Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 332
Filtrar
1.
Mol Biol Rep ; 51(1): 1032, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39352412

RESUMO

BACKGROUND: Peste des Petits Ruminants (PPR) is an economically significant transboundary viral disease of sheep and goats caused by the PPRV virus, affecting annual losses of 1.45-2.10 billion US dollars globally. We designed the current study to evaluate the positive cases, molecular characterization, phylogenetic analysis, and risk factors correlated with the disease in various districts of Khyber Pakhtunkhwa, Pakistan, with the aim of contributing to these strategies. METHODS AND RESULTS: A total of 384 samples from three selected districts, i.e., Peshawar, Charsadda and Chitral (n = 128 each), were collected, and the virus was investigated by using the sandwich ELISA, while the N gene of the virus was used as a target for molecular detection via RT-PCR. The confirmed samples were then sequenced, and phylogenetic analysis was performed. According to our findings, the highest positive cases was found in district Peshawar (50.87%), followed by Charsadda and Chitral (24.56%), respectively, while risk factor analysis showed that certain categories, such as species, sex, and age less than two years, have higher risk (P < 0.05) in contrast to their respective categories. Furthermore, sequencing and phylogenetic analysis of representative samples showed that the PPRV strains in the current study clustered in lineage IV, which is circulating in the small ruminant population of Asia, the Middle East, and African countries. Comparative residue analysis highlighted the mutation by representing 242 variable sites out of 371 locations. CONCLUSIONS: PPRV has foremost importance in Pakistan because the virus was detected in a considerable number of samples, and most of which were sourced from subsidiary areas where veterinary services are not prioritized.


Assuntos
Doenças das Cabras , Cabras , Peste dos Pequenos Ruminantes , Vírus da Peste dos Pequenos Ruminantes , Filogenia , Doenças dos Ovinos , Animais , Vírus da Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/isolamento & purificação , Paquistão/epidemiologia , Cabras/virologia , Ovinos/virologia , Peste dos Pequenos Ruminantes/virologia , Peste dos Pequenos Ruminantes/epidemiologia , Fatores de Risco , Doenças das Cabras/virologia , Doenças das Cabras/epidemiologia , Doenças dos Ovinos/virologia , Doenças dos Ovinos/epidemiologia , Feminino , Masculino
2.
Clin Lab ; 70(9)2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39257133

RESUMO

BACKGROUND: HEV is endemic in several Middle Eastern countries including Saudi Arabia, which hosts the annual pilgrimage for Muslims from around the world. One of the Hajj rituals is the sacrifice of animals, including camels, cows, goats, and sheep. HEV Zoonosis is established in swine and other suspected species, including deer, rabbits, dromedary, and Bactrian camels. HEV was identified in small, domesticized animals like goats, cows, sheep, and horses. We previously investigated HEV seroprevalence in Camels. This study aimed to evaluate HEV seroprevalence in other highly consumed ruminants in Saudi Arabia, namely cows, sheep, and goats. METHODS: Sera from cows (n = 47), goats (n = 56), and sheep (n = 67) were analyzed for the presence of HEV-IgG by using in-house developed ELISA assays. RESULTS: The highest seroprevalence was found in sheep (62.7%), followed by cows (38.3%), and then goats (14.3%), with a p-value of < 0.001. No other demographic characteristics of the animals were significantly correlated with the HEV seroprevalence. CONCLUSIONS: This study provides baseline data as the first study on the seroprevalence of HEV in ruminant animals in Saudi Arabia. The high seroprevalence found in sheep and cows must be further investigated for the potential zoonotic HEV transmission to humans. Further studies are needed to investigate the active viremia in these animal species through nucleic acid detection and sequencing to provide data on the circulating HEV genotypes among the targeted animal species. The detection of HEV in different animal products, such as milk, liver, and others, also remains an important study area to consider.


Assuntos
Cabras , Vírus da Hepatite E , Hepatite E , Ruminantes , Animais , Vírus da Hepatite E/genética , Vírus da Hepatite E/imunologia , Vírus da Hepatite E/isolamento & purificação , Hepatite E/epidemiologia , Hepatite E/veterinária , Hepatite E/virologia , Estudos Soroepidemiológicos , Cabras/virologia , Ovinos , Arábia Saudita/epidemiologia , Bovinos , Ruminantes/virologia , Feminino , Ensaio de Imunoadsorção Enzimática , Imunoglobulina G/sangue , Zoonoses/virologia , Zoonoses/epidemiologia , Zoonoses/diagnóstico , Anticorpos Anti-Hepatite/sangue , Doenças das Cabras/virologia , Doenças das Cabras/epidemiologia , Doenças das Cabras/diagnóstico , Doenças das Cabras/sangue , Masculino
3.
Emerg Infect Dis ; 30(10): 2191-2193, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39320366

RESUMO

A Rift Valley fever epizootic affected livestock in Rwanda during March-October 2022. We confirmed 3,112 infections with the virus, including 1,342 cases, 1,254 abortions, and 516 deaths among cattle, goats, and sheep. We recommend a One Health strategy for investigations and response to protect animal and human health.


Assuntos
Cabras , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Febre do Vale de Rift/epidemiologia , Ruanda/epidemiologia , Animais , Ovinos , Humanos , Cabras/virologia , Bovinos , Surtos de Doenças , Gado/virologia , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/virologia , Doenças das Cabras/epidemiologia , Doenças das Cabras/virologia , Doenças dos Ovinos/epidemiologia , Doenças dos Ovinos/virologia
4.
Viruses ; 16(8)2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39205187

RESUMO

Astroviruses are single-stranded, positive-sense RNA viruses capable of infecting humans as well as a wide range of mammalian and avian species, with a length of approximately 6.6-7.7 kb. In this study, 139 goat fecal samples collected from the Guangxi province were used for the RT-PCR detection, and two of these were positive for goat astrovirus, with a positivity rate of 1.44% (2/139). The complete genome sequence of an astrovirus strain and the partial genome sequence of a strain astrovirus, named GX WZ 2023 and GX HC 2023, were amplified and sequenced, and their sequence lengths were 6284 nt and 6213 nt, respectively. Among them, the capsid protein of goat astrovirus GX HC 2023 showed the highest amino acid identity of 95.9% with ovine astrovirus GX, which belonged to the MAstV-2 genotype. However, the closest relative of the GX WZ 2023 strain was found to be the caprine astrovirus Sichuan, with a nucleotide sequence identity of 76.8%. The ORF1ab nonstructural protein of this strain showed the highest amino acid identities of 89.2 and 95.8% with the ovine astrovirus S5.1 and caprine astrovirus G5.1 strains, respectively. However, its ORF2 capsid protein has 68.4% amino acid identity with the bovine astrovirus (BAstV) 16 2021 CHN strain and only 21.9-64% amino acid identity with all available strains of goat astrovirus. The GX WZ 2023 strain was recombined with the Chinese (BAstV 16 2021 CHN) and Japanese bovine strains (BAstV JPN 2015) in the ORF2 region. Therefore, the goat astrovirus GX WZ 2023 is proposed as a new member of the family goat astroviridae based on the species classification criteria of the International Committee on Taxonomy of Viruses. These findings enhance our understanding of the prevalence and genetic evolution of goat astrovirus and provide a scientific basis for future studies of these viruses in other animals.


Assuntos
Infecções por Astroviridae , Genoma Viral , Genótipo , Doenças das Cabras , Cabras , Mamastrovirus , Filogenia , Animais , Cabras/virologia , China/epidemiologia , Infecções por Astroviridae/veterinária , Infecções por Astroviridae/virologia , Infecções por Astroviridae/epidemiologia , Doenças das Cabras/virologia , Doenças das Cabras/epidemiologia , Mamastrovirus/genética , Mamastrovirus/classificação , Mamastrovirus/isolamento & purificação , Fezes/virologia , Proteínas do Capsídeo/genética , Recombinação Genética , RNA Viral/genética , Astroviridae/genética , Astroviridae/classificação , Astroviridae/isolamento & purificação , Ovinos , Análise de Sequência de DNA
5.
Prev Vet Med ; 232: 106328, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39191049

RESUMO

Bluetongue virus (BT) is a vector-borne virus that causes a disease, called bluetongue, which results in significant economic loss and morbidity in sheep, cattle, goats and wild ungulates across all continents of the world except Antarctica. Despite the geographical breadth of its impact, most BT epidemiological models are informed by parameters derived from the 2006-2009 BTV-8 European outbreak. The aim of this study was to develop a highly adaptable model for BT which could be used elsewhere in the world, as well as to identify the parameters which most influence outbreak dynamics, so that policy makers can be properly informed with the most current information to aid in disease planning. To provide a framework for future outbreak modelling and an updated parameterisation that reflects natural variation in infections, a newly developed and parameterised two-host, two-vector species ordinary differential equation model was formulated and analysed. The model was designed to be adaptable to be implemented in any region of the world and able to model both epidemic and endemic scenarios. It was parameterised using a systematic literature review of host-to-vector and vector-to-host transmission rates, host latent periods, host infectious periods, and vaccine protection factors. The model was demonstrated using the updated parameters, with South Africa as a setting based on the Western Cape's known cattle and sheep populations, local environmental parameters, and Culicoides spp. presence data. The sensitivity analysis identified that the duration of the infectious period for sheep and cows had the greatest impact on the outbreak length and number of animals infected at the peak of the outbreak. Transmission rates from cows and sheep to C. imicola midges greatly influenced the day on which the peak of the outbreak occurred, along with the duration of incubation period, and infectious period for cows. Finally, the protection factor of the vaccine had the greatest influence on the total number of animals infected. This knowledge could aid in the development of control measures. Due to gradual climate and anthropological change resulting in alterations in vector habitat suitability, BT outbreaks are likely to continue to increase in range and frequency. Therefore, this research provides an updated BT modelling framework for future outbreaks around the world to explore transmission, outbreak dynamics and control measures.


Assuntos
Vírus Bluetongue , Bluetongue , Doenças dos Bovinos , Surtos de Doenças , Animais , Bovinos , Bluetongue/epidemiologia , Bluetongue/transmissão , Bluetongue/virologia , Bluetongue/prevenção & controle , Vírus Bluetongue/patogenicidade , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/virologia , Doenças dos Bovinos/transmissão , Ceratopogonidae/virologia , Surtos de Doenças/prevenção & controle , Surtos de Doenças/veterinária , Cabras/virologia , Insetos Vetores/virologia , Ovinos/virologia , África do Sul/epidemiologia , Modelos Epidemiológicos
6.
Commun Biol ; 7(1): 937, 2024 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-39095591

RESUMO

Peste des petits ruminants virus (PPRV) is a multi-host pathogen with sheep and goats as main hosts. To investigate the role of cattle in the epidemiology of PPR, we simulated conditions similar to East African zero-grazing husbandry practices in a series of trials with local Zebu cattle (Bos taurus indicus) co-housed with goats (Capra aegagrus hircus). Furthermore, we developed a mathematical model to assess the impact of PPRV-transmission from cattle to goats. Of the 32 cattle intranasally infected with the locally endemic lineage IV strain PPRV/Ethiopia/Habru/2014 none transmitted PPRV to 32 co-housed goats. However, these cattle or cattle co-housed with PPRV-infected goats seroconverted. The results confirm previous studies that cattle currently play a negligible role in PPRV-transmission and small ruminant vaccination is sufficient for eradication. However, the possible emergence of PPRV strains more virulent for cattle may impact eradication. Therefore, continued monitoring of PPRV circulation and evolution is recommended.


Assuntos
Doenças das Cabras , Cabras , Peste dos Pequenos Ruminantes , Vírus da Peste dos Pequenos Ruminantes , Animais , Peste dos Pequenos Ruminantes/transmissão , Peste dos Pequenos Ruminantes/virologia , Peste dos Pequenos Ruminantes/epidemiologia , Bovinos , Vírus da Peste dos Pequenos Ruminantes/imunologia , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Cabras/virologia , Doenças das Cabras/virologia , Doenças das Cabras/transmissão , Doenças dos Bovinos/transmissão , Doenças dos Bovinos/virologia , Doenças dos Bovinos/epidemiologia , Erradicação de Doenças/métodos
7.
Arch Virol ; 169(8): 172, 2024 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-39096433

RESUMO

Goatpox and sheeppox are highly contagious and economically important viral diseases of small ruminants. Due to the risk they pose to animal health, livestock production, and international trade, capripoxviruses are a considerable threat to the livestock economy. In this study, we expressed two core proteins (A4L and A12L) and one extracellular enveloped virion protein (A33R) of goatpox virus in a baculovirus expression vector system and evaluated their use as diagnostic antigens in ELISA. Full-length A4L, A12L, and A33R genes of the GTPV Uttarkashi strain were amplified, cloned into the pFastBac HT A donor vector, and introduced into DH10Bac cells containing a baculovirus shuttle vector plasmid to generate recombinant bacmids. The recombinant baculoviruses were produced in Sf-21 cells by transfection, and proteins were expressed in TN5 insect cells. The recombinant proteins were analysed by SDS-PAGE and confirmed by western blot, with expected sizes of ~30 kDa, ~31 kDa, and ~32 kDa for A4L, A12L, and A33R, respectively. The recombinant proteins were purified, and the immunoreactivity of the purified proteins was confirmed by western blot using anti-GTPV serum. The antigenic specificity of the expressed proteins as diagnostic antigens was evaluated by testing their reactivity with infected, vaccinated, and negative GTPV/SPPV serum in indirect ELISA, and the A33R-based indirect ELISA was optimized. The diagnostic sensitivity and specificity of the A33R-based indirect ELISA were found to be of 89% and 94% for goats and 98% and 91%, for sheep, respectively. No cross-reactivity was observed with other related viruses. The recombinant-A33R-based indirect ELISA developed in the present study shows that it has potential for the detection of antibodies in GTPV and SPPV infected/vaccinated animals.


Assuntos
Baculoviridae , Capripoxvirus , Ensaio de Imunoadsorção Enzimática , Doenças das Cabras , Cabras , Proteínas do Envelope Viral , Capripoxvirus/genética , Capripoxvirus/isolamento & purificação , Baculoviridae/genética , Animais , Doenças das Cabras/virologia , Doenças das Cabras/diagnóstico , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Cabras/virologia , Ensaio de Imunoadsorção Enzimática/métodos , Infecções por Poxviridae/diagnóstico , Infecções por Poxviridae/veterinária , Infecções por Poxviridae/virologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/imunologia , Vírion/genética , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Células Sf9 , Antígenos Virais/genética , Antígenos Virais/imunologia , Linhagem Celular , Expressão Gênica
8.
BMC Vet Res ; 20(1): 376, 2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39180076

RESUMO

Rift Valley fever virus (RVFV) is a mosquito-borne RNA virus of the Phlebovirus genus in the phenuviridae family. Its genome is trisegmented with small (S), medium (M) and large (L) fragments. In nature, the virus exists as a single serotype that is responsible for outbreaks of Rift Valley fever (RVF), a zoonotic disease that often occurs in Africa and the Middle East. RVFV genomes are thought to undergo both recombination and reassortment and investigations of these events is important for monitoring the emergence of virulent strains and understanding the evolutionary characteristics of this virus. The aim of this study was to characterize the genomes of RVFV isolates from cattle, sheep, and goats collected during an interepidemic period in Kenya between June 2016 and November 2021. A total of 691 serum samples from cattle (n = 144), goats (n = 185) and sheep (n = 362) were analysed at the Central Veterinary Laboratories. The competitive IgM-capture ELISA, was used to screen the samples; 205 samples (29.67%) tested positive for RVFV. Of the 205 positive samples, 42 (20.5%) were from cattle, 57 (27.8%) from goats, and 106 (51.7%) from sheep. All the IgM-positive samples were further analyzed by qPCR, and 24 (11.71%) tested positive with Ct values ranging from 14.788 to 38.286. Two samples, 201808HABDVS from sheep and 201810CML3DVS from cattle, had Ct values of less than 20.0 and yielded whole genome sequences with 96.8 and 96.4 coverage, respectively. There was no statistically significant evidence of recombination in any of the three segments and also phylogenetic analysis showed no evidence of reassortment in the two isolated RVFV segments when compared with other isolates of different lineages from previous outbreaks whose genomes are deposited in the GenBank. No evidence of reassortment leaves room for other factors to be the most probable contributors of change in virulence, pathogenicity and emergence of highly virulent strains of the RVFV.


Assuntos
Doenças dos Bovinos , Genoma Viral , Doenças das Cabras , Cabras , Filogenia , Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Doenças dos Ovinos , Animais , Cabras/virologia , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/isolamento & purificação , Ovinos , Febre do Vale de Rift/virologia , Febre do Vale de Rift/epidemiologia , Bovinos , Quênia/epidemiologia , Doenças das Cabras/virologia , Doenças das Cabras/epidemiologia , Doenças dos Ovinos/virologia , Doenças dos Ovinos/epidemiologia , Doenças dos Bovinos/virologia , Doenças dos Bovinos/epidemiologia , Surtos de Doenças/veterinária
9.
Sci Rep ; 14(1): 15166, 2024 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-38956077

RESUMO

The study aimed to investigate molecularly the presence of flea-borne viruses in infested small ruminants with fleas. It was carried out in Egypt's Northern West Coast (NWC) and South Sinai Governorate (SSG). Three specific primers were used targeting genes, ORF103 (for Capripoxvirus and Lumpy skin disease virus), NS3 (for Bluetongue virus), and Rdrp (for Coronavirus), followed by gene sequencing and phylogenetic analyses. The results revealed that 78.94% of sheep and 65.63% of goats were infested in the NWC area, whereas 49.76% of sheep and 77.8% of goats were infested in the SSG region. Sheep were preferable hosts for flea infestations (58.9%) to goats (41.1%) in the two studied areas. Sex and age of the animals had no effects on the infestation rate (p > 0.05). The season and site of infestation on animals were significantly different between the two areas (p < 0.05). Ctenocephalides felis predominated in NWC and Ctenocephalides canis in SSG, and males of both flea species were more prevalent than females. Molecular analysis of flea DNA revealed the presence of Capripoxvirus in all tested samples, while other viral infections were absent. Gene sequencing identified three isolates as sheeppox viruses, and one as goatpox virus. The findings suggest that Capripoxvirus is adapted to fleas and may be transmitted to animals through infestation. This underscores the need for ongoing surveillance of other pathogens in different regions of Egypt.


Assuntos
Filogenia , Sifonápteros , Animais , Egito/epidemiologia , Ovinos , Sifonápteros/virologia , Cabras/virologia , Capripoxvirus/genética , Capripoxvirus/isolamento & purificação , Capripoxvirus/classificação , Infestações por Pulgas/epidemiologia , Infestações por Pulgas/veterinária , Masculino , Feminino , Doenças dos Ovinos/virologia , Doenças dos Ovinos/epidemiologia , Doenças das Cabras/virologia , Doenças das Cabras/epidemiologia
10.
Viruses ; 16(5)2024 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-38793568

RESUMO

The hepatitis E virus is a serious health concern worldwide, with 20 million cases each year. Growing numbers of autochthonous HEV infections in industrialized nations are brought on via the zoonotic transmission of HEV genotypes 3 and 4. Pigs and wild boars are the main animal reservoirs of HEV and play the primary role in HEV transmission. Consumption of raw or undercooked pork meat and close contact with infected animals are the most common causes of hepatitis E infection in industrialized countries. However, during the past few years, mounting data describing HEV distribution has led experts to believe that additional animals, particularly domestic ruminant species (cow, goat, sheep, deer, buffalo, and yak), may also play a role in the spreading of HEV. Up to now, there have not been enough studies focused on HEV infections associated with animal milk and the impact that they could have on the epidemiology of HEV. This critical analysis discusses the role of domestic ruminants in zoonotic HEV transmissions. More specifically, we focus on concerns related to milk safety, the role of mixed farming in cross-species HEV infections, and what potential consequences these may have on public health.


Assuntos
Animais Domésticos , Vírus da Hepatite E , Hepatite E , Leite , Ruminantes , Zoonoses , Animais , Hepatite E/transmissão , Hepatite E/veterinária , Hepatite E/virologia , Vírus da Hepatite E/genética , Vírus da Hepatite E/isolamento & purificação , Leite/virologia , Ruminantes/virologia , Zoonoses/virologia , Zoonoses/transmissão , Humanos , Animais Domésticos/virologia , Zoonoses Virais/transmissão , Zoonoses Virais/virologia , Cabras/virologia , Ovinos/virologia , Genótipo
11.
Virus Genes ; 60(3): 309-313, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38491264

RESUMO

Adenoviruses (AdVs) have been detected in a wide variety of animals. To date, eight types of AdVs in sheep and two types in goats have been identified, which belong to two distinct genera, Mastadenovirus and Atadenovirus. Typically, the term pneumo-enteritis is used to describe adenovirus-induced disease in small ruminants, which has been associated with both enteric and respiratory symptoms of varying severity. The aim of this study was to detect and identify AdVs of small ruminants belonging to the genera Mastadenovirus and Atadenovirus. For this purpose, diagnostic samples (47 lung, 27 intestine, and two pooled tissue samples including intestine and lung) from 49 small ruminants (39 sheep and 10 goats) were used. Following the viral DNA extraction, PCR was carried out by using the primers targeting the hexon gene in order to detect both mast- and atadenoviruses. Sequencing the amplified fragments revealed the presence of three types of ovine adenovirus (OAdV): OAdV-3, OAdV-4, and OAdV-8. Specifically, OAdV-3 was detected in two sheep and a goat while OAdV-4 and OAdV-8 were found in only one sheep each. There is still limited data on the interaction between the viruses in different adenovirus genera and the detected disease, as well as the genetic diversity of adenoviruses, especially in small ruminants. In conclusion, the detection of AdVs in lung and intestinal tissues of small ruminants in this study suggests that these viruses may have contributed to the disease and/or predisposed to other agents.


Assuntos
Infecções por Adenoviridae , Doenças das Cabras , Cabras , Mastadenovirus , Filogenia , Doenças dos Ovinos , Animais , Cabras/virologia , Ovinos/virologia , Doenças dos Ovinos/virologia , Doenças das Cabras/virologia , Infecções por Adenoviridae/veterinária , Infecções por Adenoviridae/virologia , Mastadenovirus/genética , Mastadenovirus/isolamento & purificação , Mastadenovirus/classificação , Turquia , DNA Viral/genética , Análise de Sequência de DNA , Atadenovirus/genética , Atadenovirus/isolamento & purificação , Atadenovirus/classificação , Pulmão/virologia , Adenoviridae/genética , Adenoviridae/isolamento & purificação , Adenoviridae/classificação , Adenoviridae/patogenicidade
12.
J Virol ; 98(4): e0014624, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38440983

RESUMO

Peste des petits ruminants is an acute and highly contagious disease caused by the Peste des petits ruminants virus (PPRV). Host proteins play a crucial role in viral replication. However, the effect of fusion (F) protein-interacting partners on PPRV infection is poorly understood. In this study, we found that the expression of goat plasminogen activator urokinase (PLAU) gradually decreased in a time- and dose-dependent manner in PPRV-infected goat alveolar macrophages (GAMs). Goat PLAU was subsequently identified using co-immunoprecipitation and confocal microscopy as an F protein binding partner. The overexpression of goat PLAU inhibited PPRV growth and replication, whereas silencing goat PLAU promoted viral growth and replication. Additionally, we confirmed that goat PLAU interacted with a virus-induced signaling adapter (VISA) to antagonize F-mediated VISA degradation, increasing the production of type I interferon. We also found that goat PLAU reduced the inhibition of PPRV replication in VISA-knockdown GAMs. Our results show that the host protein PLAU inhibits the growth and replication of PPRV by VISA-triggering RIG-I-like receptors and provides insight into the host protein that antagonizes PPRV immunosuppression.IMPORTANCEThe role of host proteins that interact with Peste des petits ruminants virus (PPRV) fusion (F) protein in PPRV replication is poorly understood. This study confirmed that goat plasminogen activator urokinase (PLAU) interacts with the PPRV F protein. We further discovered that goat PLAU inhibited PPRV replication by enhancing virus-induced signaling adapter (VISA) expression and reducing the ability of the F protein to degrade VISA. These findings offer insights into host resistance to viral invasion and suggest new strategies and directions for developing PPR vaccines.


Assuntos
Doenças das Cabras , Cabras , Interações Hospedeiro-Patógeno , Peste dos Pequenos Ruminantes , Vírus da Peste dos Pequenos Ruminantes , Ativador de Plasminogênio Tipo Uroquinase , Proteínas Virais de Fusão , Animais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína DEAD-box 58/metabolismo , Doenças das Cabras/imunologia , Doenças das Cabras/metabolismo , Doenças das Cabras/virologia , Cabras/imunologia , Cabras/virologia , Macrófagos Alveolares , Peste dos Pequenos Ruminantes/imunologia , Peste dos Pequenos Ruminantes/metabolismo , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/crescimento & desenvolvimento , Vírus da Peste dos Pequenos Ruminantes/imunologia , Vírus da Peste dos Pequenos Ruminantes/metabolismo , Ligação Proteica , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas Virais de Fusão/metabolismo
13.
Virol Sin ; 37(2): 229-237, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35527224

RESUMO

The Getah virus (GETV), a mosquito-borne RNA virus, is widely distributed in Oceania and Asia. GETV is not the only pathogenic to horses, pigs, cattle, foxes and boars, but it can also cause fever in humans. Since its first reported case in Chinese mainland in 2017, the number of GETV-affected provinces has increased to seventeen till now. Therefore, we performed an epidemiologic investigation of GETV in the Xinjiang region, located in northwestern China, during the period of 2017-2020. ELISA was used to analyze 3299 serum samples collected from thoroughbred horse, local horse, sheep, goat, cattle, and pigs, with thoroughbred horse (74.8%), local horse (67.3%), goat (11.7%), sheep (10.0%), cattle (25.1%) and pigs (51.1%) being positive for anti-GETV antibodies. Interestingly, the neutralizing antibody titer in horses was much higher than in other species. Four samples from horses and pigs were positive for GETV according to RT-PCR. Furthermore, from the serum of a local horse, we isolated GETV which was designated as strain XJ-2019-07, and determined its complete genome sequence. From the phylogenetic relationships, it belongs to the Group III lineage. This is the first evidence of GETV associated to domestic animals in Xinjiang. Overall, GETV is prevalent in Xinjiang and probably has been for several years. Since no vaccine against GETV is available in China, detection and monitoring strategies should be improved in horses and pigs, especially imported and farmed, in order to prevent economic losses.


Assuntos
Alphavirus , Culicidae , Alphavirus/genética , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Bovinos/virologia , China/epidemiologia , Culicidae/virologia , Cabras/virologia , Cavalos/virologia , Masculino , Filogenia , Análise de Sequência de DNA , Estudos Soroepidemiológicos , Ovinos/virologia , Suínos/virologia
14.
Viruses ; 13(12)2021 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-34960767

RESUMO

Small ruminant lentiviruses (SRLVs) exist as populations of closely related genetic variants, known as quasispecies, within an individual host. The privileged way of SRLVs transmission in goats is through the ingestion of colostrum and milk of infected does. Thus, characterization of SRLV variants transmitted through the milk, including milk epithelial cells (MEC), may provide useful information about the transmission and evolution of SRLVs. Therefore, the aim of this study was to detect SRLVs in peripheral blood leukocytes (PBLs) and milk epithelial cells of goats naturally infected with SRLVs and perform single nucleotide variations analysis to characterize the extent of genetic heterogeneity of detected SRLVs through comparison of their gag gene sequences. Blood and milk samples from 24 seropositive goats were tested in this study. The double immunolabeling against p28 and cytokeratin demonstrated that milk epithelial cells originated from naturally infected goats were infected by SRLVs. Moreover, PCR confirmed the presence of the integrated SRLVs proviral genome indicating that MECs may have a role as a reservoir of SRLVs and can transmit the virus through milk. The blood and MEC derived sequences from 7 goats were successfully sequenced using NGS and revealed that these sequences were genetically similar. The MEC and blood-derived sequences contained from 3 to 30 (mean, 10.8) and from 1 to 10 (mean, 5.4) unique SNVs, respectively. In five out of seven goats, SNVs occurred more frequent in MEC derived sequences. Non-synonymous SNVs were found in both, PBLs and MEC-derived sequences of analyzed goats and their total number differed between animals. The results of this study add to our understanding of SRLVs genomic variability. Our data provides evidence for the existence of SRLVs quasispecies and to our knowledge, this is the first study that showed quasispecies composition and minority variants of SRLVs present milk epithelial cells.


Assuntos
Cabras/virologia , Lentivirus/isolamento & purificação , Leucócitos/virologia , Leite/virologia , Animais , Células Cultivadas , Células Epiteliais/virologia , Lentivirus/genética , Polimorfismo de Nucleotídeo Único
15.
Viruses ; 13(12)2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34960798

RESUMO

Small ruminant lentiviruses (SRLVs) are a group of highly divergent viruses responsible for global infection in sheep and goats. In a previous study we showed that SRLV strains found in mixed flocks in Poland belonged to subtype A13 and A18, but this study was restricted only to the few flocks from Malopolska region. The present work aimed at extending earlier findings with the analysis of SRLVs in mixed flocks including larger numbers of animals and flocks from different part of Poland. On the basis of gag and env sequences, Polish SRLVs were assigned to the subtypes B2, A5, A12, and A17. Furthermore, the existence of a new subtypes, tentatively designed as A23 and A24, were described for the first time. Subtypes A5 and A17 were only found in goats, subtype A24 has been detected only in sheep while subtypes A12, A23, and B2 have been found in both sheep and goats. Co-infection with strains belonging to different subtypes was evidenced in three sheep and two goats originating from two flocks. Furthermore, three putative recombination events were identified within gag and env SRLVs sequences derived from three sheep. Amino acid (aa) sequences of immunodominant epitopes in CA protein were well conserved while Major Homology Region (MHR) had more alteration showing unique mutations in sequences of subtypes A5 and A17. In contrast, aa sequences of surface glycoprotein exhibited higher variability confirming type-specific variation in the SU5 epitope. The number of potential N-linked glycosylation sites (PNGS) ranged from 3 to 6 in respective sequences and were located in different positions. The analysis of LTR sequences revealed that sequences corresponding to the TATA box, AP-4, AML-vis, and polyadenylation signal (poly A) were quite conserved, while considerable alteration was observed in AP-1 sites. Interestingly, our results revealed that all sequences belonging to subtype A17 had unique substitution T to A in the fifth position of TATA box and did not have a 11 nt deletion in the R region which was noted in other sequences from Poland. These data revealed a complex picture of SRLVs population with ovine and caprine strains belonging to group A and B. We present strong and multiple evidence of dually infected sheep and goats in mixed flocks and present evidence that these viruses can recombine in vivo.


Assuntos
Cabras/virologia , Infecções por Lentivirus/transmissão , Lentivirus/genética , Recombinação Genética , Ovinos/virologia , Animais , Lentivirus/classificação , Infecções por Lentivirus/virologia , Filogenia , Sequências Repetidas Terminais
16.
Viruses ; 13(11)2021 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-34834951

RESUMO

Understanding the evolution of viral pathogens is critical to being able to define how viruses emerge within different landscapes. Host susceptibility, which is spread between different species and is a contributing factor to the subsequent epidemiology of a disease, is defined by virus detection and subsequent characterization. Peste des petits ruminants virus is a plague of small ruminant species that is a considerable burden to the development of sustainable agriculture across Africa and much of Asia. The virus has also had a significant impact on populations of endangered species in recent years, highlighting its significance as a pathogen of high concern across different regions of the globe. Here, we have re-evaluated the molecular evolution of this virus using novel genetic data to try and further resolve the molecular epidemiology of this disease. Viral isolates are genetically characterized into four lineages (I-IV), and the historic origin of these lineages is of considerable interest to the molecular evolution of the virus. Our re-evaluation of viral emergence using novel genome sequences has demonstrated that lineages I, II and IV likely originated in West Africa, in Senegal (I) and Nigeria (II and IV). Lineage III sequences predicted emergence in either East Africa (Ethiopia) or in the Arabian Peninsula (Oman and/or the United Arab Emirates), with a paucity of data precluding a more refined interpretation. Continual refinements of evolutionary emergence, following the generation of new data, is key to both understanding viral evolution from a historic perspective and informing on the ongoing genetic emergence of this virus.


Assuntos
Evolução Molecular , Genes Virais , Peste dos Pequenos Ruminantes/epidemiologia , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/classificação , Vírus da Peste dos Pequenos Ruminantes/genética , África Oriental/epidemiologia , África Ocidental/epidemiologia , Animais , Ásia/epidemiologia , Surtos de Doenças , Etiópia/epidemiologia , Genoma Viral , Doenças das Cabras/virologia , Cabras/virologia , Epidemiologia Molecular , Filogenia , Ruminantes/virologia , Senegal/epidemiologia , Análise de Sequência de DNA , Emirados Árabes Unidos/epidemiologia , Sequenciamento Completo do Genoma
17.
Emerg Microbes Infect ; 10(1): 2199-2201, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34749583

RESUMO

We report pilot studies to evaluate the susceptibility of common domestic livestock (cattle, sheep, goat, alpaca, rabbit, and horse) to intranasal infection with SARS-CoV-2. None of the infected animals shed infectious virus via nasal, oral, or faecal routes, although viral RNA was detected in several animals. Further, neutralizing antibody titres were low or non-existent one month following infection. These results suggest that domestic livestock are unlikely to contribute to SARS-CoV-2 epidemiology.


Assuntos
COVID-19/veterinária , Especificidade de Hospedeiro , Gado/virologia , SARS-CoV-2/patogenicidade , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , COVID-19/imunologia , COVID-19/virologia , Camelídeos Americanos/virologia , Bovinos/virologia , Chlorocebus aethiops , Reservatórios de Doenças/virologia , Cabras/virologia , Cavalos/virologia , Especificidade de Hospedeiro/imunologia , Humanos , Cavidade Nasal/virologia , RNA Viral/análise , Coelhos/virologia , Reto/virologia , Sistema Respiratório/virologia , SARS-CoV-2/genética , SARS-CoV-2/isolamento & purificação , Ovinos/virologia , Especificidade da Espécie , Células Vero , Eliminação de Partículas Virais , Vísceras/virologia
18.
Viruses ; 13(10)2021 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-34696389

RESUMO

South Tyrol has implemented, in 2007, a mandatory eradication program against Caprine Arthritis Encephalitis Virus (CAEV), a virus known to cause economic losses related to decreases in milk production and milk quality in goats, along with poor animal welfare and premature death. After a great initial decrease in the seroprevalence, the program has reached a tailing phase with scattered positivities. Potential risk factors associated with the multispecies farming system, a traditional approach in South Tyrol, are evaluated in this study, in order to better understand some of the potential causes leading to the tailing phenomenon. A statistically significant number of farms was selected for the present study, based on the risk factors evaluated. Even though there is no statistically significant association between the practices evaluated and the incidence of infection, the authors believe that it is important to highlight potential risks that may threaten the outcome of this eradication program.


Assuntos
Agricultura/normas , Vírus da Artrite-Encefalite Caprina/patogenicidade , Erradicação de Doenças/normas , Doenças das Cabras/epidemiologia , Infecções por Lentivirus/epidemiologia , Infecções por Lentivirus/veterinária , Agricultura/métodos , Animais , Erradicação de Doenças/métodos , Doenças das Cabras/etiologia , Cabras/virologia , Incidência , Itália/epidemiologia , Infecções por Lentivirus/etiologia , Fatores de Risco , Estudos Soroepidemiológicos
19.
Viruses ; 13(10)2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34696484

RESUMO

Small ruminant lentiviruses (SRLV) are economically important viral pathogens of sheep and goats. SRLV infection may interfere in the innate and adaptive immunity of the host, and genes associated with resistance or susceptibility to infection with SRLV have not been fully recognized. The presence of animals with relatively high and low proviral load suggests that some host factors are involved in the control of virus replication. To better understand the role of the genes involved in the host response to SRLV infection, RNA sequencing (RNA-seq) method was used to compare whole gene expression profiles in goats carrying both a high (HPL) and low (LPL) proviral load of SRLV and uninfected animals. Data enabled the identification of 1130 significant differentially expressed genes (DEGs) between control and LPL groups: 411 between control and HPL groups and 1434 DEGs between HPL and LPL groups. DEGs detected between the control group and groups with a proviral load were found to be significantly enriched in several gene ontology (GO) terms, including an integral component of membrane, extracellular region, response to growth factor, inflammatory and innate immune response, transmembrane signaling receptor activity, myeloid differentiation primary response gene 88 (MyD88)-dependent toll-like receptor signaling pathway as well as regulation of cytokine secretion. Our results also demonstrated significant deregulation of selected pathways in response to viral infection. The presence of SRLV proviral load in blood resulted in the modification of gene expression belonging to the toll-like receptor signaling pathway, the tumor necrosis factor (TNF) signaling pathway, the cytokine-cytokine receptor interaction, the phagosome, the Ras signaling pathway, the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) (PI3K-Akt) signaling pathway and rheumatoid arthritis. It is worth mentioning that the most predominant in all pathways were genes represented by toll-like receptors, tubulins, growth factors as well as interferon gamma receptors. DEGs detected between LPL and HPL groups were found to have significantly enriched regulation of signaling receptor activity, the response to toxic substances, nicotinamide adenine dinucleotide (NADH) dehydrogenase complex assembly, cytokine production, vesicle, and vacuole organization. In turn, the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway tool classified DEGs that enrich molecular processes such as B and T-cell receptor signaling pathways, natural killer cell-mediated cytotoxicity, Fc gamma R-mediated phagocytosis, toll-like receptor signaling pathways, TNF, mammalian target of rapamycin (mTOR) signaling and forkhead box O (Foxo) signaling pathways, etc. Our data indicate that changes in SRLV proviral load induced altered expression of genes related to different biological processes such as immune response, inflammation, cell locomotion, and cytokine production. These findings provide significant insights into defense mechanisms against SRLV infection. Furthermore, these data can be useful to develop strategies against SRLV infection by selection of animals with reduced SRLV proviral concentration that may lead to a reduction in the spread of the virus.


Assuntos
Vírus da Artrite-Encefalite Caprina/genética , Cabras/virologia , Vírus Visna-Maedi/genética , Imunidade Adaptativa/genética , Animais , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/genética , Doenças das Cabras/virologia , Cabras/genética , Interações entre Hospedeiro e Microrganismos/genética , Imunidade Inata/genética , Infecções por Lentivirus/veterinária , Lentivirus Ovinos-Caprinos/genética , Provírus/genética , Análise de Sequência de RNA , Transcriptoma/genética , Carga Viral/métodos , Replicação Viral
20.
Front Immunol ; 12: 745315, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34671358

RESUMO

Peste des petits ruminants (PPR) is an acute transboundary infectious viral disease of small ruminants, mainly sheep and goats. Host susceptibility varies considerably depending on the PPR virus (PPRV) strain, the host species and breed. The effect of strains with different levels of virulence on the modulation of the immune system has not been thoroughly compared in an experimental setting so far. In this study, we used a multi-omics approach to investigate the host cellular factors involved in different infection phenotypes. Peripheral blood mononuclear cells (PBMCs) from Saanen goats were activated with a T-cell mitogen and infected with PPRV strains of different virulence: Morocco 2008 (high virulence), Ivory Coast 1989 (low virulence) and Nigeria 75/1 (live attenuated vaccine strain). Our results showed that the highly virulent strain replicated better than the other two in PBMCs and rapidly induced cell death and a stronger inhibition of lymphocyte proliferation. However, all the strains affected lymphocyte proliferation and induced upregulation of key antiviral genes and proteins, meaning a classical antiviral response is orchestrated regardless of the virulence of the PPRV strain. On the other hand, the highly virulent strain induced stronger inflammatory responses and activated more genes related to lymphocyte migration and recruitment, and inflammatory processes. Both transcriptomic and proteomic approaches were successful in detecting viral and antiviral effectors under all conditions. The present work identified key immunological factors related to PPRV virulence in vitro.


Assuntos
Cabras/imunologia , Leucócitos Mononucleares/imunologia , Peste dos Pequenos Ruminantes/imunologia , Vírus da Peste dos Pequenos Ruminantes/patogenicidade , Virulência/imunologia , Animais , Perfilação da Expressão Gênica , Cabras/virologia , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/imunologia , Proteômica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA