Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Microbiol Resour Announc ; 13(8): e0021824, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39046228

RESUMEN

In Bangladesh, free-range duck farms provide opportunities for the generation of novel influenza A viruses as evidenced by the emergence of an unusual A(H1N7) virus in 2023. Continued surveillance of such environments for the potential emergence of influenza A viruses with novel properties remains a priority.

2.
J Virol ; 98(7): e0088124, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38958444

RESUMEN

In March 2024, clade 2.3.4.4b H5N1 highly pathogenic avian influenza virus (HPAIV) was detected in dairy cattle in the US, and it was discovered that the virus could be detected in raw milk. Although affected cow's milk is diverted from human consumption and current pasteurization requirements are expected to reduce or eliminate infectious HPAIV from the milk supply, a study was conducted to characterize whether the virus could be detected by quantitative real-time RT-PCR (qrRT-PCR) in pasteurized retail dairy products and, if detected, to determine whether the virus was viable. From 18 April to 22 April 2024, a total of 297 samples of Grade A pasteurized retail milk products (23 product types) were collected from 17 US states that represented products from 132 processors in 38 states. Viral RNA was detected in 60 samples (20.2%), with qrRT-PCR-based quantity estimates (non-infectious) of up to 5.4log1050% egg infectious doses per mL, with a mean and median of 3.0log10/mL and 2.9log10/mL, respectively. Samples that were positive for type A influenza by qrRT-PCR were confirmed to be clade 2.3.4.4 H5 HPAIV by qrRT-PCR. No infectious virus was detected in any of the qrRT-PCR-positive samples in embryonating chicken eggs. Further studies are needed to monitor the milk supply, but these results provide evidence that the infectious virus did not enter the US pasteurized milk supply before control measures for HPAIV were implemented in dairy cattle.IMPORTANCEHighly pathogenic avian influenza virus (HPAIV) infections in US dairy cattle were first confirmed in March 2024. Because the virus could be detected in raw milk, a study was conducted to determine whether it had entered the retail food supply. Pasteurized dairy products were collected from 17 states in April 2024. Viral RNA was detected in one in five samples, but infectious virus was not detected. This provides a snapshot of HPAIV in milk products early in the event and reinforces that with current safety measures, infectious viruses in milk are unlikely to enter the food supply.


Asunto(s)
Productos Lácteos , Leche , ARN Viral , Animales , Bovinos , Leche/virología , Estados Unidos , Productos Lácteos/virología , ARN Viral/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Pasteurización , Gripe Aviar/virología , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa
3.
Microbiol Resour Announc ; 13(8): e0026024, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39037308

RESUMEN

The influenza virus strain A/mute swan/Mangystau/1-S24R-2/2024 (H5N1; clade 2.3.4.4b) was isolated in embryonated chicken eggs from the lung of a dead swan found around Lake Karakol (Kazakhstan) during a highly pathogenic avian influenza outbreak in 2024. The aim of this study was to characterize the genetic profile of the isolated strain.

4.
Microbiol Resour Announc ; 13(7): e0015824, 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-38860813

RESUMEN

The whole genome sequence of a low pathogenicity avian influenza virus (H6N2) was sequenced from a Brazilian teal (Amazonetta brasiliensis) in Brazil, 2023. Phylogenetic analysis of the whole genome revealed a distinct genome pertaining to South American LPAIV from 2014 to 2016, indicating extensive circulation among South American wild birds.

5.
Vet Res ; 55(1): 54, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38671518

RESUMEN

This article reviews the avian viruses that infect the skin of domestic farm birds of primary economic importance: chicken, duck, turkey, and goose. Many avian viruses (e.g., poxviruses, herpesviruses, Influenza viruses, retroviruses) leading to pathologies infect the skin and the appendages of these birds. Some of these viruses (e.g., Marek's disease virus, avian influenza viruses) have had and/or still have a devasting impact on the poultry economy. The skin tropism of these viruses is key to the pathology and virus life cycle, in particular for virus entry, shedding, and/or transmission. In addition, for some emergent arboviruses, such as flaviviruses, the skin is often the entry gate of the virus after mosquito bites, whether or not the host develops symptoms (e.g., West Nile virus). Various avian skin models, from primary cells to three-dimensional models, are currently available to better understand virus-skin interactions (such as replication, pathogenesis, cell response, and co-infection). These models may be key to finding solutions to prevent or halt viral infection in poultry.


Asunto(s)
Enfermedades de las Aves de Corral , Virosis , Animales , Aves de Corral/virología , Enfermedades de las Aves de Corral/virología , Piel/virología , Virosis/veterinaria , Virosis/virología
6.
Microbiol Spectr ; 11(6): e0305523, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37982626

RESUMEN

IMPORTANCE: With the circulation of high pathogenicity avian influenza viruses having intensified considerably in recent years, the European Union is considering the vaccination of farmed birds. A prerequisite for this vaccination is the implementation of drastic surveillance protocols. Environmental sampling is a relevant alternative to animal sampling. However, environmental samples often contain inhibitory compounds in large enough quantities to inhibit RT-qPCR reactions. As bovine serum albumin is a molecule used in many fields to overcome this inhibitory effect, we tested its use on dust samples from poultry farms in areas heavily affected by HPAIV epizootics. Our results show that its use significantly increases the sensitivity of the method.


Asunto(s)
Virus de la Influenza A , Gripe Aviar , Enfermedades de las Aves de Corral , Animales , Albúmina Sérica Bovina , Polvo , Virulencia , Virus de la Influenza A/genética , Aves de Corral , Filogenia
7.
J Virol ; 97(10): e0078023, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37702486

RESUMEN

IMPORTANCE: AAVs are extensively studied as promising therapeutic gene delivery vectors. In order to circumvent pre-existing antibodies targeting primate-based AAV capsids, the AAAV capsid was evaluated as an alternative to primate-based therapeutic vectors. Despite the high sequence diversity, the AAAV capsid was found to bind to a common glycan receptor, terminal galactose, which is also utilized by other AAVs already being utilized in gene therapy trials. However, contrary to the initial hypothesis, AAAV was recognized by approximately 30% of human sera tested. Structural and sequence comparisons point to conserved epitopes in the fivefold region of the capsid as the reason determinant for the observed cross-reactivity.


Asunto(s)
Antígenos Virales , Cápside , Parvovirinae , Animales , Humanos , Cápside/química , Proteínas de la Cápside/química , Dependovirus/química , Vectores Genéticos , Primates/genética , Antígenos Virales/química , Parvovirinae/química
8.
Microbiol Spectr ; 11(4): e0435122, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37318353

RESUMEN

Ducks have recently received a lot of attention from the research community due to their importance as natural reservoirs of avian influenza virus (AIV). Still, there is a lack of tools to efficiently determine the immune status of ducks. The purpose of this work was to develop an automated differential blood count for the mallard duck (Anas platyrhynchos), to assess reference values of white blood cell (WBC) counts in this species, and to apply the protocol in an AIV field study. We established a flow cytometry-based duck WBC differential based on a no-lyse no-wash single-step one-tube technique, applying a combination of newly generated monoclonal antibodies with available duck-specific as well as cross-reacting chicken markers. The blood cell count enables quantification of mallard thrombocytes, granulocytes, monocytes, B cells, CD4+ T cells (T helper) and CD8+ cytotoxic T cells. The technique is reproducible, accurate, and much faster than traditional evaluations of blood smears. Stabilization of blood samples enables analysis up to 1 week after sampling, thus allowing for evaluation of blood samples collected in the field. We used the new technique to investigate a possible influence of sex, age, and AIV infection status on WBC counts in wild mallards. We show that age has an effect on the WBC counts in mallards, as does sex in juvenile mallards. Interestingly, males naturally infected with low pathogenic AIV showed a reduction of lymphocytes (lymphocytopenia) and thrombocytes (thrombocytopenia), which are both common in influenza A infection in humans. IMPORTANCE Outbreaks of avian influenza in poultry and humans are a global public health concern. Aquatic birds are the primary natural reservoir of avian influenza viruses (AIVs), and strikingly, AIVs mainly cause asymptomatic or mild infection in these species. Hence, immunological studies in aquatic birds are important for investigating variation in disease outcome of different hosts to AIV and may aid in early recognition and a better understanding of zoonotic events. Unfortunately, immunological studies in these species were so far hampered by the lack of diagnostic tools. Here, we present a technique that enables high-throughput white blood cell (WBC) analysis in the mallard and report changes in WBC counts in wild mallards naturally infected with AIV. Our protocol permits large-scale immune status monitoring in a widespread wild and domesticated duck species and provides a tool to further investigate the immune response in an important reservoir host of zoonotic viruses.


