Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.215
Filtrar
1.
Front Immunol ; 15: 1322879, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38482020

RESUMO

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


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza , Vírus do Orf , Infecções por Orthomyxoviridae , Animais , Suínos , Hemaglutininas/genética , Infecções por Orthomyxoviridae/prevenção & controle , Vírus da Influenza A Subtipo H1N1/genética , Anticorpos Antivirais , Imunoglobulina G , Epitopos
2.
Emerg Microbes Infect ; 13(1): 2284294, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37966008

RESUMO

H5N1 avian influenza viruses bearing the clade 2.3.2.1 hemagglutinin (HA) gene have been widely detected in birds and poultry in several countries. During our routine surveillance, we isolated 28 H5N1 viruses between January 2017 and October 2020. To investigate the genetic relationship of the globally circulating H5N1 viruses and the biological properties of those detected in China, we performed a detailed phylogenic analysis of 274 representative H5N1 strains and analyzed the antigenic properties, receptor-binding preference, and virulence in mice of the H5N1 viruses isolated in China. The phylogenic analysis indicated that the HA genes of the 274 viruses belonged to six subclades, namely clades 2.3.2.1a to 2.3.2.1f; these viruses acquired gene mutations and underwent complicated reassortment to form 58 genotypes, with G43 being the dominant genotype detected in eight Asian and African countries. The 28 H5N1 viruses detected in this study carried the HA of clade 2.3.2.1c (two strains), 2.3.2.1d (three strains), or 2.3.2.1f (23 strains), and formed eight genotypes. These viruses were antigenically well-matched with the H5-Re12 vaccine strain used in China. Animal studies showed that the pathogenicity of the H5N1 viruses ranged from non-lethal to highly lethal in mice. Moreover, the viruses exclusively bound to avian-type receptors and have not acquired the ability to bind to human-type receptors. Our study reveals the overall picture of the evolution of clade 2.3.2.1 H5N1 viruses and provides insights into the control of these viruses.


Assuntos
Virus da Influenza A Subtipo H5N1 , Influenza Aviária , Animais , Humanos , Camundongos , Hemaglutininas/genética , Aves , Aves Domésticas , Filogenia , Galinhas , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química
3.
Genes (Basel) ; 14(10)2023 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-37895321

RESUMO

The avian influenza viruses (AIV) of the H5 subtype have the ability to mutate from low pathogenic (LPAI) to highly pathogenic (HPAI), which can cause high mortality in poultry. Little is known about the pathogenic switching apart from the mutations at the haemagglutinin cleavage site, which significantly contributes to the virus virulence switching phenomenon. Therefore, this study aimed to compare the molecular markers in the haemagglutinin (HA), neuraminidase (NA), and matrix (M) genes of a locally isolated LPAI AIV strain H5N2 from Malaysia with the reference HPAI strains using bioinformatics approaches, emphasising the pathogenic properties of the viral genes. First, the H5N2 strain A/Duck/Malaysia/8443/2004 was propagated in SPF eggs. The viral presence was verified by haemagglutination assay, RT-PCR, and sequencing. Results showed successful amplifications of HA (1695 bp), NA (1410 bp), and M (1019 bp) genes. The genes were sequenced and the deduced amino acid sequences were analysed computationally using MEGA 11 and NetNGlyc software. Analysis of the HA protein showed the absence of the polybasic cleavage motif, but presence of two amino acid residues that are known to affect pathogenicity. There were also two glycosylation sites (glycosites) compared to the reference HPAI viruses, which had three or more at the HA globular head domain. No NA stalk deletion was detected but the haemadsorbing and active centres of the studied NA protein were relatively similar to the reference HPAI H5N2 isolates of duck but not chicken origins. Six NA glycosites were also identified. Finally, we observed a consistent M1 and M2 amino acid sequences between our LPAI isolate with the other HPAI H5N1 or H5N2 reference proteins. These data demonstrate distinct characteristics of the Malaysian LPAI H5N2, compared to HPAI H5N2 or H5N1 from ducks or chickens, potentially aiding the epidemiological research on genetic dynamics of circulating AIV in poultry.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A Subtipo H5N2 , Vírus da Influenza A , Influenza Aviária , Animais , Patos/genética , Vírus da Influenza A Subtipo H5N2/genética , Influenza Aviária/genética , Galinhas/genética , Virus da Influenza A Subtipo H5N1/genética , Hemaglutininas/genética , Aves Domésticas/genética , Análise de Sequência
4.
J Biol Chem ; 299(12): 105364, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37865319

RESUMO

Nucleoid-associated proteins (NAPs) regulate multiple cellular processes such as gene expression, virulence, and dormancy throughout bacterial species. NAPs help in the survival and adaptation of Mycobacterium tuberculosis (Mtb) within the host. Fourteen NAPs have been identified in Escherichia coli; however, only seven NAPs are documented in Mtb. Given its complex lifestyle, it is reasonable to assume that Mtb would encode for more NAPs. Using bioinformatics tools and biochemical experiments, we have identified the heparin-binding hemagglutinin (HbhA) protein of Mtb as a novel sequence-independent DNA-binding protein which has previously been characterized as an adhesion molecule required for extrapulmonary dissemination. Deleting the carboxy-terminal domain of HbhA resulted in a complete loss of its DNA-binding activity. Atomic force microscopy showed HbhA-mediated architectural modulations in the DNA, which may play a regulatory role in transcription and genome organization. Our results showed that HbhA colocalizes with the nucleoid region of Mtb. Transcriptomics analyses of a hbhA KO strain revealed that it regulates the expression of ∼36% of total and ∼29% of essential genes. Deletion of hbhA resulted in the upregulation of ∼73% of all differentially expressed genes, belonging to multiple pathways suggesting it to be a global repressor. The results show that HbhA is a nonessential NAP regulating gene expression globally and acting as a plausible transcriptional repressor.


Assuntos
Proteínas de Bactérias , Hemaglutininas , Mycobacterium tuberculosis , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , DNA/química , DNA/metabolismo , Hemaglutininas/genética , Hemaglutininas/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Regulação Bacteriana da Expressão Gênica/genética , Deleção de Genes , Proteínas de Ligação a DNA/genética , Domínios Proteicos/genética , Microscopia de Força Atômica
5.
Vet Microbiol ; 285: 109872, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37690146

RESUMO

Vaccines are widely used to prevent Newcastle disease virus (NDV). Under the pressure of immunization, NDVs with mutations among epitopes of F and HN protein were isolated, which indicates that the efficiency of vaccine may decrease in terms of preventing emerged NDV. However, the lack of evidences to support whether these mutations contribute to antigenic mutation and immune escape in NDV leading to the controversy that the matched vaccine is more effective than the mismatched vaccine. In this study, a genotype VII velogenic NDV strain (C22) was isolated from a vaccinated farm in Tibet, China. We found that this strain was close to NDV from east China, but it had a specific mutation (K138R) in one epitope (131DYIGGIGKE139) of HN protein. This mutation might change the interaction between amino acids in stalk-head link region of HN protein and then induce the specific antibody to worse recognize the C22 strain, but it did not alter viral virulence and growth ability. Then, the C22 strain was attenuated via modification of the F protein cleavage site to generate a matched vaccine. Comparing to a mismatched vaccine (LaSota), this matched vaccine showed advantages in inhibiting viral shedding and tissue damage. However, both vaccines induced chicken to generate similar level of neutralizing antibodies against C22, C22mut (R138K) and LaSota. These results suggest that the epitope mutation is insufficient to help NDV escaping neutralizing antibodies of vaccinated chicken, supporting that the merits of NDV matched vaccine are not totally related to humoral immunity.


Assuntos
Doença de Newcastle , Vacinas Virais , Animais , Vírus da Doença de Newcastle , Hemaglutininas/genética , Neuraminidase/genética , Tibet , Proteína HN/genética , Vacinas Virais/genética , Galinhas , Proteínas Virais/genética , Anticorpos Neutralizantes/genética , China , Variação Antigênica , Epitopos/genética , Anticorpos Antivirais , Genótipo
6.
Viruses ; 15(9)2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37766279

RESUMO

Canine distemper (CD), caused by canine distemper virus (CDV), is a highly contagious and lethal disease in domestic and wild carnivores. Although CDV live-attenuated vaccines have reduced the incidence of CD worldwide, low levels of protection are achieved in the presence of maternal antibodies in juvenile animals. Moreover, live-attenuated CDV vaccines may retain residual virulence in highly susceptible species and cause disease. Here, we generated several CDV DNA vaccine candidates based on the biscistronic vector (pIRES) co-expressing virus wild-type or codon-optimized hemagglutinin (H) and nucleocapsid (N) or ferret interferon (IFN)-γ, as a molecular adjuvant, respectively. Apparently, ferret (Mustela putorius furo)-specific codon optimization increased the expression of CDV H and N proteins. A ferret model of CDV was used to evaluate the protective immune response of the DNA vaccines. The results of the vaccinated ferrets showed that the DNA vaccine co-expressing the genes of codon-optimized H and ferret IFN-γ (poptiH-IRES-IFN) elicited the highest anti-CDV serum-neutralizing antibodies titer (1:14) and cytokine responses (upregulated TNF-α, IL-4, IL-2, and IFN-γ expression) after the third immunization. Following vaccination, the animals were challenged with a lethal CDV 5804Pe/H strain with a dose of 105.0 TCID50. Protective immune responses induced by the DNA vaccine alleviated clinical symptoms and pathological changes in CDV-infected ferrets. However, it cannot completely prevent virus replication and viremia in vivo as well as virus shedding due to the limited neutralizing antibody level, which eventually contributed to a survival rate of 75% (3/4) against CDV infection. Therefore, the improved strategies for the present DNA vaccines should be taken into consideration to develop more protective immunity, which includes increasing antigen expression or alternative delivery routes, such as gene gun injection.


Assuntos
Vírus da Cinomose Canina , Cinomose , Vacinas de DNA , Animais , Cães , Furões , Vacinas de DNA/genética , Hemaglutininas/genética , Vírus da Cinomose Canina/genética , Interferon gama , Anticorpos Neutralizantes , Cinomose/prevenção & controle
7.
Emerg Microbes Infect ; 12(2): 2249558, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37585307

RESUMO

H9N2 avian influenza viruses (AIVs) pose an increasing threat to the poultry industry worldwide and have pandemic potential. Vaccination has been principal prevention strategy to control H9N2 in China since 1998, but vaccine effectiveness is persistently challenged by the emergence of the genetic and/or antigenic variants. Here, we analysed the genetic and antigenic characteristics of H9N2 viruses in China, including 70 HA sequences of H9N2 isolates from poultry, 7358 from online databases during 2010-2020, and 15 from the early reference strains. Bayesian analyses based on hemagglutinin (HA) gene revealed that a new designated clade16 emerged in April 2012, and was prevalent and co-circulated with clade 15 since 2013 in China. Clade 16 viruses exhibited decreased cross-reactivity with those from clade 15. Antigenic Cartography analyses showed represent strains were classified into three antigenic groups named as Group1, Group2 and Group3, and most of the strains in Group 3 (15/17, 88.2%) were from Clade 16 while most of the strains in Group2 (26/29, 89.7%) were from Clade 15. The mean distance between Group 3 and Group 2 was 4.079 (95%CI 3.605-4.554), revealing that major switches to antigenic properties were observed over the emergence of clade 16. Genetic analysis indicated that 11 coevolving amino acid substitutions primarily at antigenic sites were associated with the antigenic differences between clade 15 and clade 16. These data highlight complexities of the genetic evolution and provide a framework for the genetic basis and antigenic characterization of emerging clade 16 of H9N2 subtype avian influenza virus.


Assuntos
Vírus da Influenza A Subtipo H9N2 , Influenza Aviária , Animais , Influenza Aviária/epidemiologia , Hemaglutininas/genética , Deriva e Deslocamento Antigênicos , Teorema de Bayes , Galinhas , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Aves Domésticas , China/epidemiologia , Filogenia
8.
Arch Virol ; 168(8): 203, 2023 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-37418014

RESUMO

The hemagglutinin-neuraminidase (HN) protein of Newcastle disease virus (NDV) is a multifunctional protein with receptor recognition ability that plays an important role in the infection of cells by NDV. An alignment of NDV HN protein sequences of different genotypes showed that vaccine strains of NDV, such as the LaSota strain, generally have an HN protein of 577 amino acids. In comparison, the HN protein of the V4 strain has 616 amino acids, with 39 more amino acids at the C-terminus. In this study, we generated a recombinant NDV (rNDV) with a 39-amino-acid truncation at the HN C-terminus based on the full-length cDNA clone of the V4 strain. This rNDV, named rV4-HN-tr, displayed thermostability similar to that of the parental V4 strain. However, growth kinetics and pathogenicity analysis suggested that rV4-HN-tr is more virulent than the V4 strain. Notably, the C-terminus of HN affected the ability of the virus to adsorb onto cells. Structural predictions further suggested that the C-terminus of HN may obstruct the sialic acid binding site. Immunization of chickens with rV4-HN-tr induced a 3.5-fold higher level of NDV-specific antibodies than that obtained with the V4 strain and provided 100% immune protection against NDV challenge. Our study suggests that rV4-HN-tr is a thermostable, safe, and highly efficient vaccine candidate against Newcastle disease.


Assuntos
Doença de Newcastle , Vacinas Virais , Animais , Vírus da Doença de Newcastle , Galinhas , Virulência , Neuraminidase/genética , Hemaglutininas/genética , Proteína HN/genética , Proteína HN/metabolismo , Vacinas Virais/genética , Anticorpos Antivirais , Aminoácidos
9.
J Cancer Res Clin Oncol ; 149(12): 9903-9918, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37249647

RESUMO

BACKGROUND: Canine distemper virus (CDV) has been shown to have oncolytic activity against primary canine tumors. Previous studies from this laboratory had confirmed that CDV induces apoptosis in canine mammary tumor (CMT) cells, although the molecular mechanism remains unknown. METHODS: The CDV N, P, M, F, H, L, C, and V genes were identified in CDV-L and cloned separately. Mutants with deletions in the 5' region (pCMV-F L△60, pCMV-FL△107, and pCMV-FL△114) or with site-directed mutagenesis in the 3' region (pCMV-FLA602-610) of the F gene were generated. Late-stage apoptotic cells were detected by Hoechst 33342. Early-stage apoptotic cells were detected by AnnexinV-FITC/PI. Quantitative real-time PCR was performed to detect the mRNA levels of target genes of apoptotic and NF-κB pathway. Western blot analysis was performed to detect the expression or phosphorylation levels of target proteins of apoptotic or NF-κB pathway. Immunofluorescence assay was performed to detect the nuclear translocation of p65 protein. Recombinant viruses (rCDV-FL△60 and rCDV-FLA602-610) were rescued by a BHK-T7-based system. 5-week-old female BALB/c nude mice were used to detect the oncolytic activity of recombinant viruses. RESULTS: In this study, it was first confirmed that none of the structural or non-structural proteins of CDV-L, a vaccine strain, was individually able to induce apoptosis in canine mammary tubular adenocarcinoma cells (CIPp) or intraductal papillary carcinoma cells (CMT-7364). However, when CIPp or CMT-7364 cells were co-transfected with glycoprotein fusion (F) and hemagglutinin (H) proteins of CDV-L, nuclear fragmentation was observed and a high proportion of early apoptotic cells were detected, as well as cleaved caspase-3, caspase-8 and poly (ATP ribose) polymerase (PARP). Cleaved caspase-3 and PARP were down-regulated by apoptosis broad-spectrum inhibitor Z-VAD-FMK and caspase-8 pathway inhibitor Z-IETD-FMK, confirming that the F and H proteins coinduced apoptosis in CMT cells via the caspase-8 and caspase-3 pathways. F and H proteins co-induced phosphorylation of p65 and IκBα and nuclear translocation of p65, confirming activation of the NF-κB pathway, inhibition of which down-regulated cleaved caspase-3 and cleaved PARP. Recombinant F protein with enhanced fusion activity and H protein co-induced more cleaved caspase-3 and PARP than parental F protein, while the corresponding recombinant virus exhibited the same properties both in CIPp cells and in a subcutaneous xenograft mouse model. CONCLUSIONS: F and H proteins of CDV-L co-induce apoptosis in CMT cells, while the NF-κB pathway and fusion activity of F protein paly essential roles in the process.


Assuntos
Neoplasias da Mama , Vírus da Cinomose Canina , Feminino , Animais , Cães , Humanos , Camundongos , Caspase 3 , Vírus da Cinomose Canina/genética , Hemaglutininas/genética , Caspase 8 , NF-kappa B , Camundongos Nus , Inibidores de Poli(ADP-Ribose) Polimerases , Apoptose
10.
Arch Microbiol ; 205(6): 253, 2023 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-37254016

RESUMO

Newcastle disease has been endemic within the Iranian poultry industry for decades. However, the genetic nature of the circulating Hemagglutinin-Neuraminidase (HN) gene among Iranian domesticated bird populations is broadly unexplored. The presented study was carried out to gain insights into the biological and molecular characterization of four complete HN genes isolated from turkey, peacock, and broiler isolates in Iran between 2018 and 2020. The phylogenetic analysis revealed that the isolates belong to the Newcastle disease virus (NDV) subgenotype VII.1.1, previously known as VIIL. Further analysis demonstrated the thermostable substitutions S315P and I369V within the isolates. Finding the N-glycosylation site (NIS) at positions 144-146 and the cysteine residue 123 might influence the fusogenicity abilities of the isolates, while identification of multiple amino acid substitutions in both antigenic sites, especially I514V and E347Q, and the binding sites of the HN protein, raised concern about the pathogenicity of the isolates. In addition, the annual rate of change based on the HN gene of Iranian NDV was calculated at about 1.8088E-3 between 2011 and 2020. In conclusion, a new NDV variant with multiple site mutagenesis is circulating not only among chickens but also in turkey and captive birds such as peafowls, and failure of routine vaccination programs could be attributed to the differences between circulating NDV strains and those used in vaccine manufacturing. Therefore, future legislation aimed at providing vaster vaccination cover and biosecurity plans will be needed to control the spread of circulating NDV strains.


Assuntos
Galinhas , Vírus da Doença de Newcastle , Animais , Vírus da Doença de Newcastle/genética , Filogenia , Neuraminidase , Hemaglutininas/genética , Irã (Geográfico) , Genótipo , Proteínas Virais/genética
11.
PLoS One ; 18(1): e0280811, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36662890

RESUMO

Manufactured influenza vaccines have to contain a defined amount of hemagglutinin (HA) antigen. Therefore, vaccine viruses with a high HA antigen yield (HAY) are preferable for manufacturing vaccines, particularly vaccines in response to a pandemic, when vaccines need to be rapidly produced. However, the viral properties associated with a high HAY have not yet been fully clarified. To identify the HAY-associated traits, we first propagated 26 H5 candidate vaccine viruses (CVVs) in eggs, which were previously developed based on genetic reassortment methods using master viruses, to determine their total protein yield (TPY), ratio of HA to total viral protein (%-HA content) and HAY. The results revealed that the HAY was correlated with the TPY but not with the %-HA content. We further found that altering the sequences of the 3' noncoding region of HA vRNA or replacing the master virus improved the HAYs and TPYs of the low-HAY CVVs to approximately double the values of the original CVVs but did not change the %-HA content, which a previous study suggested was associated with the HAY. Analyses based on real-time PCR assays and scanning electron microscopy revealed that the virus samples with an improved HAY contained more copies of the virus genome and viral particles than the original samples. The results suggest that an improvement in virus growth (i.e., an increase in the amount of viral particles) leads to an increase in the TPY and thus in the HAY, regardless of the %-HA content. The approximately twofold increase in the HAY shown in this study may not appear to represent a large improvement, but the impact will be significant given the millions of chicken eggs used to produce vaccines. These findings will be informative for developing high-HAY vaccine viruses.


Assuntos
Vacinas contra Influenza , Orthomyxoviridae , Animais , Hemaglutininas/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Galinhas , Anticorpos Antivirais
12.
Int J Infect Dis ; 125: 114-119, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36283676

RESUMO

OBJECTIVES: We evaluated the VE and the mutations of the viruses present in the Mexican population at the beginning of 2018. METHODS: We diagnosed influenza in outpatients with a high-performance Rapid Influenza Diagnostic Test (RIDT) qRT-PCR. Descriptive statistics were used to describe the study population, while the chi-square test was used to determine clinical variables. VE was analyzed through a negative test design. We sequenced the hemagglutinin (HA) gene, performed a phylogenetic analysis, and analyzed the nonsynonymous substitutions both in and outside antigenic sites. RESULTS: Of the 240 patients analyzed, 42.5% received the trivalent vaccine, and 37.5% were positive for influenza. The VE for the general population for any influenza virus type or subtype was 37.0%, while the VE for the predominant influenza A(H3N2) subtype was the lowest (19.7%). The phylogenetic analysis of HA showed the co-circulation of clades and subclades 3C.2a1, 3C.2a1b, 3C.2a2, 3C.2a2re, 3C.2a3, and 3C.3a with identities approximately 97-98% similar to the vaccine composition. CONCLUSION: Low VE was related to the co-circulation of multiple clades and subclades of influenza A(H3N2), with sufficient genetic and phenotypic distance to allow for the infection of vaccinated individuals.


Assuntos
Vacinas contra Influenza , Influenza Humana , Humanos , Influenza Humana/diagnóstico , Influenza Humana/epidemiologia , Influenza Humana/prevenção & controle , Vírus da Influenza A Subtipo H3N2/genética , Filogenia , Estações do Ano , México/epidemiologia , Eficácia de Vacinas , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , RNA Viral/genética , Variação Antigênica , Hemaglutininas/genética
13.
Front Immunol ; 13: 974210, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36275684

RESUMO

The zoonotic H7N9 avian influenza (AI) virus first emerged in 2013 as a low pathogenic (LPAI) strain, and has repeatedly caused human infection resulting in severe respiratory illness and a mortality of ~39% (>600 deaths) across five epidemic waves. This virus has circulated in poultry with little to no discernible clinical signs, making detection and control difficult. Contrary to published data, our group has observed a subset of specific pathogen free chickens infected with the H7N9 virus succumb to disease, showing clinical signs consistent with highly pathogenic AI (HPAI). Viral genome sequencing revealed two key mutations had occurred following infection in the haemagglutinin (HA 226 L>Q) and nucleoprotein (NP 373 A>T) proteins. We further investigated the impact of the NP mutation and demonstrated that only chickens bearing a single nucleotide polymorphism (SNP) in their IFITM1 gene were susceptible to the H7N9 virus. Susceptible chickens demonstrated a distinct loss of CD8+ T cells from the periphery as well as a dysregulation of IFNγ that was not observed for resistant chickens, suggesting a role for the NP mutation in altered T cell activation. Alternatively, it is possible that this mutation led to altered polymerase activity, as the mutation occurs in the NP 360-373 loop which has been previously show to be important in RNA binding. These data have broad ramifications for our understanding of the pathobiology of AI in chickens and humans and provide an excellent model for investigating the role of antiviral genes in a natural host species.


Assuntos
Subtipo H7N9 do Vírus da Influenza A , Influenza Aviária , Animais , Humanos , Influenza Aviária/genética , Influenza Aviária/epidemiologia , Subtipo H7N9 do Vírus da Influenza A/genética , Galinhas/genética , Hemaglutininas/genética , Nucleoproteínas/genética , Linfócitos T CD8-Positivos/patologia , Mutação , Antivirais , RNA
14.
Sci Rep ; 12(1): 17089, 2022 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-36224196

RESUMO

Influenza A(H3N2) virus exhibited complex seasonal patterns to evade pre-existing antibodies, resulting in changes in the antigenicity of the viron surface protein hemagglutinin (HA). To monitor the currently imported influenza viruses as well as to assess the capacity of health emergencies at the Shanghai port, we collected respiratory specimens of passengers from different countries and regions including some of Europe with influenza-like illness at the Shanghai port during 2016/2017, examined amino acid substitutions, and calculated the perfect-match vaccine efficacy using the p epitope model. Phylogenetic analysis of the HA genes revealed that influenza A(H3N2) viruses belonging to eight subclades were detected, and three amino acid substitutions in the subclade 3C.2a.4 were also added. Besides, two epidemic influenza virus strains were found in the 2016/2017 winter and 2016 summer. The results of lower predicted vaccine effectiveness in summer suggest that the imported A(H3N2) strains were not a good match for the A/Hong Kong/4801/2014 vaccine strain since the summer of 2017. Therefore, the Shanghai Port might stop the risk of the international spread of influenza for the first time, and curb the entry of A(H3N2) from overseas at the earliest stage of a probable influenza pandemic.


Assuntos
Vírus da Influenza A , Vacinas contra Influenza , Influenza Humana , China/epidemiologia , Epitopos , Variação Genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Hemaglutininas/genética , Humanos , Vírus da Influenza A Subtipo H3N2 , Influenza Humana/epidemiologia , Influenza Humana/prevenção & controle , Proteínas de Membrana/genética , Filogenia , Estações do Ano
15.
Viruses ; 14(10)2022 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-36298794

RESUMO

Adaptive laboratory evolution has been used to improve production of influenza hemagglutinin (HA)-displaying virus-like particles (VLPs) in insect cells. However, little is known about the underlying biological mechanisms promoting higher HA-VLP expression in such adapted cell lines. In this article, we present a study of gene expression patterns associated with high-producer insect High Five cells adapted to neutral pH, in comparison to non-adapted cells, during expression of influenza HA-VLPs. RNA-seq shows a decrease in the amount of reads mapping to host cell genomes along infection, and an increase in those mapping to baculovirus and transgenes. A total of 1742 host cell genes were found differentially expressed between adapted and non-adapted cells throughout infection, 474 of those being either up- or down-regulated at both time points evaluated (12 and 24 h post-infection). Interestingly, while host cell genes were found up- and down-regulated in an approximately 1:1 ratio, all differentially expressed baculovirus genes were found to be down-regulated in infected adapted cells. Pathway analysis of differentially expressed genes revealed enrichment of ribosome biosynthesis and carbohydrate, amino acid, and lipid metabolism. In addition, oxidative phosphorylation and protein folding, sorting and degradation pathways were also found to be overrepresented. These findings contribute to our knowledge of biological mechanisms of insect cells during baculovirus-mediated transient expression and will assist the identification of potential engineering targets to increase recombinant protein production in the future.


Assuntos
Vacinas contra Influenza , Influenza Humana , Animais , Humanos , Hemaglutininas/genética , Baculoviridae/genética , Insetos/genética , Proteínas Recombinantes/genética , Expressão Gênica , Aminoácidos/genética , Carboidratos , RNA
16.
Biosystems ; 220: 104740, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35934256

RESUMO

Influenza viruses mutate rapidly and can pose a threat to public health, especially to those in vulnerable groups. Throughout history, influenza A viruses have caused pandemics between different species. It is important to identify the origin of a virus in order to prevent the spread of an outbreak. Recently, there has been increasing interest in using machine learning algorithms to provide fast and accurate predictions for viral sequences. In this study, real testing data sets and a variety of evaluation metrics were used to evaluate machine learning algorithms at different taxonomic levels. As hemagglutinin is the major protein in the immune response, only hemagglutinin sequences were used and represented by position-specific scoring matrix and word embedding. The results suggest that the 5-grams-transformer neural network is the most effective algorithm for predicting viral sequence origins, with approximately 99.54% AUCPR, 98.01% F1 score and 96.60% MCC at a higher classification level, and approximately 94.74% AUCPR, 87.41% F1 score and 80.79% MCC at a lower classification level.


Assuntos
Vírus da Influenza A , Orthomyxoviridae , Algoritmos , Hemaglutininas/genética , Vírus da Influenza A/genética , Aprendizado de Máquina
17.
J Vet Diagn Invest ; 34(5): 874-878, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35879873

RESUMO

Rapid and reliable identification of the hemagglutinin (HA) and neuraminidase (NA) genetic clades of an influenza A virus (IAV) sequence from swine can inform control measures and multivalent vaccine composition. Current approaches to genetically characterize HA or NA sequences are based on nucleotide similarity or phylogenetic analyses. Public databases exist to acquire IAV genetic sequences for comparison, but personnel at the diagnostic or production level have difficulty in adequately updating and maintaining relevant sequence datasets for IAV in swine. Further, phylogenetic analyses are time intensive, and inference drawn from these methods is impacted by input sequence data and associated metadata. We describe here the use of the IAV multisequence identity tool as an integrated public webpage located on the Iowa State University Veterinary Diagnostic Laboratory (ISU-VDL) FLUture website: https://influenza.cvm.iastate.edu/. The multisequence identity tool uses sequence data derived from IAV-positive cases sequenced at the ISU-VDL, employs a BLAST algorithm that identifies sequences that are genetically similar to submitted query sequences, and presents a tabulation and visualization of the most genetically similar IAV sequence and associated metadata from the FLUture database. Our tool removes bioinformatic barriers and allows clients, veterinarians, and researchers to rapidly classify and identify IAV sequences similar to their own sequences to augment interpretation of results.


Assuntos
Vírus da Influenza A , Infecções por Orthomyxoviridae , Doenças dos Suínos , Animais , Hemaglutininas/genética , Humanos , Vírus da Influenza A/genética , Neuraminidase/genética , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Orthomyxoviridae/veterinária , Filogenia , Suínos , Doenças dos Suínos/diagnóstico , Doenças dos Suínos/epidemiologia , Estados Unidos/epidemiologia
18.
Trop Biomed ; 39(2): 160-169, 2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35838085

RESUMO

Highly Pathogenic Avian Influenza (HPAI) is a highly contagious disease in poultry. The outbreaks can lead to flock mortality up to 100% in two to three days. In July 2018, high mortality in a commercial layer farm in Kauluan village, Sabah was reported. Samples were sent to Veterinary Research Institute Ipoh for diagnosis. Virus isolation and molecular detection is carried out simultaneously. The causative agent was then identified as AI H5N1 virus by real time reverse transcription-polymerase chain reaction (RT-PCR). The virus was then subjected for further nucleotide sequencing of full length hemagglutinin (HA) and neuraminidase (NA) gene. The PQRERRRKR/GLF motif at the HA cleavage site indicated that the isolate was of HPAI virus. Phylogenetic analysis of the HA gene showed that the isolate was belonged to the clade 2.3.2.1c virus. In the HA gene, besides the S133A substitution, the virus possesses conserved amino acid at most of the avian receptor binding sites including the glutamine (Q) and glycine (G) at position 222 and 224 respectively, indicating that the virus retains the avian-type receptor binding preference. As such, the zoonotic potential of the virus was relatively low. On the other hand, though the N154D and T156A substitution were detected in the same gene, the pandemic potential of this Sabah 2.3.2.1c virus is low in the absence of the Q222L, G224S, H103Y, N220K and T315I. A typical 20 amino acid deletion with loss of four corresponding glycosylation sites in the NA stalk region was visible. Though three NA resistance markers were detected, the virus was predicted to be sensitive to NA inhibitor. This is the first HPAI H5N1 outbreak in Sabah. The introduction of this virus into East Malaysia for the first time raised an alert alarm of the future epidemic potential. Strict farm biosecurity, continuous surveillance programme in poultry, wild birds, migratory birds; molecular epidemiology as well as risk assessment for the virus with pandemic potential are needed in dealing with emergence of new influenza virus in the country.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A , Influenza Aviária , Aminoácidos/genética , Animais , Aves , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Hemaglutininas/genética , Humanos , Virus da Influenza A Subtipo H5N1/genética , Influenza Aviária/epidemiologia , Malásia/epidemiologia , Neuraminidase/genética , Filogenia , Aves Domésticas
19.
ACS Infect Dis ; 8(8): 1543-1552, 2022 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-35819162

RESUMO

Understanding mechanisms of resistance to antiviral inhibitors can reveal nuanced features of targeted viral mechanisms and, in turn, lead to improved strategies for inhibitor design. Arbidol is a broad-spectrum antiviral that binds to and prevents the fusion-associated conformational changes in the trimeric influenza A virus (IAV) hemagglutinin (HA). The rate-limiting step during the HA-mediated membrane fusion is the release of the hydrophobic fusion peptides from a conserved pocket on HA. Here, we investigated how destabilizing or stabilizing mutations in or near the fusion peptide affect viral sensitivity to Arbidol. The degree of sensitivity was proportional to the extent of fusion-peptide stability on the prefusion HA: stabilized mutants were more sensitive, and destabilized ones were resistant to Arbidol. Single-virion membrane fusion experiments for representative wild-type (WT) and mutant viruses demonstrated that resistance is a direct consequence of fusion-peptide destabilization not requiring reduced Arbidol binding to HA. Our results support the model whereby the probability of individual HAs extending to engage the target membrane is determined by the composite of two critical forces: a "tug" on the fusion peptide by HA rearrangements near the Arbidol binding site and the key interactions stabilizing the fusion peptide in the prefusion pocket. Arbidol increases and destabilizing mutations decrease the free-energy cost for fusion-peptide release, accounting for the observed resistance. Our findings have broad implications for fusion inhibitor design, viral mechanisms of resistance, and our basic understanding of HA-mediated membrane fusion.


Assuntos
Vírus da Influenza A , Antivirais/química , Antivirais/farmacologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Hemaglutininas/genética , Indóis , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Sulfetos
20.
PLoS One ; 17(6): e0268957, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35671300

RESUMO

Avian influenza virus (AIV) represents a major concern with productive implications in poultry systems but it is also a zoonotic agent that possesses an intrinsic pandemic risk. AIV is an enveloped, negative-sense and single-stranded RNA virus with a segmented genome. The eight genomic segments, comprising the whole genome, encode for eleven proteins. Within these proteins, Hemagglutinin (HA) and Neuraminidase (NA) are the most relevant for studies of evolution and pathogenesis considering their role in viral replication, and have also been used for classification purposes. Migratory birds are the main hosts and play a pivotal role in viral evolution and dissemination due to their migratory routes that comprise large regions worldwide. Altogether, viral and reservoir factors contribute to the emergence of avian influenza viruses with novel features and pathogenic potentials. The study aimed to conduct surveillance of AIVs in wild birds from Peru. A multi-site screening of feces of migratory birds was performed to isolate viruses and to characterize the whole genome sequences, especially the genes coding for HA and NA proteins. Four-hundred-twenty-one (421) fecal samples, collected between March 2019 and March 2020 in Lima, were obtained from 21 species of wild birds. From these, we isolated five AIV from whimbrel, kelp gull, Franklin's gulls and Mallard, which were of low pathogenicity, including four subtypes as H6N8, H13N6, H6N2 and H2N6. Genetic analysis of HA and NA genes revealed novel features in these viruses and phylogenetic analysis exhibited a close relationship with those identified in North America (US and Canada). Furthermore, H2N6 isolate presented a NA sequence with higher genetic relationship to Chilean isolates. These results highlight that the geographical factor is of major relevance in the evolution of AIV, suggesting that AIV circulating in Peru might represent a new site for the emergence of reassortant AIVs.


Assuntos
Charadriiformes , Vírus da Influenza A , Influenza Aviária , Animais , Animais Selvagens , Aves , Hemaglutininas/genética , Neuraminidase/genética , Peru/epidemiologia , Filogenia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...