fifty undetermined samples (2020-2021) from the previous study were retested with the newly designed probes and found to be positive for H275 and/or Y275. The results obtained were similar to the Sanger sequencing results from the previous study, suggesting that the redesigned probes were efficient in discriminating between wild-type and mutant-type viruses. Furthermore, 133 samples from 2022, making a total of 183 samples (2020-2022), were tested using improved allelic discrimination real-time RT-PCR, and the overall prevalence rate of oseltamivir resistance in 2020-2022 was found to be 0.54%.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human , Humans , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Influenza A Virus, H1N1 Subtype/genetics , Reverse Transcriptase Polymerase Chain Reaction , Mutation, Missense , Viral Proteins/genetics , Drug Resistance, Viral/genetics , Mutation , Neuraminidase/genetics
12.
J Biomol Struct Dyn ; 42(4): 2013-2033, 2024.
Article in English | MEDLINE | ID: mdl-37166274

ABSTRACT

The advent of influenza A (H1N1) drug-resistant strains led to the search quest for more potent inhibitors of the influenza A virus, especially in this devastating COVID-19 pandemic era. Hence, the present research utilized some molecular modelling strategies to unveil new camphor imine-based compounds as anti-influenza A (H1N1) pdm09 agents. The 2D-QSAR results revealed GFA-MLR (R2train = 0.9158, Q2=0.8475) and GFA-ANN (R2train = 0.9264, Q2=0.9238) models for the anti-influenza A (H1N1) pdm09 activity prediction which have passed the QSAR model acceptability thresholds. The results from the 3D-QSAR studies also revealed CoMFA (R2train =0.977, Q2=0.509) and CoMSIA_S (R2train =0.976, Q2=0.527) models for activity predictions. Based on the notable information derived from the 2D-QSAR, 3D-QSAR, and docking analysis, ten (10) new camphor imine-based compounds (22a-22j) were designed using the most active compound 22 as the template. Furthermore, the high predicted activity and binding scores of compound 22j were further justified by the high reactive sites shown in the electrostatic potential maps and other quantum chemical calculations. The MD simulation of 22j in the active site of the influenza hemagglutinin (HA) receptor confirmed the dynamic stability of the complex. Moreover, the appraisals of drug-likeness and ADMET properties of the proposed compounds showed zero violation of Lipinski's criteria with good pharmacokinetic profiles. Hence, the outcomes in this work recommend further in-depth in vivo and in-vitro investigations to validate these theoretical findings.Communicated by Ramaswamy H. Sarma.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human , Humans , Influenza, Human/drug therapy , Camphor/pharmacology , Camphor/chemistry , Imines/pharmacology , Imines/chemistry , Pandemics , Quantitative Structure-Activity Relationship , Antibodies , Molecular Docking Simulation
13.
Emerg Infect Dis ; 30(1): 168-171, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38147510

ABSTRACT

We detected high titers of cross-reactive neuraminidase inhibition antibodies to influenza A(H5N1) virus clade 2.3.4.4b in 96.8% (61/63) of serum samples from healthy adults in Hong Kong in 2020. In contrast, antibodies at low titers were detected in 42% (21/50) of serum samples collected in 2009. Influenza A(H1N1)pdm09 and A(H5N1) titers were correlated.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A Virus, H5N1 Subtype , Influenza Vaccines , Influenza in Birds , Influenza, Human , Adult , Animals , Humans , Neuraminidase , Antibodies, Viral
14.
Viruses ; 15(12)2023 11 30.
Article in English | MEDLINE | ID: mdl-38140609

ABSTRACT

In 2009, a novel H1N1 influenza virus caused the first influenza pandemic of the 21st century. Studies have shown that the influenza M gene played important roles in the pathogenicity and transmissibility of the 2009 H1N1 pandemic ((H1N1)pdm09), whilst the underlying mechanism remains unclear. The influenza M gene encodes two proteins, matrix protein 1 and matrix protein 2, which play important roles in viral replication and assembly. In this study, it is found that the M2 protein of the (H1N1)pdm09 virus showed a lower mobility rate than the North America triple-reassortant influenza M2 protein in Polyacrylamide Gel Electrophoresis (PAGE). The site-directed mutations of the amino acids of (H1N1)pdm09 M2 revealed that E79 is responsible for the mobility rate change. Further animal studies showed that the (H1N1)pdm09 containing a single M2-E79K was significantly attenuated compared with the wild-type virus in mice and induced lower proinflammatory cytokines and IFNs in mouse lungs. Further in vitro studies indicated that this mutation also affected NLRP3 inflammasome activation. To reveal the reason why they have different mobility rates, a circular dichroism spectra assay was employed and showed that the two M2 proteins displayed different secondary structures. Overall, our findings suggest that M2 E79 is important for the virus replication and pathogenicity of (H1N1)pdm09 through NLRP3 inflammasome and proinflammatory response.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A virus , Influenza, Human , Orthomyxoviridae Infections , Animals , Mice , Humans , Influenza A Virus, H1N1 Subtype/physiology , NLR Family, Pyrin Domain-Containing 3 Protein , Virulence , Inflammasomes
15.
Int J Mol Sci ; 24(24)2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38139214