Asunto(s)
Virus de la Influenza A , Gripe Aviar , Animales , Humanos , Patos , Citometría de Flujo , Virus de la Influenza A/fisiología , Aves
9.
J Appl Microbiol ; 134(1)2023 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-36626750

RESUMEN

Development of molecular biology and understanding structures and functions of various biological molecules and entities allowed to construct various sophisticated tools for different biotechnological, medical, and veterinary applications. One of them is the phage display technology, based on the possibility to create specific bacteriophages bearing fusion genes, which code for fusion proteins consisting of a phage coat protein and a peptide of any amino acid sequence. Such proteins retain their biological functions as structural elements of phage virions while exposing foreign peptide sequences on their surfaces. Genetic manipulations allow to construct phage display libraries composed of billions of variants of exposed peptides; such libraries can be used to select peptides of desired features. Although the phage display technology has been widely used in biotechnology and medicine, its applications in veterinary and especially in poultry science were significantly less frequent. Nevertheless, many interesting discoveries have been reported also in the latter field, providing evidence for a possibility of effective applications of phage display-related methods in developing novel diagnostic tools, new vaccines, and innovative potential therapies dedicated to poultry. Especially, infectious diseases caused by avian viruses, bacteria, and unicellular eukaryotic parasites were investigated in this field. These studies are summarized and discussed in this review, with presentation of various possibilities provided by different phage display systems in development of useful and effective products facilitating management of the problem of infectious diseases of poultry.


Asunto(s)
Bacteriófagos , Enfermedades Transmisibles , Animales , Humanos , Biblioteca de Péptidos , Aves de Corral , Péptidos/química , Bacteriófagos/genética , Bacteriófagos/metabolismo
10.
mBio ; 14(1): e0339922, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36645303

RESUMEN

Host restriction limits the emergence of novel pandemic strains from the influenza A virus avian reservoir. For efficient replication in mammalian cells, the avian influenza RNA-dependent RNA polymerase must adapt to use human orthologues of the host factor ANP32, which lack a 33-amino-acid insertion relative to avian ANP32A. Here, we find that influenza polymerase requires ANP32 proteins to support both steps of genome replication: cRNA and vRNA synthesis. However, avian strains are only restricted in vRNA synthesis in human cells. Therefore, avian influenza polymerase can use human ANP32 orthologues to support cRNA synthesis, without acquiring mammalian adaptations. This implies a fundamental difference in the mechanism by which ANP32 proteins support cRNA versus vRNA synthesis. IMPORTANCE To infect humans and cause a pandemic, avian influenza must first adapt to use human versions of the proteins the virus hijacks for replication, instead of the avian orthologues found in bird cells. One critical host protein is ANP32. Understanding the details of how host proteins such as ANP32 support viral activity may allow the design of new antiviral strategies that disrupt these interactions. Here, we use cells that lack ANP32 to unambiguously demonstrate ANP32 is needed for both steps of influenza genome replication. Unexpectedly, however, we found that avian influenza can use human ANP32 proteins for the first step of replication, to copy a complementary strand, without adaptation but can only utilize avian ANP32 for the second step of replication that generates new genomes. This suggests ANP32 may have a distinct role in supporting the second step of replication, and it is this activity that is specifically blocked when avian influenza infects human cells.


Asunto(s)
Virus de la Influenza A , Gripe Aviar , Gripe Humana , Animales , Humanos , ARN Complementario/metabolismo , Línea Celular , Virus de la Influenza A/genética , Replicación Viral , ARN Viral/metabolismo , Mamíferos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
11.
Curr Res Microb Sci ; 3: 100169, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36518169

RESUMEN

Caliciviruses are ssRNA viruses that can infect a wide range of hosts, including birds. While several avian caliciviruses have been discovered, their taxonomy and host distribution are largely unknown. We molecularly characterized a novel calicivirus (trumpeter swan calicivirus: TruSCV) in trumpeter swans over-wintering in south-west British Columbia, Canada. The positivity rate was 20.3% (14/69) and there were no significant differences in infection rates between males (5/34, 14.7%) and females (9/35, 25.7%) or among considered age groups (juveniles: 4/14, 28.6%; sub-adults: 1/9, 11.1%; adults: 9/46, 19.6%). Twelve infected swans died of lead poisoning, one because of starvation, and one from physical injuries. TruSCV complete genome possessed the typical organization and protein motifs of caliciviruses and a type 2 IRES and its closest relative was a virus circulating in Australian ducks. Phylogenetic analyses showed the existence of 34 different but monophyletic avian caliciviruses. These viruses, while having conserved genomic organization and protein motifs, possess different IRES types and group in several divergent clades, with only two of them corresponding to currently defined genera, highlighting the need for epidemiological investigations and systematic analyses to better define their taxonomy. Follow-up studies are needed to elucidate the diversity, distribution, and pathogenic potential of TruSCV.

12.
J Virol ; 96(18): e0077622, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36069546

RESUMEN

The nonstructural protein 1 (NS1) of influenza A viruses is an important virulence factor that controls host cell immune responses. In human cells, NS1 proteins inhibit the induction of type I interferon by several mechanisms, including potentially, by preventing the activation of the retinoic acid-inducible gene I (RIG-I) receptor by the ubiquitin ligase tripartite motif-containing protein 25 (TRIM25). It is unclear whether the inhibition of human TRIM25 is a universal function of all influenza A NS1 proteins or is strain dependent. It is also unclear if NS1 proteins similarly target the TRIM25 of mallard ducks, a natural reservoir host of avian influenza viruses with a long coevolutionary history and unique disease dynamics. To answer these questions, we compared the ability of five different NS1 proteins to interact with human and duck TRIM25 using coimmunoprecipitation and microscopy and assessed the consequence of this on RIG-I ubiquitination and signaling in both species. We show that NS1 proteins from low-pathogenic and highly pathogenic avian influenza viruses potently inhibit RIG-I ubiquitination and reduce interferon promoter activity and interferon-beta protein secretion in transfected human cells, while the NS1 of the mouse-adapted PR8 strain does not. However, all the NS1 proteins, when cloned into recombinant viruses, suppress interferon in infected alveolar cells. In contrast, avian NS1 proteins do not suppress duck RIG-I ubiquitination and interferon promoter activity, despite interacting with duck TRIM25. IMPORTANCE Influenza A viruses are a major cause of human and animal disease. Periodically, avian influenza viruses from wild waterfowl, such as ducks, pass through intermediate agricultural hosts and emerge into the human population as zoonotic diseases with high mortality rates and epidemic potential. Because of their coevolution with influenza A viruses, ducks are uniquely resistant to influenza disease compared to other birds, animals, and humans. Here, we investigate a mechanism of influenza A virus interference in an important antiviral signaling pathway that is orthologous in humans and ducks. We show that NS1 proteins from four avian influenza strains can block the coactivation and signaling of the human RIG-I antiviral receptor, while none block the coactivation and signaling of duck RIG-I. Understanding host-pathogen dynamics in the natural reservoir will contribute to our understanding of viral disease mechanisms, viral evolution, and the pressures that drive it, which benefits global surveillance and outbreak prevention.


Asunto(s)
Proteínas Aviares , Virus de la Influenza A , Gripe Aviar , Interferón beta , Receptores de Ácido Retinoico , Transducción de Señal , Proteínas no Estructurales Virales , Animales , Antivirales/metabolismo , Proteínas Aviares/metabolismo , Patos , Humanos , Virus de la Influenza A/genética , Interferón Tipo I/metabolismo , Interferón beta/metabolismo , Ratones , Receptores de Ácido Retinoico/metabolismo , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitinación , Proteínas no Estructurales Virales/metabolismo
13.
Front Immunol ; 13: 901913, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35634318

RESUMEN

Innate immunity is not only the first line of host defense against pathogenic infection, but also the cornerstone of adaptive immune response. Upon pathogenic infection, pattern recognition receptors (PRRs) of host engage pathogen-associated molecular patterns (PAMPs) of pathogens, which initiates IFN production by activating interferon regulatory transcription factors (IRFs), nuclear factor-kappa B (NF-κB), and/or activating protein-1 (AP-1) signal transduction pathways in host cells. In order to replicate and survive, pathogens have evolved multiple strategies to evade host innate immune responses, including IFN-I signal transduction, autophagy, apoptosis, necrosis, inflammasome and/or metabolic pathways. Some avian viruses may not be highly pathogenic but they have evolved varied strategies to evade or suppress host immune response for survival, causing huge impacts on the poultry industry worldwide. In this review, we focus on the advances on innate immune evasion by several important avian immunosuppressive viruses (infectious bursal disease virus (IBDV), Marek's disease virus (MDV), avian leukosis virus (ALV), etc.), especially their evasion of PRRs-mediated signal transduction pathways (IFN-I signal transduction pathway) and IFNAR-JAK-STAT signal pathways. A comprehensive understanding of the mechanism by which avian viruses evade or suppress host immune responses will be of help to the development of novel vaccines and therapeutic reagents for the prevention and control of infectious diseases in chickens.


Asunto(s)
Evasión Inmune , Virus , Animales , Pollos/metabolismo , Inmunidad Innata , Inmunosupresores , Factores Reguladores del Interferón/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos , Receptores de Reconocimiento de Patrones/metabolismo , Virus/metabolismo
14.
J Virol ; 96(6): e0205921, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35044208

RESUMEN

The Gammacoronavirus infectious bronchitis virus (IBV) is a highly contagious global pathogen prevalent in all types of poultry flocks. IBV is responsible for economic losses and welfare issues in domestic poultry, resulting in a significant risk to food security. IBV vaccines are currently generated by serial passage of virulent IBV field isolates through embryonated hens' eggs. The different patterns of genomic variation accumulated during this process means that the exact mechanism of attenuation is unknown and presents a risk of reversion to virulence. Additionally, the passaging process adapts the virus to replicate in chicken embryos, increasing embryo lethality. Vaccines produced in this manner are therefore unsuitable for in ovo application. We have developed a reverse genetics system, based on the pathogenic IBV strain M41, to identify genes which can be targeted for rational attenuation. During the development of this reverse genetics system, we identified four amino acids, located in nonstructural proteins (nsps) 10, 14, 15, and 16, which resulted in attenuation both in vivo and in ovo. Further investigation highlighted a role of amino acid changes, Pro85Leu in nsp 10 and Val393Leu in nsp 14, in the attenuated in vivo phenotype observed. This study provides evidence that mutations in nsps offer a promising mechanism for the development of rationally attenuated live vaccines against IBV, which have the potential for in ovo application. IMPORTANCE The Gammacoronavirus infectious bronchitis virus (IBV) is the etiological agent of infectious bronchitis, an acute, highly contagious, economically important disease of poultry. Vaccination is achieved using a mixture of live attenuated vaccines for young chicks and inactivated vaccines as boosters for laying hens. Live attenuated vaccines are generated through serial passage in embryonated hens' eggs, an empirical process which achieves attenuation but retains immunogenicity. However, these vaccines have a risk of reversion to virulence, and they are lethal to the embryo. In this study, we identified amino acids in the replicase gene which attenuated IBV strain M41, both in vivo and in ovo. Stability assays indicate that the attenuating amino acids are stable and unlikely to revert. The data in this study provide evidence that specific modifications in the replicase gene offer a promising direction for IBV live attenuated vaccine development, with the potential for in ovo application.


Asunto(s)
Aminoácidos , Infecciones por Coronavirus , Virus de la Bronquitis Infecciosa , Enfermedades de las Aves de Corral , Proteínas no Estructurales Virales , Vacunas Virales , Aminoácidos/química , Aminoácidos/genética , Animales , Embrión de Pollo , Pollos , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Femenino , Virus de la Bronquitis Infecciosa/genética , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología , Vacunas Atenuadas/genética , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Vacunas Virales/genética
15.
J Virol ; 96(1): e0136621, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-34613804

RESUMEN

Highly pathogenic avian influenza viruses (HPAIV) emerge from low-pathogenic avian influenza viruses (LPAIV) through the introduction of basic amino acids at the hemagglutinin (HA) cleavage site. Following viral evolution, the newly formed HPAIV likely represents a minority variant within the index host, predominantly infected with the LPAIV precursor. Using reverse genetics-engineered H5N8 viruses differing solely at the HA cleavage, we tested the hypothesis that the interaction between the minority HPAIV and the majority LPAIV could modulate the risk of HPAIV emergence and that the nature of the interaction could depend on the host species. In chickens, we observed that the H5N8LP increased H5N8HP replication and pathogenesis. In contrast, the H5N8LP antagonized H5N8HP replication and pathogenesis in ducks. Ducks mounted a more potent antiviral innate immune response than chickens against the H5N8LP, which correlated with H5N8HP inhibition. These data provide experimental evidence that HPAIV may be more likely to emerge in chickens than in ducks and underscore the importance of within-host viral variant interactions in viral evolution. IMPORTANCE Highly pathogenic avian influenza viruses represent a threat to poultry production systems and to human health because of their impact on food security and because of their zoonotic potential. It is therefore crucial to better understand how these viruses emerge. Using a within-host competition model between high- and low-pathogenic avian influenza viruses, we provide evidence that highly pathogenic avian influenza viruses could be more likely to emerge in chickens than in ducks. These results have important implications for highly pathogenic avian influenza virus emergence prevention, and they underscore the importance of within-host viral variant interactions in virus evolution.


Asunto(s)
Pollos , Susceptibilidad a Enfermedades , Patos , Interacciones Huésped-Patógeno , Subtipo H5N8 del Virus de la Influenza A/fisiología , Gripe Aviar/virología , Enfermedades de las Aves de Corral/virología , Animales , Biomarcadores , Biopsia , Células Cultivadas , Coinfección , Genotipo , Inmunohistoquímica , Gripe Aviar/metabolismo , Gripe Aviar/patología , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/patología , ARN Viral , Especificidad de la Especie , Carga Viral , Virulencia , Replicación Viral
16.
J Virol ; 96(5): e0040821, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33853954

RESUMEN

PA-X is a nonstructural protein of influenza A virus (IAV), which is encoded by the polymerase acidic (PA) N-terminal region that contains a C-terminal +1 frameshifted sequence. IAV PA-X protein modulates virus-induced host innate immune responses and viral pathogenicity via suppression of host gene expression or cellular shutoff, through cellular mRNA cleavage. Highly pathogenic avian influenza viruses (HPAIV) of the H5N1 subtype naturally infect different avian species, they have an enormous economic impact in the poultry farming, and they also have zoonotic and pandemic potential, representing a risk to human public health. In the present study, we describe a novel bacterium-based approach to identify amino acid residues in the PA-X protein of the HPAIV A/Viet Nam/1203/2004 H5N1 that are important for its ability to inhibit host protein expression or cellular shutoff activity. Identified PA-X mutants displayed a reduced shutoff activity compared to that of the wild-type A/Viet Nam/1203/2004 H5N1 PA-X protein. Notably, this new bacterium-based screening allowed us to identify amino acid residues widely distributed over the entire N-terminal region of PA-X. Furthermore, we found that some of the residues affecting A/Viet Nam/1203/2004 H5N1 PA-X host shutoff activity also affect PA polymerase activity in a minigenome assay. This information could be used for the rational design of new and more effective compounds with antiviral activity against IAV. Moreover, our results demonstrate the feasibility of using this bacterium-based approach to identify amino acid residues important for the activity of viral proteins to inhibit host gene expression. IMPORTANCE Highly pathogenic avian influenza viruses continue to pose a huge threat to global animal and human health. Despite of the limited genome size of Influenza A virus (IAV), the virus encodes eight main viral structural proteins and multiple accessory nonstructural proteins, depending on the IAV type, subtype, or strain. One of the IAV accessory proteins, PA-X, is encoded by the polymerase acidic (PA) protein and is involved in pathogenicity through the modulation of IAV-induced host inflammatory and innate immune responses. However, the molecular mechanism(s) of IAV PA-X regulation of the host immune response is not well understood. Here, we used, for the first time, a bacterium-based approach for the identification of amino acids important for the ability of IAV PA-X to induce host shutoff activity and describe novel residues relevant for its ability to inhibit host gene expression, and their contribution in PA polymerase activity.


Asunto(s)
Aminoácidos , Expresión Génica , Interacciones Huésped-Patógeno , Subtipo H5N1 del Virus de la Influenza A , Proteínas Represoras , Proteínas no Estructurales Virales , Aminoácidos/genética , Aminoácidos/inmunología , Animales , Bacterias/virología , Aves/inmunología , Expresión Génica/genética , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/inmunología , Gripe Aviar/inmunología , Gripe Aviar/virología , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Vietnam , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología
17.
mBio ; 12(5): e0178521, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34488450

RESUMEN

Avian influenza viruses pose a continuous threat to both poultry and human health, with significant economic impact. The ability of viruses to reassort and jump the species barrier into mammalian hosts generates a constant pandemic threat. H10Nx avian viruses have been shown to replicate in mammalian species without prior adaptation and have caused significant human infection and fatalities. They are able to rapidly reassort with circulating poultry strains and go undetected due to their low pathogenicity in chickens. Novel detections of both human reassortant strains and increasing endemicity of H10Nx poultry infections highlight the increasing need for heightened surveillance and greater understanding of the distribution, tropism, and infection capabilities of these viruses. In this minireview, we highlight the gap in the current understanding of this subtype and its prevalence across a vast range of host species and geographical locations.


Asunto(s)
Gripe Aviar/transmisión , Aves de Corral/virología , Zoonosis Virales/transmisión , Animales , Australia , Aves , Pollos , Especificidad del Huésped , Humanos , Gripe Aviar/clasificación , Gripe Aviar/genética , Gripe Humana/virología , Infecciones por Orthomyxoviridae/virología , Pandemias , Porcinos
18.
Viruses ; 13(5)2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-34068495

RESUMEN

Avian influenza virus A (H7N9), after circulating in avian hosts for decades, was identified as a human pathogen in 2013. Herein, amino acid substitutions possibly essential for human adaptation were identified by comparing the 4706 aligned overlapping nonamer position sequences (1-9, 2-10, etc.) of the reported 2014 and 2017 avian and human H7N9 datasets. The initial set of virus sequences (as of year 2014) exhibited a total of 109 avian-to-human (A2H) signature amino acid substitutions. Each represented the most prevalent substitution at a given avian virus nonamer position that was selectively adapted as the corresponding index (most prevalent sequence) of the human viruses. The majority of these avian substitutions were long-standing in the evolution of H7N9, and only 17 were first detected in 2013 as possibly essential for the initial human adaptation. Strikingly, continued evolution of the avian H7N9 virus has resulted in avian and human protein sequences that are almost identical. This rapid and continued adaptation of the avian H7N9 virus to the human host, with near identity of the avian and human viruses, is associated with increased human infection and a predicted greater risk of human-to-human transmission.


Asunto(s)
Adaptación Biológica , Interacciones Huésped-Patógeno , Subtipo H7N9 del Virus de la Influenza A/fisiología , Gripe Aviar/virología , Gripe Humana/virología , Infecciones por Orthomyxoviridae/virología , Sustitución de Aminoácidos , Animales , Aves , Variación Genética , Humanos , ARN Viral , Especificidad de la Especie
19.
Viruses ; 13(2)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525386

RESUMEN

Parvoviruses are small single-stranded DNA viruses that can infect both vertebrates and invertebrates. We report here the full characterization of novel viruses we identified in ducks, including two viral species within the subfamily Hamaparvovirinae (duck-associated chapparvovirus, DAC) and a novel species within the subfamily Densovirinae (duck-associated ambidensovirus, DAAD). Overall, 5.7% and 21.1% of the 123 screened ducks (American black ducks, mallards, northern pintail) were positive for DAC and DAAD, respectively, and both viruses were more frequently detected in autumn than in winter. Genome organization and predicted transcription profiles of DAC and DAAD were similar to viruses of the genera Chaphamaparvovirus and Protoambidensovirus, respectively. Their association to these genera was also demonstrated by subfamily-wide phylogenetic and distance analyses of non-structural protein NS1 sequences. While DACs were included in a highly supported clade of avian viruses, no definitive conclusions could be drawn about the host type of DAAD because it was phylogenetically close to viruses found in vertebrates and invertebrates and analyses of codon usage bias and nucleotide frequencies of viruses within the family Parvoviridae showed no clear host-based viral segregation. This study highlights the high parvoviral diversity in the avian reservoir with many avian-associated parvoviruses likely yet to be discovered.


Asunto(s)
Patos/virología , Infecciones por Parvoviridae/veterinaria , Parvoviridae/genética , Animales , Animales Salvajes/virología , Uso de Codones , ADN Viral/genética , Patos/clasificación , Genoma Viral/genética , Especificidad del Huésped , Parvoviridae/clasificación , Infecciones por Parvoviridae/epidemiología , Infecciones por Parvoviridae/virología , Filogenia , Estaciones del Año , Proteínas no Estructurales Virales/genética
20.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-972799

RESUMEN

Aims@#Psittacine birds such as parrots, macaws, cockatoos, lovebirds and parakeets, are widely reared as household pets or at aviary due to their attractive features. However, the status of virus-causing diseases of psittacine species in Malaysia is fairly under-documented. Therefore, this study was aimed to detect the presence of three common avian viruses that infect psittacine birds, i.e. beak and feather disease virus (BFDV), avian polyomavirus and avian papillomavirus. @*Methodology and results@#Faecal samples from twelve asymptomatic captive psittacine birds of different species were collected from an undisclosed animal garden in Serdang, Selangor, Malaysia. Briefly, the sample was homogenised and resuspended with SM buffer with the ratio 1:1 (weight of sample/g: volume of SM buffer/mL) before centrifugation at 1,000 × g for 20 min. The supernatant was collected and filtered before subjected to genomic DNA extraction using a commercialised kit. Polymerase chain reaction (PCR) technique was used to screen the V1, VP1 and L1 genes of beak and feather disease virus (BFDV), avian polyomavirus and avian papillomavirus, respectively. Findings revealed that the samples were negative for BFDV and avian polyomavirus. However, positive results of 1.5 kbp PCR amplicon were detected for avian papillomavirus in four out of the 12 samples (33.33%), which was from the white-crested cockatoo, African grey parrot, yellow-collared macaw and Senegal parrot. Sequence analysis of the L1 gene from the Senegal parrot Poicephalus senegalus revealed 93% identity to a reference Psittacus erithacus timneh avian papillomavirus.@*Conclusion, significance and impact of study@#This study added to the limited prevalence data of three important avian viruses which infect captive psittacines in Seri Kembangan, Selangor, Malaysia. Avian papillomavirus, but not BFDV and avian polyomavirus, was detected in the collected captive psittacine birds. Therefore, a routine screening can be performed to monitor the health status of birds despite their asymptomatic manifestation, in order to prevent possible virus transmission.


Asunto(s)
Virosis , Aves
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA