Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.317
Filter
1.
PLoS Pathog ; 20(7): e1012257, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38950082

ABSTRACT

An important aspect of how viruses spread and infect is the viral burst size, or the number of new viruses produced by each infected cell. Surprisingly, this value remains poorly characterized for influenza A virus (IAV), commonly known as the flu. In this study, we screened tens of thousands of cells using a microfluidic method called droplet quantitative PCR (dqPCR). The high-throughput capability of dqPCR enabled the measurement of a large population of infected cells producing progeny virus. By measuring the fully assembled and successfully released viruses from these infected cells, we discover that the viral burst sizes for both the seasonal H3N2 and the 2009 pandemic H1N1 strains vary significantly, with H3N2 ranging from 101 to 104 viruses per cell, and H1N1 ranging from 101 to 103 viruses per cell. Some infected cells produce average numbers of new viruses, while others generate extensive number of viruses. In fact, we find that only 10% of the single-cell infections are responsible for creating a significant portion of all the viruses. This small fraction produced approximately 60% of new viruses for H3N2 and 40% for H1N1. On average, each infected cell of the H3N2 flu strain produced 709 new viruses, whereas for H1N1, each infected cell produced 358 viruses. This novel method reveals insights into the flu virus and can lead to improved strategies for managing and preventing the spread of viruses.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza, Human , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/virology , Real-Time Polymerase Chain Reaction/methods , Single-Cell Analysis/methods , Animals , Madin Darby Canine Kidney Cells , Influenza A virus/genetics , Dogs , Virus Replication
2.
Emerg Microbes Infect ; 13(1): 2368202, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38970562

ABSTRACT

Influenza A viruses (IAV) impose significant respiratory disease burdens in both swine and humans worldwide, with frequent human-to-swine transmission driving viral evolution in pigs and highlighting the risk at the animal-human interface. Therefore, a comprehensive One Health approach (interconnection among human, animal, and environmental health) is needed for IAV prevention, control, and response. Animal influenza genomic surveillance remains limited in many Latin American countries, including Colombia. To address this gap, we genetically characterized 170 swine specimens from Colombia (2011-2017). Whole genome sequencing revealed a predominance of pandemic-like H1N1 lineage, with a minority belonging to H3N2 and H1N2 human seasonal-like lineage and H1N1 early classical swine lineages. Significantly, we have identified reassortant and recombinant viruses (H3N2, H1N1) not previously reported in Colombia. This suggests a broad genotypic viral diversity, likely resulting from reassortment between classical endemic viruses and new introductions established in Colombia's swine population (e.g. the 2009 H1N1 pandemic). Our study highlights the importance of a One Health approach in disease control, particularly in an ecosystem where humans are a main source of IAV to swine populations, and emphasizes the need for continued surveillance and enhanced biosecurity measures. The co-circulation of multiple subtypes in regions with high swine density facilitates viral exchange, underscoring the importance of monitoring viral evolution to inform vaccine selection and public health policies locally and globally.


Subject(s)
Evolution, Molecular , Genetic Variation , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections , Phylogeny , Swine Diseases , Animals , Swine , Colombia/epidemiology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/epidemiology , Swine Diseases/virology , Swine Diseases/epidemiology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/classification , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H1N1 Subtype/isolation & purification , One Health , Humans , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Whole Genome Sequencing , Genome, Viral , Epidemiological Monitoring , Reassortant Viruses/genetics , Reassortant Viruses/classification , Reassortant Viruses/isolation & purification , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/isolation & purification , Influenza A Virus, H1N2 Subtype/classification , Influenza, Human/virology , Influenza, Human/epidemiology
3.
PLoS Pathog ; 20(7): e1012345, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38968329

ABSTRACT

The CRISPR-Cas13 system has been proposed as an alternative treatment of viral infections. However, for this approach to be adopted as an antiviral, it must be optimized until levels of efficacy rival or exceed the performance of conventional approaches. To take steps toward this goal, we evaluated the influenza viral RNA degradation patterns resulting from the binding and enzymatic activity of mRNA-encoded LbuCas13a and two crRNAs from a prior study, targeting PB2 genomic and messenger RNA. We found that the genome targeting guide has the potential for significantly higher potency than originally detected, because degradation of the genomic RNA is not uniform across the PB2 segment, but it is augmented in proximity to the Cas13 binding site. The PB2 genome targeting guide exhibited high levels (>1 log) of RNA degradation when delivered 24 hours post-infection in vitro and maintained that level of degradation over time, with increasing multiplicity of infection (MOI), and across modern influenza H1N1 and H3N2 strains. Chemical modifications to guides with potent LbuCas13a function, resulted in nebulizer delivered efficacy (>1-2 log reduction in viral titer) in a hamster model of influenza (Influenza A/H1N1/California/04/09) infection given prophylactically or as a treatment (post-infection). Maximum efficacy was achieved with two doses, when administered both pre- and post-infection. This work provides evidence that mRNA-encoded Cas13a can effectively mitigate Influenza A infections opening the door to the development of a programmable approach to treating multiple respiratory infections.


Subject(s)
CRISPR-Cas Systems , Influenza, Human , RNA Stability , RNA, Messenger , RNA, Viral , Animals , RNA, Viral/genetics , RNA, Viral/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Humans , Influenza, Human/virology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Orthomyxoviridae Infections/virology , Antiviral Agents/pharmacology , Dogs , Cricetinae , Viral Proteins/genetics , Viral Proteins/metabolism , Mesocricetus , Madin Darby Canine Kidney Cells
4.
Arch Virol ; 169(8): 164, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38990242

ABSTRACT

Upregulation of ADAMTS-4 has been reported to have an important role in lung injury, and ADAMTS-4 expression is regulated by miR-126a-5p in abdominal aortic aneurysms. The aim of this study was to investigate whether miR-126a-5p/ADAMTS-4 plays a role in influenza-virus-induced lung injury. Lung fibroblasts were infected with H1N1 influenza virus to detect changes in miR-126a-5p and ADAMTS-4 expression, and cell viability was measured by CCK-8 assay. Inflammatory factors and matrix protease levels were examined using ELISA kits, and cell apoptosis was assessed by measuring the levels of apoptosis-related proteins. A dual luciferase assay was used to verify the regulatory relationship between miR-126a-5p and ADAMTS-4. H1N1 influenza virus reduced fibroblast viability, inhibited miR-126a-5p expression, and promoted ADAMTS-4 expression. Overexpression of miR-126a-5p attenuated the cellular inflammatory response, apoptosis, matrix protease secretion, and virus replication. Luciferase reporter assays revealed that miR-126a-5p inhibited ADAMTS-4 expression by targeting ADAMTS-4 mRNA. Further experiments showed that overexpression of ADAMTS-4 significantly reversed the inhibitory effects of miR-126a-5p on fibroblast inflammation, apoptosis, matrix protease secretion, and virus replication. Upregulation of miR-126a-5p inhibits H1N1-induced apoptosis, inflammatory factors, and matrix protease secretion, as well as virus replication in lung fibroblasts.


Subject(s)
ADAMTS4 Protein , Apoptosis , Fibroblasts , Inflammation , Influenza A Virus, H1N1 Subtype , Lung , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Fibroblasts/virology , Fibroblasts/metabolism , Humans , Lung/virology , Lung/pathology , ADAMTS4 Protein/genetics , ADAMTS4 Protein/metabolism , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/physiology , Inflammation/genetics , Cell Survival , Virus Replication , Influenza, Human/virology , Influenza, Human/genetics , Influenza, Human/metabolism , Cell Line
5.
Influenza Other Respir Viruses ; 18(6): e13342, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923314

ABSTRACT

BACKGROUND: The 2022-23 US influenza season peaked early in fall 2022. METHODS: Late-season influenza vaccine effectiveness (VE) against outpatient, laboratory-confirmed influenza was calculated among participants of the US Influenza VE Network using a test-negative design. RESULTS: Of 2561 participants enrolled from December 12, 2022 to April 30, 2023, 91 laboratory-confirmed influenza cases primarily had A(H1N1)pdm09 (6B.1A.5a.2a.1) or A(H3N2) (3C.2a1b.2a.2b). Overall, VE was 30% (95% confidence interval -9%, 54%); low late-season activity precluded estimation for most subgroups. CONCLUSIONS: 2022-23 late-season outpatient influenza VE was not statistically significant. Genomic characterization may improve the identification of influenza viruses that circulate postinfluenza peak.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza Vaccines , Influenza, Human , Outpatients , Seasons , Vaccine Efficacy , Humans , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Influenza, Human/epidemiology , Influenza, Human/immunology , Influenza, Human/virology , Adult , Male , Female , United States/epidemiology , Middle Aged , Young Adult , Adolescent , Aged , Child , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Child, Preschool , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/genetics , Outpatients/statistics & numerical data , Infant , Vaccination/statistics & numerical data , Aged, 80 and over
6.
J Mol Diagn ; 26(7): 599-612, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38901927

ABSTRACT

The high disease burden of influenza virus poses a significant threat to human health. Optimized diagnostic technologies that combine speed, sensitivity, and specificity with minimal equipment requirements are urgently needed to detect the many circulating species, subtypes, and variants of influenza at the point of need. Here, we introduce such a method using Streamlined Highlighting of Infections to Navigate Epidemics (SHINE), a clustered regularly interspaced short palindromic repeats (CRISPR)-based RNA detection platform. Four SHINE assays were designed and validated for the detection and differentiation of clinically relevant influenza species (A and B) and subtypes (H1N1 and H3N2). When tested on clinical samples, these optimized assays achieved 100% concordance with quantitative RT-PCR. Duplex Cas12a/Cas13a SHINE assays were also developed to detect two targets simultaneously. This study demonstrates the utility of this duplex assay in discriminating two alleles of an oseltamivir resistance (H275Y) mutation as well as in simultaneously detecting influenza A and human RNAse P in patient samples. These assays have the potential to expand influenza detection outside of clinical laboratories for enhanced influenza diagnosis and surveillance.


Subject(s)
CRISPR-Cas Systems , Influenza, Human , Humans , Influenza, Human/diagnosis , Influenza, Human/virology , CRISPR-Cas Systems/genetics , Sensitivity and Specificity , RNA, Viral/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Molecular Diagnostic Techniques/methods , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A virus/genetics , Influenza A virus/isolation & purification , Influenza A virus/classification
7.
Nat Commun ; 15(1): 5025, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871701

ABSTRACT

Influenza A viruses in swine have considerable genetic diversity and continue to pose a pandemic threat to humans due to a potential lack of population level immunity. Here we describe a pipeline to characterize and triage influenza viruses for their pandemic risk and examine the pandemic potential of two widespread swine origin viruses. Our analysis reveals that a panel of human sera collected from healthy adults in 2020 has no cross-reactive neutralizing antibodies against a α-H1 clade strain (α-swH1N2) but do against a γ-H1 clade strain. The α-swH1N2 virus replicates efficiently in human airway cultures and exhibits phenotypic signatures similar to the human H1N1 pandemic strain from 2009 (H1N1pdm09). Furthermore, α-swH1N2 is capable of efficient airborne transmission to both naïve ferrets and ferrets with prior seasonal influenza immunity. Ferrets with H1N1pdm09 pre-existing immunity show reduced α-swH1N2 viral shedding and less severe disease signs. Despite this, H1N1pdm09-immune ferrets that became infected via the air can still onward transmit α-swH1N2 with an efficiency of 50%. These results indicate that this α-swH1N2 strain has a higher pandemic potential, but a moderate level of impact since there is reduced replication fitness and pathology in animals with prior immunity.


Subject(s)
Ferrets , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H1N2 Subtype , Influenza, Human , Orthomyxoviridae Infections , Pandemics , Animals , Ferrets/virology , Humans , Swine , Influenza, Human/virology , Influenza, Human/epidemiology , Influenza, Human/immunology , Influenza, Human/blood , Influenza, Human/transmission , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/blood , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Swine Diseases/virology , Swine Diseases/epidemiology , Swine Diseases/immunology , Swine Diseases/transmission , Swine Diseases/blood , Female , Virus Shedding , Male , Adult , Virus Replication
8.
Biomacromolecules ; 25(7): 4281-4291, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38843459

ABSTRACT

Messenger ribonucleic acid (mRNA) vaccines, serving as a rapid and easily scalable emergency preventive measure, have played a pivotal role in preventing infectious diseases. The effectiveness of mRNA vaccines heavily relies on the delivery carrier, but the current market options are predominantly lipid nanoparticles. Their intricate preparation process and high transportation costs pose challenges for widespread use in remote areas. In this study, we harnessed FDA-approved polymer PLGA and lipid components widely employed in clinical experiments to craft a ready-to-use mRNA vaccine delivery system known as lipid-polymer hybrid nanoparticles (LPP). Following formulation optimization, the PDCD nanoparticles emerged as the most effective, showcasing exceptional mRNA delivery capabilities both in vitro and in vivo. Loading PDCD nanoparticles with mRNA encoding the H1N1 influenza virus HA antigen-fused M2e peptide enabled the successful induction of M2e-specific antibodies and T cell immune responses in immunized mice. After three rounds of vaccine immunization, the mice demonstrated weight recovery to normal levels and maintained a survival rate exceeding 80% following an encounter with the H1N1 influenza virus. The innovative mRNA delivery system that we designed demonstrates outstanding effectiveness in preventing infectious diseases, with the potential to play an even more significant role in future clinical applications.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Animals , Mice , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/chemistry , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/genetics , Nanoparticles/chemistry , mRNA Vaccines , Mice, Inbred BALB C , Female , Orthomyxoviridae Infections/prevention & control , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/administration & dosage , Humans , Influenza, Human/prevention & control , United States , Lipids/chemistry
9.
Anal Methods ; 16(27): 4582-4589, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38919038

ABSTRACT

As of now, the global COVID-19 pandemic caused by SARS-CoV-2, which began in 2019, has been effectively controlled. However, the symptoms of influenza A virus infection were similar to those of SARS-CoV-2 infection, but they required different treatment approaches. To make the detection more accurate and the treatment more targeted. We developed a system that integrates RPA and CRISPR assays, allowing for the rapid, highly specific, and sensitive detection and differentiation of SARS-CoV-2, H1N1, and H3N2. Under isothermal amplification conditions, the RPA-CRISPR Cas12a detection system achieved a detection limit as low as 5 copies per µL, demonstrating excellent specificity. The measurement time was approximately 30 minutes. The RPA-CRISPR Cas12a detection system combined with the microfluidic chip we designed to simultaneously detect three viruses, providing a potential solution for efficient and reliable diagnosis.


Subject(s)
COVID-19 , Influenza, Human , SARS-CoV-2 , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Humans , Influenza, Human/diagnosis , COVID-19/diagnosis , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H1N1 Subtype/genetics , Nucleic Acid Amplification Techniques/methods , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/genetics , CRISPR-Cas Systems/genetics , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/methods , Microfluidic Analytical Techniques/instrumentation , Limit of Detection , Molecular Diagnostic Techniques/methods , Influenza A virus/isolation & purification , Influenza A virus/genetics , Sensitivity and Specificity
10.
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
11.
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
12.
ACS Appl Mater Interfaces ; 16(26): 33897-33906, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38902962

ABSTRACT

We have developed an automated sensing system for the repeated detection of a specific microRNA (miRNA) of the influenza A (H1N1) virus. In this work, magnetic particles functionalized with DNAs, target miRNAs, and alkaline phosphate (ALP) enzymes formed sandwich structures. These particles were trapped on nickel (Ni) patterns of our sensor chip by an external magnetic field. Then, additional electrical signals from electrochemical markers generated by ALP enzymes were measured using the sensor, enabling the highly sensitive detection of target miRNA. The magnetic particles used on the sensor were easily removed by applying the opposite direction of external magnetic fields, which allowed us to repeat sensing measurements. As a proof of concept, we demonstrated the detection of miRNA-1254, one of the biomarkers for the H1N1 virus, with a high sensitivity down to 1 aM in real time. Moreover, our sensor could selectively detect the target from other miRNA samples. Importantly, our sensor chip showed reliable electrical signals even after six repeated miRNA sensing measurements. Furthermore, we achieved technical advances to utilize our sensor platform as part of an automated sensing system. In this regard, our reusable sensing platform could be utilized for versatile applications in the field of miRNA detection and basic research.


Subject(s)
Influenza A Virus, H1N1 Subtype , MicroRNAs , MicroRNAs/analysis , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H1N1 Subtype/genetics , Biosensing Techniques/methods , Biomarkers/analysis , Humans , Electrochemical Techniques/methods , Nickel/chemistry , Alkaline Phosphatase/metabolism , Alkaline Phosphatase/chemistry , Influenza, Human/diagnosis , Influenza, Human/virology
13.
BMC Infect Dis ; 24(1): 550, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824508

ABSTRACT

BACKGROUND: Influenza A virus infections can occur in multiple species. Eurasian avian-like swine influenza A (H1N1) viruses (EAS-H1N1) are predominant in swine and occasionally infect humans. A Eurasian avian-like swine influenza A (H1N1) virus was isolated from a boy who was suffering from fever; this strain was designated A/Shandong-binzhou/01/2021 (H1N1). The aims of this study were to investigate the characteristics of this virus and to draw attention to the need for surveillance of influenza virus infection in swine and humans. METHODS: Throat-swab specimens were collected and subjected to real-time fluorescent quantitative polymerase chain reaction (RT‒PCR). Positive clinical specimens were inoculated onto Madin-Darby canine kidney (MDCK) cells to isolate the virus, which was confirmed by a haemagglutination assay. Then, whole-genome sequencing was carried out using an Illumina MiSeq platform, and phylogenetic analysis was performed with MEGA X software. RESULTS: RT‒PCR revealed that the throat-swab specimens were positive for EAS-H1N1, and the virus was subsequently successfully isolated from MDCK cells; this strain was named A/Shandong-binzhou/01/2021 (H1N1). Whole-genome sequencing and phylogenetic analysis revealed that A/Shandong-binzhou/01/2021 (H1N1) is a novel triple-reassortant EAS-H1N1 lineage that contains gene segments from EAS-H1N1 (HA and NA), triple-reassortant swine influenza H1N2 virus (NS) and A(H1N1) pdm09 viruses (PB2, PB1, PA, NP and MP). CONCLUSIONS: The isolation and analysis of the A/Shandong-binzhou/01/2021 (H1N1) virus provide further evidence that EAS-H1N1 poses a threat to human health, and greater attention should be given to the surveillance of influenza virus infections in swine and humans.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human , Phylogeny , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H1N1 Subtype/classification , China/epidemiology , Humans , Male , Animals , Influenza, Human/virology , Influenza, Human/epidemiology , Dogs , Madin Darby Canine Kidney Cells , Child , Swine , Whole Genome Sequencing , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/epidemiology , Genome, Viral
14.
Virology ; 596: 110125, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38805804

ABSTRACT

Influenza viruses present a significant threat to global health. The production of a universal vaccine is considered essential due to the ineffectiveness of current seasonal influenza vaccines against mutant strains. mRNA technology offers new prospects in vaccinology, with various candidates for different infectious diseases currently in development and testing phases. In this study, we encapsulated a universal influenza mRNA vaccine. The vaccine encoded influenza hemagglutinin (HA), nucleoprotein (NP), and three tandem repeats of matrix protein 2 (3M2e). Twice-vaccinated mice exhibited strong humoral and cell-mediated immune responses in vivo. Notably, these immune responses led to a significant reduction in viral load of the lungs in challenged mice, and also conferred protection against future wild-type H1N1, H3N2, or H5N1 influenza virus challenges. Our findings suggest that this mRNA-universal vaccine strategy for influenza virus may be instrumental in mitigating the impact of future influenza pandemics.


Subject(s)
Antibodies, Viral , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H3N2 Subtype , Influenza Vaccines , Mice, Inbred BALB C , Orthomyxoviridae Infections , Viral Matrix Proteins , mRNA Vaccines , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Mice , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Antibodies, Viral/immunology , mRNA Vaccines/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Female , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Cross Protection/immunology , Viral Load , Lung/virology , Lung/immunology , Humans , Viroporin Proteins
15.
Antiviral Res ; 227: 105918, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38795911

ABSTRACT

The most widely used class of antivirals available for Influenza treatment are the neuraminidase inhibitors (NAI) Oseltamivir and Zanamivir. However, amino acid (AA) substitutions in the neuraminidase may cause reduced inhibition or high antiviral resistance. In Mexico, the current state of knowledge about NAI susceptibility is scarce, in this study we report the results of 14 years of Influenza surveillance by phenotypic and genotypic methods. A total of 255 isolates were assessed with the NAI assay, including Influenza A(H1N1)pdm09, A(H3N2) and Influenza B (IBV). Furthermore, 827 sequences contained in the GISAID platform were analyzed in search of relevant mutations.Overall, five isolates showed highly reduced inhibition or reduced inhibition to Oseltamivir, and two showed reduced inhibition to Zanamivir in the NAI assays. Additionally, five A(H1N1)pdm09 sequences from the GISAID possessed AA substitutions associated to reduced inhibition to Oseltamivir and none to Zanamivir. Oseltamivir resistant A(H1N1)pdm09 harbored the H275Y mutation. No genetic mutations were identified in Influenza A(H3N2) and IBV. Overall, these results show that in Mexico the rate of NAI resistance is low (0.6%), but it is essential to continue the Influenza surveillance in order to understand the drug susceptibility of circulating strains.


Subject(s)
Antiviral Agents , Drug Resistance, Viral , Influenza B virus , Influenza, Human , Neuraminidase , Oseltamivir , Zanamivir , Drug Resistance, Viral/genetics , Antiviral Agents/pharmacology , Mexico/epidemiology , Humans , Influenza B virus/drug effects , Influenza B virus/genetics , Influenza, Human/virology , Influenza, Human/drug therapy , Influenza, Human/epidemiology , Oseltamivir/pharmacology , Zanamivir/pharmacology , Neuraminidase/genetics , Neuraminidase/antagonists & inhibitors , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Mutation , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/genetics , Adult , Influenza A virus/drug effects , Influenza A virus/genetics , Adolescent , Child , Amino Acid Substitution , Young Adult , Middle Aged , Female , Child, Preschool , Genotype , Male , Aged , Microbial Sensitivity Tests , Viral Proteins/genetics
17.
Sci Rep ; 14(1): 10436, 2024 05 07.
Article in English | MEDLINE | ID: mdl-38714669

ABSTRACT

Influenza (sometimes referred to as "flu") is a contagious viral infection of the airways in the lungs that affects a significant portion of the world's population. Clinical symptoms of influenza virus infections can range widely, from severe pneumonia to moderate or even asymptomatic sickness. If left untreated, influenza can have more severe effects on the heart, brain, and lungs than on the respiratory tract and can necessitate hospitalization. This study was aimed to investigate and characterize all types of influenza cases prevailing in Nepal and to analyze seasonal occurrence of Influenza in Nepal in the year 2019. A cross sectional, retrospective and descriptive study was carried out at National Influenza Center (NIC), National Public Health Laboratory Kathmandu Nepal for the period of one year (Jan-Dec 2019). A total of 3606 throat swab samples from various age groups and sexes were processed at the NIC. The specimens were primarily stored at 4 °C and processed using ABI 7500 RT PCR system for the identification of Influenza virus types and subtypes. Data accessed for research purpose were retrieved from National Influenza Centre (NIC) on 1st Jan 2020. Of the total 3606 patients suspected of having influenza infection, influenza viruses were isolated from 1213 (33.6%) patients with male predominance. The highest number of infection was caused by Influenza A/Pdm09 strain 739 (60.9%) followed by Influenza B 304 (25.1%) and Influenza A/H3 169 (13.9%) and most remarkable finding of this study was the detection of H5N1 in human which is the first ever case of such infection in human from Nepal. Similar to other tropical nations, influenza viruses were detected year-round in various geographical locations of Nepal. The influenza virus type and subtypes that were in circulation in Nepal were comparable to vaccine candidate viruses, which the currently available influenza vaccine may prevent.


Subject(s)
Influenza, Human , Humans , Nepal/epidemiology , Influenza, Human/epidemiology , Influenza, Human/virology , Female , Male , Child , Adult , Adolescent , Middle Aged , Child, Preschool , Infant , Retrospective Studies , Young Adult , Cross-Sectional Studies , Aged , Influenza B virus/genetics , Influenza B virus/isolation & purification , Seasons , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/isolation & purification
18.
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
19.
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
20.
Vet Res ; 55(1): 65, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773540

ABSTRACT

In 2020, a new genotype of swine H1N2 influenza virus (H1avN2-HA 1C.2.4) was identified in France. It rapidly spread within the pig population and supplanted the previously predominant H1avN1-HA 1C.2.1 virus. To characterize this new genotype which is genetically and antigenically distant from the other H1avNx viruses detected in France, an experimental study was conducted to compare the outcomes of H1avN2 and H1avN1 infections in pigs and evaluate the protection conferred by the only inactivated vaccine currently licensed in Europe containing an HA 1C (clade 1C.2.2) antigen. Infection with H1avN2 induced stronger clinical signs and earlier shedding than H1avN1. The neutralizing antibodies produced following H1avN2 infection were unable to neutralize H1avN1, and vice versa, whereas the cellular-mediated immunity cross-reacted. Vaccination slightly altered the impact of H1avN2 infection at the clinical level, but did not prevent shedding of infectious virus particles. It induced a cellular-mediated immune response towards H1avN2, but did not produce neutralizing antibodies against this virus. As in vaccinated animals, animals previously infected by H1avN1 developed a cross-reacting cellular immune response but no neutralizing antibodies against H1avN2. However, H1avN1 pre-infection induced a better protection against the H1avN2 infection than vaccination, probably due to higher levels of non-neutralizing antibodies and a mucosal immunity. Altogether, these results showed that the new H1avN2 genotype induced a severe respiratory infection and that the actual vaccine was less effective against this H1avN2-HA 1C.2.4 than against H1avN1-HA 1C.2.1, which may have contributed to the H1avN2 epizootic and dissemination in pig farms in France.


Subject(s)
Genotype , Influenza A Virus, H1N2 Subtype , Orthomyxoviridae Infections , Swine Diseases , Animals , Swine , Swine Diseases/virology , Swine Diseases/immunology , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/immunology , France/epidemiology , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza Vaccines/immunology , Virulence , Antibodies, Neutralizing/blood , Immunity, Cellular
SELECTION OF CITATIONS
SEARCH DETAIL