ABSTRACT

Influenza virus strain A/South Africa/3626/2013 (H1N1)pdm09 (SA-WT) is a non-mouse-adapted model strain that has naturally high pathogenic properties in mice. It has been suggested that the high pathogenicity of this strain for mice could be due to the three strain-specific substitutions in the polymerase complex (Q687R in PB1, N102T in PB2, and E358E/K heterogeneity in PB2). To evaluate the role of these replacements, SA-WT was passaged five times in mouse lungs, and the genome of the mouse-adapted version of the SA-WT strain (SA-M5) was sequenced. SA-M5 lost E358E/K heterogeneity and retained E358, which is the prevalent amino acid at this position among H1N1pdm09 strains. In addition, in the hemagglutinin of SA-M5, two heterogeneous substitutions (G155G/E and S190S/R) were identified. Both viruses, SA-M5 and SA-WT, were compared for their toxicity, ability to replicate, pathogenicity, and immunogenicity in mice. In mice infected with SA-M5 or SA-WT strains, toxicity, virus titer in pulmonary homogenates, and mouse survival did not differ significantly. In contrast, an increase in the immunogenicity of SA-M5 compared to SA-WT was observed. This increase could be due to the substitutions G155G/E and S190S/R in the HA of SA-M5. The prospects for using SA-M5 in studying the immunogenicity mechanisms were also discussed.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human , Humans , Influenza A Virus, H1N1 Subtype/genetics , Virulence/genetics , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Phylogeny
16.
J Med Virol ; 95(9): e29106, 2023 09.
Article in English | MEDLINE | ID: mdl-37712255

ABSTRACT

China experienced a severe influenza season that began at the end of February 2023. The aim of this post hoc analysis was to investigate the clinical, epidemiological, and genomic features of this outbreak in Beijing. The number of cases increased rapidly from the end of February and reached its peak in March, with 7262 confirmed cases included in this study. The median age was 33 years, and 50.3% of them were male. The average daily positive rate reached 69% during the peak period. The instantaneous reproduction number (Rt) showed a median of 2.1, exceeded 2.5 initially, and remaining above 1 for the following month. The most common symptoms were fever (75.0%), cough (51.0%), and expectoration (42.9%), with a median body temperature of 38.5°C (interquartile range 38-39). Eight clinical symptoms were more likely to be observed in cases with fever, with odds ratio greater than 1. Viral shedding time ranged from 3 to 25 days, with median of 7.5 days. The circulating viruses in Beijing mainly included H1N1pdm09 (clades 5a.2a and 5a.2a.1), following with H3N2 (clade 2a.2a.3a.1). The descriptive study suggests that influenza viruses in this influenza season had a higher transmissibility and longer shedding duration, with fever being the most common symptom.


Subject(s)
Influenza A virus , Influenza, Human , Male , Humans , Adult , Female , Seasons , Beijing/epidemiology , Influenza A virus/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/epidemiology , Genomics , Disease Outbreaks , Fever/epidemiology
17.
Viruses ; 15(5)2023 05 04.
Article in English | MEDLINE | ID: mdl-37243200

ABSTRACT

Influenza virus can infect the vascular endothelium and cause endothelial dysfunction. Persons at higher risk for severe influenza are patients with acute and chronic cardiovascular disorders; however, the mechanism of influenza-induced cardiovascular system alteration remains not fully understood. The aim of the study was to assess the functional activity of mesenteric blood vessels of Wistar rats with premorbid acute cardiomyopathy infected with Influenza A(H1N1)pdm09 virus. For this, we determined (1) the vasomotor activity of mesenteric blood vessels of Wistar rats using wire myography, (2) the level of expression of three endothelial factors: endothelial nitric oxide synthase (eNOS), plasminogen activator inhibitor-1 (PAI-1), and tissue plasminogen activator (tPA) in the endothelium of mesenteric blood vessels using immunohistochemistry, and (3) the concentration of PAI-1 and tPA in the blood plasma using ELISA. Acute cardiomyopathy in animals was induced by doxorubicin (DOX) following infection with rat-adapted Influenza A(H1N1)pdm09 virus. The functional activity of mesenteric blood vessels was analyzed at 24 and 96 h post infection (hpi). Thus, the maximal response of mesenteric arteries to both vasoconstrictor and vasodilator at 24 and 96 hpi was significantly decreased compared with control. Expression of eNOS in the mesenteric vascular endothelium was modulated at 24 and 96 hpi. PAI-1 expression increased 3.47-fold at 96 hpi, while the concentration of PAI-1 in the blood plasma increased 6.43-fold at 24 hpi compared with control. The tPA concentration in plasma was also modulated at 24 hpi and 96 hpi. The obtained data indicate that influenza A(H1N1)pdm09 virus aggravates the course of premorbid acute cardiomyopathy in Wistar rats, causing pronounced dysregulation of endothelial factor expression and vasomotor activity impairment of mesenteric arteries.


Subject(s)
Cardiomyopathies , Influenza A Virus, H1N1 Subtype , Influenza, Human , Rats , Animals , Humans , Rats, Wistar , Tissue Plasminogen Activator , Plasminogen Activator Inhibitor 1
18.
J Med Virol ; 95(5): e28764, 2023 05.
Article in English | MEDLINE | ID: mdl-37212286

ABSTRACT

Influenza viruses can mutate genetically and cause a range of respiratory ailments. The H275Y mutation in the neuraminidase (NA) gene reduces the effectiveness of oseltamivir, a widely used drug for the treatment of Influenza A and B virus infection. The World Health Organization (WHO) recommends single-nucleotide polymorphism assays to detect this mutation. The present study aims to estimate the prevalence of H275Y mutation conferring oseltamivir resistance in Influenza A(H1N1)pdm09 virus among hospitalized patients from June 2014 to December 2021. Following the WHO protocol, allelic discrimination real-time RT-PCR was performed for 752 samples. Out of the 752 samples, 1 tested positive for Y275 gene mutation by allelic discrimination real-time RT-PCR. In samples of years 2020 and 2021, neither the H275 nor Y275 genotype was detected. Sequencing of the NA gene of all negative samples showed a mismatch between the NA sequence and the probes used in the allelic discrimination assay. Also, Y275 mutation was detected in only 1 sample from 2020. The prevalence of oseltamivir resistance was estimated as 0.27% among the Influenza A(H1N1)pdm09 patients during 2014-2021. The study highlights that the WHO-recommended probes for detecting H275Y mutation may not be useful to detect 2020 and 2021 circulating strains of Influenza A(H1N1)pdm09, emphasizing the need for continuous monitoring of mutations in the influenza virus.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A virus , Influenza, Human , Humans , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Influenza, Human/drug therapy , Influenza, Human/epidemiology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Influenza A Virus, H1N1 Subtype/genetics , Reverse Transcriptase Polymerase Chain Reaction , Mutation, Missense , Mutation , Influenza A virus/genetics , Neuraminidase/genetics , Drug Resistance, Viral/genetics
19.
Viruses ; 15(2)2023 02 14.
Article in English | MEDLINE | ID: mdl-36851749

ABSTRACT

The influenza A(H1N1)pdm09 virus that emerged in 2009 causes seasonal epidemic worldwide. The virus acquired several amino acid substitutions that were responsible for antigenic drift until the 2018-2019 influenza season. Viruses possessing mutations in the NA and PA proteins that cause reduced susceptibility to NA inhibitors and baloxavir marboxil, respectively, have been detected after antiviral treatment, albeit infrequently. Here, we analyzed HA, NA, and PA sequences derived from A(H1N1)pdm09 viruses that were isolated during the 2018-2019 and 2019-2020 influenza seasons in Japan. We found that A(H1N1)pdm09 viruses possessing the D187A and Q189E substitutions in HA emerged and dominated during the 2019-2020 season; these substitutions in the antigenic site Sb, a high potency neutralizing antibody-eliciting site for humans, changed the antigenicity of A(H1N1)pdm09 viruses. Furthermore, we found that isolates possessing the N156K substitution, which was predicted to affect the antigenicity of A(H1N1)pdm09 virus at the laboratory level, were detected at a frequency of 1.0% in the 2018-2019 season but 10.1% in the 2019-2020 season. These findings indicate that two kinds of antigenically drifted viruses-N156K and D187A/Q189E viruses-co-circulated during the 2019-2020 influenza season in Japan.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A virus , Influenza, Human , Humans , Influenza A Virus, H1N1 Subtype/genetics , Seasons , Japan/epidemiology , Influenza, Human/epidemiology
20.
J Med Virol ; 95(2): e28498, 2023 02.
Article in English | MEDLINE | ID: mdl-36653733

ABSTRACT

Community surveillance found the 2019-2020 A(H1N1)pdm09 predominant influenza season in Israel to be a high-intensity season with an early and steep morbidity peak. To further characterize disease severity in the 2019-2020 season, we analyzed a cohort of hospitalized patients with laboratory-confirmed influenza from this season (n = 636). Quantitative polymerase chain reaction was performed on clinical samples to detect the presence of influenza. Demographic, clinical, and laboratory data were retrieved via electronic health records and MDClone. Electronic health records were accessed to obtain data on intensive care unit patients, missing data and for data verification purposes. Univariate analysis was performed to compare demographic, comorbidity, and clinical characteristics across the three influenza strains. The A(H1N1)pdm09 predominant 2019-2020 influenza season in Israel was characterized by an early and steep morbidity peak, vaccine delays and shortages, and with the A(H3N2) and B/Victoria strains disproportionately targeting children and young adults, most probably due to reduced immunity to these strains. A greater proportion of children <5 years infected with A(H3N2) and B/Victoria developed severe influenza compared with those infected with A(H1N1)pdm09. Our study emphasizes the vulnerability of infants and young children in the face of rapidly evolving influenza strains and underscores the importance of influenza prevention measures in this population.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Influenza, Human , Child , Infant , Young Adult , Humans , Child, Preschool , Influenza, Human/epidemiology , Influenza A Virus, H3N2 Subtype , Seasons , Israel , Morbidity , Influenza B virus
SELECTION OF CITATIONS
SEARCH DETAIL