Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Bioinformatics ; 36(7): 2098-2104, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31790143

RESUMEN

MOTIVATION: The potentially low precision associated with the geographic origin of sampled sequences represents an important limitation for spatially explicit (i.e. continuous) phylogeographic inference of fast-evolving pathogens such as RNA viruses. A substantial proportion of publicly available sequences is geo-referenced at broad spatial scale such as the administrative unit of origin, rather than more precise locations (e.g. geographic coordinates). Most frequently, such sequences are either discarded prior to continuous phylogeographic inference or arbitrarily assigned to the geographic coordinates of the centroid of their administrative area of origin for lack of a better alternative. RESULTS: We here implement and describe a new approach that allows to incorporate heterogeneous prior sampling probabilities over a geographic area. External data, such as outbreak locations, are used to specify these prior sampling probabilities over a collection of sub-polygons. We apply this new method to the analysis of highly pathogenic avian influenza H5N1 clade data in the Mekong region. Our method allows to properly include, in continuous phylogeographic analyses, H5N1 sequences that are only associated with large administrative areas of origin and assign them with more accurate locations. Finally, we use continuous phylogeographic reconstructions to analyse the dispersal dynamics of different H5N1 clades and investigate the impact of environmental factors on lineage dispersal velocities. AVAILABILITY AND IMPLEMENTATION: Our new method allowing heterogeneous sampling priors for continuous phylogeographic inference is implemented in the open-source multi-platform software package BEAST 1.10. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Animales , Brotes de Enfermedades , Filogenia , Filogeografía , Probabilidad
2.
Nucleic Acids Res ; 45(D1): D466-D474, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-27679478

RESUMEN

The Influenza Research Database (IRD) is a U.S. National Institute of Allergy and Infectious Diseases (NIAID)-sponsored Bioinformatics Resource Center dedicated to providing bioinformatics support for influenza virus research. IRD facilitates the research and development of vaccines, diagnostics and therapeutics against influenza virus by providing a comprehensive collection of influenza-related data integrated from various sources, a growing suite of analysis and visualization tools for data mining and hypothesis generation, personal workbench spaces for data storage and sharing, and active user community support. Here, we describe the recent improvements in IRD including the use of cloud and high performance computing resources, analysis and visualization of user-provided sequence data with associated metadata, predictions of novel variant proteins, annotations of phenotype-associated sequence markers and their predicted phenotypic effects, hemagglutinin (HA) clade classifications, an automated tool for HA subtype numbering conversion, linkouts to disease event data and the addition of host factor and antiviral drug components. All data and tools are freely available without restriction from the IRD website at https://www.fludb.org.


Asunto(s)
Biología Computacional/métodos , Bases de Datos Factuales , Virus de la Influenza A , Investigación , Programas Informáticos , Virus de la Influenza A/clasificación , Virus de la Influenza A/fisiología , Tipificación Molecular/métodos , Fenotipo , Filogenia , Proteínas Virales/genética , Virulencia
3.
Avian Pathol ; 47(6): 559-575, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29985640

RESUMEN

In May 2016, highly pathogenic avian influenza virus of the subtype A/H5N1 was detected in Cameroon in an industrial poultry farm at Mvog-Betsi, Yaoundé (Centre region), with a recorded sudden increase of deaths among chickens, and an overall mortality rate of 75%. The virus spread further and caused new outbreaks in some parts of the country. In total, 21 outbreaks were confirmed from May 2016 to March 2017 (six in the Centre, six in the West, eight in the South and one in the Adamaoua regions). This resulted in an estimated total loss of 138,252 birds (44,451 deaths due to infection and 93,801 stamped out). Only domestic birds (chickens, ducks and geese) were affected in farms as well as in poultry markets. The outbreaks occurred in three waves, the first from May to June 2016, the second in September 2016 and the last wave in March 2017. The topology of the phylogeny based on the haemagglutinin gene segment indicated that the causative H5N1 viruses fall within the genetic clade 2.3.2.1c, within the same group as the A/H5N1 viruses collected in Niger in 2015 and 2016. More importantly, the gene constellation of four representative viruses showed evidence of H5N1/H9N2 intra-clade reassortment. Additional epidemiological and genetic data from affected countries in West Africa are needed to better trace the origin, spread and evolution of A/H5N1 in Cameroon. RESEARCH HIGHLIGHTS HPAI A/H5N1 was detected in May 2016 in domestic chickens in Yaoundé-Cameroon. Twenty-one outbreaks in total were confirmed from May 2016 to March 2017. The causative H5N1 viruses fall within the genetic clade 2.3.2.1c. The viral gene constellation showed evidence of H5N1/H9N2 intra-clade reassortment.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/genética , Gripe Aviar/virología , Enfermedades de las Aves de Corral/virología , Virus Reordenados/genética , Animales , Camerún/epidemiología , Pollos/virología , Brotes de Enfermedades/veterinaria , Patos/virología , Gansos/virología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/epidemiología , Filogenia , Aves de Corral , Enfermedades de las Aves de Corral/epidemiología , Virus Reordenados/patogenicidad
4.
Euro Surveill ; 22(11)2017 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-28333616

RESUMEN

A cross-sectional study was conducted in Egypt to determine the prevalence of Middle East respiratory syndrome coronavirus (MERS-CoV) in imported and resident camels and bats, as well as to assess possible transmission of the virus to domestic ruminants and equines. A total of 1,031 sera, 1,078 nasal swabs, 13 rectal swabs, and 38 milk samples were collected from 1,078 camels in different types of sites. In addition, 145 domestic animals and 109 bats were sampled. Overall, of 1,031 serologically-tested camels, 871 (84.5%) had MERS-CoV neutralising antibodies. Seroprevalence was significantly higher in imported (614/692; 88.7%) than resident camels (257/339; 5.8%) (p < 0.05). Camels from Sudan (543/594; 91.4%) had a higher seroprevalence than those from East Africa (71/98; 72.4%) (p < 0.05). Sampling site and age were also associated with MERS-CoV seroprevalence (p < 0.05). All tested samples from domestic animals and bats were negative for MERS-CoV antibodies except one sheep sample which showed a 1:640 titre. Of 1,078 camels, 41 (3.8%) were positive for MERS-CoV genetic material. Sequences obtained were not found to cluster with clade A or B MERS-CoV sequences and were genetically diverse. The presence of neutralising antibodies in one sheep apparently in contact with seropositive camels calls for further studies on domestic animals in contact with camels.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Camelus/virología , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Coronavirus/aislamiento & purificación , Brotes de Enfermedades/veterinaria , Reservorios de Enfermedades/virología , Zoonosis/diagnóstico , Animales , Secuencia de Bases , Bovinos/sangre , Coronavirus/genética , Infecciones por Coronavirus/diagnóstico , Estudios Transversales , Egipto/epidemiología , Genotipo , Cabras/sangre , Inmunoglobulina G/sangre , Coronavirus del Síndrome Respiratorio de Oriente Medio/clasificación , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Datos de Secuencia Molecular , Prevalencia , ARN Viral/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ARN , Estudios Seroepidemiológicos , Ovinos/sangre , Zoonosis/epidemiología , Zoonosis/virología
6.
J Virol ; 89(7): 3746-62, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25609805

RESUMEN

UNLABELLED: Vaccines are used in integrated control strategies to protect poultry against H5N1 high-pathogenicity avian influenza (HPAI). H5N1 HPAI was first reported in Indonesia in 2003, and vaccination was initiated in 2004, but reports of vaccine failures began to emerge in mid-2005. This study investigated the role of Indonesian licensed vaccines, specific vaccine seed strains, and emerging variant field viruses as causes of vaccine failures. Eleven of 14 licensed vaccines contained the manufacturer's listed vaccine seed strains, but 3 vaccines contained a seed strain different from that listed on the label. Vaccines containing A/turkey/Wisconsin/1968 (WI/68), A/chicken/Mexico/28159-232/1994 (Mex/94), and A/turkey/England/N28/1973 seed strains had high serological potency in chickens (geometric mean hemagglutination inhibition [HI] titers, ≥ 1:169), but vaccines containing strain A/chicken/Guangdong/1/1996 generated by reverse genetics (rg; rgGD/96), A/chicken/Legok/2003 (Legok/03), A/chicken/Vietnam/C57/2004 generated by rg (rgVN/04), or A/chicken/Legok/2003 generated by rg (rgLegok/03) had lower serological potency (geometric mean HI titers, ≤ 1:95). In challenge studies, chickens immunized with any of the H5 avian influenza vaccines were protected against A/chicken/West Java/SMI-HAMD/2006 (SMI-HAMD/06) and were partially protected against A/chicken/Papua/TA5/2006 (Papua/06) but were not protected against A/chicken/West Java/PWT-WIJ/2006 (PWT/06). Experimental inactivated vaccines made with PWT/06 HPAI virus or rg-generated PWT/06 low-pathogenicity avian influenza (LPAI) virus seed strains protected chickens from lethal challenge, as did a combination of a commercially available live fowl poxvirus vaccine expressing the H5 influenza virus gene and inactivated Legok/03 vaccine. These studies indicate that antigenic variants did emerge in Indonesia following widespread H5 avian influenza vaccine usage, and efficacious inactivated vaccines can be developed using antigenic variant wild-type viruses or rg-generated LPAI virus seed strains containing the hemagglutinin and neuraminidase genes of wild-type viruses. IMPORTANCE: H5N1 high-pathogenicity avian influenza (HPAI) virus has become endemic in Indonesian poultry, and such poultry are the source of virus for birds and mammals, including humans. Vaccination has become a part of the poultry control strategy, but vaccine failures have occurred in the field. This study identified possible causes of vaccine failure, which included the use of an unlicensed virus seed strain and induction of low levels of protective antibody because of an insufficient quantity of vaccine antigen. However, the most important cause of vaccine failure was the appearance of drift variant field viruses that partially or completely overcame commercial vaccine-induced immunity. Furthermore, experimental vaccines using inactivated wild-type virus or reverse genetics-generated vaccines containing the hemagglutinin and neuraminidase genes of wild-type drift variant field viruses were protective. These studies indicate the need for surveillance to identify drift variant viruses in the field and update licensed vaccines when such variants appear.


Asunto(s)
Anticuerpos Antivirales/sangre , Protección Cruzada , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Aviar/prevención & control , Animales , Variación Antigénica , Pollos , Flujo Genético , Indonesia , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Vacunas contra la Influenza/administración & dosificación , Gripe Aviar/inmunología , Análisis de Supervivencia , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología
7.
Virol J ; 13: 49, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-27000533

RESUMEN

BACKGROUND: Highly pathogenic avian influenza (HPAI) viruses of the H5N1 subtype are widely distributed within poultry populations in Egypt and have caused multiple human infections. Linking the epidemiological and sequence data is important to understand the transmission, persistence and evolution of the virus. This work describes the phylogenetic dynamics of H5N1 based on molecular characterization of the hemagglutinin (HA) gene of isolates collected from February 2006 to May 2014. METHODS: Full-length HA sequences of 368 H5N1 viruses were generated and were genetically analysed to study their genetic evolution. They were collected from different poultry species, production sectors, and geographic locations in Egypt. The Bayesian Markov Chain Monte Carlo (BMCMC) method was applied to estimate the evolutionary rates among different virus clusters; additionally, an analysis of selection pressures in the HA gene was performed using the Single Likelihood Ancestor Counting (SLAC) method. RESULTS: The phylogenetic analysis of the H5 gene from 2006-14 indicated the presence of one virus introduction of the classic clade (2.2.1) from which two main subgroups were originated, the variant subgroup which was further subdivided into 2 sub-divisions (2.2.1.1 and 2.2.1.1a) and the endemic subgroup (2.2.1.2). The clade 2.2.1.2 showed a high evolution rate over a period of 6 years (6.9 × 10(-3) sub/site/year) in comparison to the 2.2.1.1a variant cluster (7.2 × 10(-3) over a period of 4 years). Those two clusters are under positive selection as they possess 5 distinct positively selected sites in the HA gene. The mutations at 120, 154, and 162 HA antigenic sites and the other two mutations (129∆, I151T) that occurred from 2009-14 were found to be stable in the 2.2.1.2 clade. Additionally, 13 groups of H5N1 HPAI viruses were identified based on their amino acid sequences at the cleavage site and "EKRRKKR" became the dominant pattern beginning in 2013. CONCLUSIONS: Continuous evolution of H5N1 HPAI viruses in Egypt has been observed in all poultry farming and production systems in almost all regions of the country. The wide circulation of the 2.2.1.2 clade carrying triple mutations (120, 129∆, I151T) associated with increased binding affinity to human receptors is an alarming finding of public health importance.


Asunto(s)
Genotipo , Subtipo H5N1 del Virus de la Influenza A/clasificación , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Aviar/epidemiología , Gripe Aviar/virología , Filogenia , Secuencia de Aminoácidos , Animales , Teorema de Bayes , Sitios de Unión , Aves , Análisis por Conglomerados , Egipto/epidemiología , Evolución Molecular , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores Virales/química , Receptores Virales/metabolismo , Selección Genética
8.
Avian Pathol ; 44(5): 333-41, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26023824

RESUMEN

Vaccination is the main tool implemented in Egypt since 2007 to control H5N1 avian influenza. The present study aimed at comparing the effectiveness of three avian influenza vaccination regimes in commercial broiler chickens carrying high levels of maternally derived antibodies (MDAs). Day-old chicks were divided into four experimental groups. Group I received only the rHVT-H5 vaccine (recombinant turkey herpesvirus (HVT) which carries a H5 clade 2.2 insert) administered at D1. Group II received only the KV-H5 (an oil emulsion killed vaccine prepared from reassortant HPAI virus (A/duck/Anhui/1/06)) vaccine (inactivated reverse genetic H5N1 clade 2.3.4 virus) administered at D8. Group III received rHVT-H5 and KV-H5 as prime/boost. Group IV served as unvaccinated control. Weekly serological monitoring was conducted using the haemagglutination inhibition test. Two challenge experiments were conducted at D28 and D35 using HPAI H5N1 clade 2.2.1 virus. Birds were monitored daily 14 days post-challenge for morbidity and mortality, and oropharyngeal swabs were collected for virological monitoring. Initially, day-old chicks had high mean MDA titres (9 + 0.9 log2). The MDA half-life was >7 and <7 days, respectively, for unvaccinated and vaccinated birds. Group III showed the highest post-vaccination humoral immune response and seroconversion rate. The highest protection rate against morbidity (80-90%) and mortality (90-90%) was obtained in Group III after challenge at D28 and D35, respectively, as compared to Group I (70-70%) and (80-90%) and Group II (0-0%) and (30-30%). Groups I and III had lower number of shedder birds. The vaccination regime with prime/boost conferred the highest and earliest protection, and can hence be recommended for the broiler production sector in endemic and high HPAI H5N1 challenge areas.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Gripe Aviar/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Vacunación , Animales , Pollos , Egipto , Pruebas de Inhibición de Hemaglutinación , Vacunas contra la Influenza/inmunología , Gripe Aviar/epidemiología , Gripe Aviar/inmunología , Enfermedades de las Aves de Corral/epidemiología , Enfermedades de las Aves de Corral/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
9.
Avian Pathol ; 43(6): 514-23, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25245772

RESUMEN

The effectiveness of recombinant turkey herpesvirus avian influenza (A/swan/Hungary/4999/2006(H5N1)) clade 2.2 virus (rHVT-H5) vaccine was evaluated in two layer chicken breeds (White Bovans [WB] and Brown Shaver [BS]). One dose of rHVT-H5 vaccine was administered at day 1 and birds were monitored serologically (haemagglutination inhibition test) and virologically for 19 weeks. Maternally-derived antibody and post-vaccination H5 antibody titres were measured using the Chinese (A/Goose/Guangdong/1/96(H5N1)) HA and the Egyptian (A/chicken/Egypt/128s/2012(H5N1)) HA as antigens. The challenge was conducted at 19 weeks of age and on six experimental groups: Groups I (WB) and II (BS), both vaccinated and challenged; Groups III (WB) and IV (BS), both vaccinated but not challenged; Groups V and VI, unvaccinated specific pathogen free chickens, serving respectively as positive and negative controls. The challenge virus was the clade 2.2.1 highly pathogenic avian influenza H5N1 A/chicken/Egypt/128s/2012 at a dose of 10(6) median embryo infective dose. For both breeds, complete maternally-derived antibody waning occurred at the age of 4 weeks. The immune response to rHVT-H5 vaccination was detected from the sixth week. The seroconversion rates for both breeds reached 85.7 to 100% in the eighth week of age. Protection levels of 73.3%, 60% and 0% were respectively recorded in Groups I, II and V. No mortalities occurred in the unchallenged groups. Group I showed superior results for all measured post-challenge parameters. In conclusion, a single rHVT-H5 hatchery vaccination conferred a high level of protection for a relatively extended period. This vaccine could be an important tool for future A/H5N1 prevention/control in endemic countries. Further studies on persistence of immunity beyond 19 weeks, need for booster with inactivated vaccines, breed susceptibility and vaccinal response, and transmissibility are recommended.


Asunto(s)
Pollos/inmunología , Herpesvirus Meleágrido 1/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Gripe Aviar/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Vacunación/veterinaria , Animales , Egipto , Femenino , Vacunas contra la Influenza/inmunología , Gripe Aviar/inmunología , Gripe Aviar/virología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Organismos Libres de Patógenos Específicos , Vacunas de Productos Inactivados/inmunología , Vacunas Sintéticas
10.
Vaccine ; 42(15): 3410-3419, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38641498

RESUMEN

The application of recombinant herpesvirus of turkey, expressing the H9 hemagglutinin gene from low pathogenic avian influenza virus (LPAIV) H9N2 and the avian orthoavulavirus-1 (AOAV-1) (commonly known as Newcastle Disease virus (NDV)) fusion protein (F) as an rHVT-H9-F vaccine, is an alternative to currently used classical vaccines. This study investigated H9- and ND-specific humoral and mucosal responses, H9-specific cell-mediated immunity, and protection conferred by the rHVT-H9-F vaccine in specific pathogen-free (SPF) chickens. Vaccination elicited systemic NDV F- and AIV H9-specific antibody response but also local antibodies in eye wash fluid and oropharyngeal swabs. The ex vivo H9-specific stimulation of splenic and pulmonary T cells in the vaccinated group demonstrated the ability of vaccination to induce systemic and local cellular responses. The clinical protection against a challenge using a LPAIV H9N2 strain of the G1 lineage isolated in Morocco in 2016 was associated with a shorter duration of shedding along with reduced viral genome load in the upper respiratory tract and reduced cloacal shedding compared to unvaccinated controls.


Asunto(s)
Anticuerpos Antivirales , Pollos , Subtipo H9N2 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Aviar , Esparcimiento de Virus , Animales , Subtipo H9N2 del Virus de la Influenza A/inmunología , Subtipo H9N2 del Virus de la Influenza A/genética , Pollos/inmunología , Gripe Aviar/prevención & control , Gripe Aviar/inmunología , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Esparcimiento de Virus/inmunología , Organismos Libres de Patógenos Específicos , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/genética , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Inmunidad Celular , Herpesvirus Meleágrido 1/inmunología , Herpesvirus Meleágrido 1/genética , Vacunación/métodos , Inmunidad Humoral , Vectores Genéticos/inmunología , Inmunogenicidad Vacunal , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/administración & dosificación , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética
12.
Vaccine ; 41(1): 145-158, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36411134

RESUMEN

In France during winter 2016-2017, 487 outbreaks of clade 2.3.4.4b H5N8 subtype high pathogenicity (HP) avian influenza A virus (AIV) infections were detected in poultry and captive birds. During this epizootic, HPAIV A/decoy duck/France/161105a/2016 (H5N8) was isolated and characterized in an experimental infection transmission model in conventional mule ducks. To investigate options to possibly protect such ducks against this HPAIV, three vaccines were evaluated in controlled conditions. The first experimental vaccine was derived from the hemagglutinin gene of another clade 2.3.4.4b A(H5N8) HPAIV. It was injected at three weeks of age, either alone (Vac1) or after a primer injection at day-old (Vac1 + boost). The second vaccine (Vac2) was a commercial bivalent adjuvanted vaccine containing an expressed hemagglutinin modified from a clade 2.3.2 A(H5N1) HPAIV. Vac2 was administered as a single injection at two weeks of age. The third experimental vaccine (Vac3) also incorporated a homologous 2.3.4.4b H5 HA gene and was administered as a single injection at three weeks of age. Ducks were challenged with HPAIV A/decoy duck/France/161105a/2016 (H5N8) at six weeks of age. Post-challenge virus excretion was monitored in vaccinated and control birds every 2-3 days for two weeks using real-time reverse-transcription polymerase chain reaction and serological analyses (haemagglutination inhibition test against H5N8, H5 ELISA and AIV ELISA) were performed. Vac1 abolished oropharyngeal and cloacal shedding to almost undetectable levels, whereas Vac3 abolished cloacal shedding only (while partially reducing respiratory shedding) and Vac2 only partly reduced the respiratory and intestinal excretion of the challenge virus. These results provided relevant insights in the immunogenicity of recombinant H5 vaccines in mule ducks, a rarely investigated hybrid between Pekin and Muscovy duck species that has played a critical role in the recent H5 HPAI epizootics in France.


Asunto(s)
Patos , Subtipo H5N1 del Virus de la Influenza A , Subtipo H5N8 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Aviar , Enfermedades de las Aves de Corral , Animales , Equidae , Hemaglutininas , Enfermedades de las Aves de Corral/prevención & control , Vacunas Sintéticas , Virulencia
13.
Emerg Infect Dis ; 18(12): 2076-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23171740

RESUMEN

We investigated the influence of a mass poultry vaccination campaign on passive surveillance of highly pathogenic avian influenza subtype (H5N1) outbreaks among poultry in Egypt. Passive reporting dropped during the campaign, although probability of infection remained unchanged. Future poultry vaccination campaigns should consider this negative impact on reporting for adapting surveillance strategies.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Vacunas contra la Influenza , Gripe Aviar/epidemiología , Gripe Aviar/prevención & control , Gripe Humana/epidemiología , Animales , Egipto/epidemiología , Humanos , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunación Masiva/veterinaria , Aves de Corral , Vigilancia en Salud Pública , Estaciones del Año
14.
Virus Res ; 319: 198877, 2022 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-35872282

RESUMEN

The control of infectious bronchitis (IB) is largely based on routine vaccine administration, often using live-attenuated vaccines. However, their capability to replicate and be transmitted among animals and farms implies significant risks. The detection of strains genetically related to vaccines complicates the diagnostic process and understanding of the viral molecular epidemiology. Moreover, reversion to virulence and associated clinical outbreaks can occur although the underlying mechanism are often unknown. In the present study, three vaccine vials, based on IBV GI-23 lineage (also known as Variant2) were deep sequenced through Next Generation Sequencing (NGS) to investigate the presence and features of viral subpopulations. To elucidate the consequences in the field and identify potential markers suitable for a DIVA strategy, the S1 sequences of strains originating from farms in different countries were sequenced and classified based on the knowledge of their vaccination history and similarity with the applied vaccine. Although all considered vaccine batches shared the same consensus sequence, different subpopulations were identified suggesting independent and poorly constrained evolutionary processes. When compared with strains sampled from farms, the vaccine consensus sequences and the respective subpopulations clustered with vaccine strains and no genetic features were consistently shared with field strains. Therefore, if vaccine-induced outbreaks occur, they are more likely to originate from in vivo evolution rather than selection of already present subpopulations. Although some amino acid residues were most commonly detected in field or vaccine strains, no consistent marker could be identified. The occurrence of subpopulations within IBV GI-23-based vaccines and variability featuring different production batches was demonstrated. Being such a phenomenon apparently driven by random genetic drift rather than directional selection, the differentiation between field and vaccine-derived strains appears extremely challenging based on sequence analysis alone. The knowledge of farm management and vaccination history should thus be considered for a proper epidemiological investigation.


Asunto(s)
Infecciones por Coronavirus , Virus de la Bronquitis Infecciosa , Enfermedades de las Aves de Corral , Vacunas Virales , Animales , Pollos , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/veterinaria , Virus de la Bronquitis Infecciosa/genética , Vacunas Atenuadas/genética , Vacunas Virales/genética
15.
Front Vet Sci ; 9: 1057040, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36825205

RESUMEN

The first Food and Agriculture Organization of the United Nations (FAO) Action Plan on antimicrobial resistance (AMR), published in 2016, identified the need to develop capacity for AMR surveillance and monitoring in food and agriculture sectors. As part of this effort, FAO has developed the "Assessment Tool for Laboratories and AMR Surveillance Systems" (FAO-ATLASS) to assist countries in systematically assessing their AMR surveillance system in food and agriculture. FAO-ATLASS includes two different modules for surveillance and laboratory assessment. Each module includes two questionnaires that collect either qualitative or semi-quantitative data to describe and score the performance of national AMR surveillance system data production network, data collection and analysis, governance, communication and overall sustainability in a standardized manner. Based on information captured in the questionnaire by trained assessors (1) tables and figures describing the outputs of the surveillance system are automatically generated (2) a Progressive Improvement Pathway (PIP) stage, ranging from "1-limited" to "5-sustainable", is assigned to each laboratory assessed in the country, each area of the surveillance system and also to the overarching national AMR surveillance system. FAO-ATLASS allows national authorities to implement a strategic stepwise approach to improving their AMR surveillance systems via the FAO-ATLASS PIP system and provides an evidence base for actions and advocacy. The implementation of FAO-ATLASS at regional and global levels can contribute to harmonize and better coordinate strategies aimed at implementing an integrated AMR surveillance system under the One Health approach.

16.
Acta Trop ; 216: 105787, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33385361

RESUMEN

Despite declaration as a national priority disease, dog rabies remains endemic in Liberia, with surveillance systems and disease control activities still developing. The objective of these initial efforts was to establish animal rabies diagnostics, foster collaboration between all rabies control stakeholders, and develop a short-term action plan with estimated costs for rabies control and elimination in Liberia. Four rabies diagnostic tests, the direct fluorescent antibody (DFA) test, the direct immunohistochemical test (dRIT), the reverse transcriptase polymerase chain reaction (RT-PCR) assay and the rapid immunochromatographic diagnostic test (RIDT), were implemented at the Central Veterinary Laboratory (CVL) in Monrovia between July 2017 and February 2018. Seven samples (n=7) out of eight suspected animals were confirmed positive for rabies lyssavirus, and molecular analyses revealed that all isolates belonged to the Africa 2 lineage, subgroup H. During a comprehensive in-country One Health rabies stakeholder meeting in 2018, a practical workplan, a short-term action plan and an accurately costed mass dog vaccination strategy were developed. Liberia is currently at stage 1.5/5 of the Stepwise Approach towards Rabies Elimination (SARE) tool, which corresponds with countries that are scaling up local-level interventions (e.g. dog vaccination campaigns) to the national level. Overall an estimated 5.3 - 8 million USD invested over 13 years is needed to eliminate rabies in Liberia by 2030. Liberia still has a long road to become free from dog-rabies. However, the dialogue between all relevant stakeholders took place, and disease surveillance considerably improved through implementing rabies diagnosis at the CVL. The joint efforts of diverse national and international stakeholders laid important foundations to achieve the goal of zero dog-mediated human rabies deaths by 2030.


Asunto(s)
Pruebas Diagnósticas de Rutina/veterinaria , Vacunas Antirrábicas/administración & dosificación , Rabia/diagnóstico , Rabia/prevención & control , Animales , ADN Viral , Pruebas Diagnósticas de Rutina/métodos , Enfermedades de los Perros/diagnóstico , Enfermedades de los Perros/prevención & control , Enfermedades de los Perros/virología , Perros/virología , Femenino , Humanos , Liberia/epidemiología , Masculino , Vacunación Masiva/veterinaria , Técnicas de Diagnóstico Molecular/métodos , Técnicas de Diagnóstico Molecular/veterinaria , Filogenia , Reacción en Cadena de la Polimerasa , Vacunas Antirrábicas/economía , Virus de la Rabia/genética , Virus de la Rabia/aislamiento & purificación
17.
Avian Dis ; 54(1 Suppl): 380-3, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20521664

RESUMEN

The World Organisation for Animal Health (OIE)/United Nations Food and Agriculture Organization (FAO) joint network of expertise on animal influenza (OFFLU) includes all ten OIE/FAO reference laboratories and collaborating centers for avian influenza, other diagnostic laboratories, research and academic institutions, and experts in the fields of virology, epidemiology, vaccinology, and molecular biology. OFFLU has made significant progress in improving its infrastructure, in identifying and addressing technical gaps, and in establishing associations among leading veterinary institutions. Interaction with the World Health Organization (WHO) Global Influenza Program is also critical, and mechanisms for permanent interaction are being developed. OFFLU played a key role in the WHO/OIE/FAO Joint Technical Consultation held in Verona (October 7-9, 2008), which provided an opportunity to highlight and share knowledge and identify potential gaps regarding issues at the human-animal interface for avian influenza. OFFLU experts also contributed to the working group for the Unified Nomenclature System for H5N1 influenza viruses based on hemagglutinin gene phylogeny (WHO/OIE/FAO, H5N1 Evolution Working Group, Towards a unified nomenclature system for highly pathogenic avian influenza virus (H5N1) in Emerging Infectious Diseases 14:el, 2008). OFFLU technical activities, led by expert scientists from OIE/FAO reference institutions and coordinated by OIE and FAO focal points, have been prioritized to include commercial diagnostic kit evaluation, applied epidemiology, biosafety, vaccination, proficiency testing, development of standardized reference materials for sera and RNA, and issues at the human-animal interface. The progress to date and future plans for these groups will be presented. OFFLU is also involved in two national projects implemented by FAO in Indonesia and Egypt that seek to establish sustainable mechanisms for monitoring virus circulation, including viral characterization, and for streamlining the process to update poultry vaccines for avian influenza.


Asunto(s)
Salud Global , Gripe Aviar/prevención & control , Cooperación Internacional , Naciones Unidas , Animales , Virus de la Influenza A/clasificación , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/fisiología , Gripe Aviar/epidemiología , Gripe Aviar/virología , Aves de Corral , Salud Pública
18.
PLoS One ; 15(3): e0230567, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32196515

RESUMEN

Poultry production has significantly increased worldwide, along with the number of avian influenza (AI) outbreaks and the potential threat for human pandemic emergence. The role of wild bird movements in this global spread has been extensively studied while the role of animal, human and fomite movement within commercial poultry production and trade networks remains poorly understood. The aim of this work is to better understand these roles in relation to the different routes of AI spread. A scoping literature review was conducted according to the PRISMA guidelines (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) using a search algorithm combining twelve domains linked to AI spread and animal/human movements within poultry production and trade networks. Only 28 out of 3,978 articles retrieved dealt especially with the role of animal, human and fomite movements in AI spread within the international trade network (4 articles), the national trade network (8 articles) and the production network (16 articles). While the role of animal movements in AI spread within national trade networks has been largely identified, human and fomite movements have been considered more at risk for AI spread within national production networks. However, the role of these movements has never been demonstrated with field data, and production networks have only been partially studied and never at international level. The complexity of poultry production networks and the limited access to production and trade data are important barriers to this knowledge. There is a need to study the role of animal and human movements within poultry production and trade networks in the global spread of AI in partnership with both public and private actors to fill this gap.


Asunto(s)
Gripe Aviar/epidemiología , Aves de Corral/virología , Crianza de Animales Domésticos , Animales , Comercio , Humanos , Virus de la Influenza A/aislamiento & purificación , Internacionalidad , Productos Avícolas/virología
19.
Prev Vet Med ; 184: 105129, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33002655

RESUMEN

France recently faced two epizootic waves of highly pathogenic avian influenza (HPAI) in poultry (H5N6 in 2015-2016 and H5N8 in 2016-2017), mainly in the fattening duck production sector. Vaccination against avian influenza (AI) is currently not authorised in France even though its potential benefits were discussed during these epizootic events. The objective of this work was to evaluate the potential efficiency of different vaccination strategies that could be applied against AI in France. The EVACS tool, which is a decision support tool developed to evaluate vaccination strategies, was applied in several French poultry production sectors: broiler, layer, turkey, duck and guinea fowl. EVACS was used to simulate the performance of vaccination strategies in terms of vaccination coverage, immunity levels and spatial distribution of the immunity level. A cost-benefit analysis was then applied based on EVACS results to identify the most efficient strategy. For each sector, vaccination protocols were tested according to the production type (breeders/production, indoor/outdoor), the integration level (integrated/independent) and the type of vaccine (hatchery vaccination using a recombinant vaccine/farm vaccination using an inactivated vaccine). The most efficient protocols for each sector were then combined to test different overall vaccination strategies at the national level. Even if it was not possible to compare vaccination protocols with the two vaccines types in "foie gras" duck, meat duck and guinea fowl production sectors as no hatchery vaccine currently exist for these species, these production sectors were also described and included in this simulation. Both types of vaccination (at hatchery and farm level) enabled protective immunity levels for the control of AI, but higher poultry population immunity level was reached (including independent farms) using hatchery vaccination. We also showed that hatchery vaccination was more efficient (higher benefit-cost ratio) than farm vaccination. Sufficient and homogeneously spatially distributed protective levels were reached in the overall poultry population with vaccination strategies targeting breeders, chicken layers and broilers and turkeys, without the need to include ducks and guinea fowls. However, vaccination strategies involving the highest number of species and production types were the most efficient in terms of cost-benefit. This study provides critical information on the efficiency of different vaccination strategies to support future decision making in case vaccination was applied to prevent and control HPAI in France.


Asunto(s)
Control de Enfermedades Transmisibles/métodos , Técnicas de Apoyo para la Decisión , Brotes de Enfermedades/veterinaria , Patos , Galliformes , Gripe Aviar/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Vacunación/veterinaria , Animales , Pollos , Francia/epidemiología , Gripe Aviar/epidemiología , Gripe Aviar/virología , Enfermedades de las Aves de Corral/epidemiología , Enfermedades de las Aves de Corral/virología , Pavos
20.
Avian Dis ; 63(sp1): 193-202, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31131577

RESUMEN

The most recent pandemic clade of highly pathogenic avian influenza (HPAI) H5, clade 2.3.4.4, spread widely, with the involvement of wild birds, most importantly wild waterfowl, carrying the virus (even asymptomatically) from Asia to North America, Europe, and Africa. Domestic waterfowl being in regular contact with wild birds played a significant role in the H5Nx epizootics. Therefore, protection of domestic waterfowl from H5Nx avian influenza infection would likely cut the transmission chain of these viruses and greatly enhance efforts to control and prevent disease outbreak in other poultry and animal species, as well as infection of humans. The expectation for such a vaccine is not only to provide clinical protection, but also to control challenge virus transmission efficiently and ensure that the ability to differentiate infected from vaccinated animals is retained. A water-in-oil emulsion virus-like particle vaccine, containing homologous hemagglutinin antigen to the current European H5N8 field strains, has been developed to meet these requirements. The vaccine was tested in commercial Pekin and mule ducks by vaccinating them either once, at 3 wk of age, or twice (at 1 day and at 3 wk of age). Challenge was performed at 6 wk of age with a Hungarian HPAIV H5N8 isolate (2.3.4.4 Group B). Efficacy of vaccination was evaluated on the basis of clinical signs, amount of virus shedding, and transmission. Vaccination resulted in complete clinical protection and prevention of challenge virus transmission from the directly challenged vaccinated ducks to the vaccinated contact animals.


Una vacuna basada en partículas similares a virus proporciona un alto nivel de protección contra el desafío con un virus homólogo de influenza aviar de alta patogenicidad H5N8 en patos mula y Pekin, incluida la prevención de la transmisión. El clado pandémico más reciente de influenza aviar altamente patógena H5, clado 2.3.4.4, se diseminó ampliamente, con la participación de aves silvestres, siendo las aves acuáticas más importantes, portando el virus (incluso asintomáticamente) de Asia a América del Norte, Europa, y África. Las aves acuáticas domésticas en contacto regular con aves silvestres desempeñaron un papel importante en las epizootias H5Nx. Por lo tanto, la protección de las aves acuáticas domésticas contra la infección por influenza aviar H5Nx probablemente cortaría la cadena de transmisión de estos virus y aumentaría en gran medida los esfuerzos para controlar y prevenir brotes de enfermedades en otras aves comerciales y especies animales, así como la infección en humanos. La expectativa de una vacuna de este tipo es no solo brindar protección clínica, sino también controlar la transmisión del virus de desafío de manera eficiente y garantizar que se mantenga la capacidad de diferenciar a los animales vacunados. Se ha desarrollado una vacuna emulsionada en aceite con partículas similares al virus, que contiene el antígeno de hemaglutinina homóloga a las cepas de campo H5N8 europeas actuales, para cumplir con estos requisitos. La vacuna se probó en patos de Pekín y mulas comerciales, vacunándolos una vez, a las tres semanas de edad, o dos veces (al primer día y a las tres semanas de edad). El desafío se realizó a las seis semanas de edad con un aislado de alta patogenicidad H5N8 húngaro (2.3.4.4 Grupo B). La eficacia de la vacunación se evaluó en función de los signos clínicos, la eliminación viral y la transmisión. La vacunación dio como resultado una protección clínica completa y la prevención de la transmisión del virus de desafío de los patos vacunados.


Asunto(s)
Patos , Subtipo H5N8 del Virus de la Influenza A/efectos de los fármacos , Gripe Aviar/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Vacunas de Partículas Similares a Virus/farmacología , Vacunas Virales/farmacología , Animales , Patos/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/administración & dosificación , Gripe Aviar/transmisión , Enfermedades de las Aves de Corral/transmisión , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA