Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mem Inst Oswaldo Cruz ; 117: e220177, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36651456

RESUMO

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections in domestic animals have occurred from the beginning of the pandemic to the present time. Therefore, from the perspective of One Health, investigating this topic is of global scientific and public interest. OBJECTIVES: The present study aimed to determine the presence of SARS-CoV-2 in domestic animals whose owners had coronavirus disease 2019 (COVID-19). METHODS: Nasopharyngeal and faecal samples were collected in Uruguay. Using quantitative polymerase chain reaction (qPCR), we analysed the presence of the SARS-CoV-2 genome. Complete genomes were obtained using ARTIC enrichment and Illumina sequencing. Sera samples were used for virus neutralisation assays. FINDINGS: SARS-CoV-2 was detected in an asymptomatic dog and a cat. Viral genomes were identical and belonged to the P.6 Uruguayan SARS-CoV-2 lineage. Only antiserum from the infected cat contained neutralising antibodies against the ancestral SARS-CoV-2 strain and showed cross-reactivity against the Delta but not against the B.A.1 Omicron variant. MAIN CONCLUSIONS: Domestic animals and the human SARS-CoV-2 P.6 variant comparison evidence a close relationship and gene flow between them. Different SARS-CoV-2 lineages infect dogs and cats, and no specific variants are adapted to domestic animals. This first record of SARS-CoV-2 in domestic animals from Uruguay supports regular surveillance of animals close to human hosts.


Assuntos
COVID-19 , Doenças do Gato , Doenças do Cão , Gatos , Animais , Humanos , Cães , SARS-CoV-2/genética , Uruguai , Doenças do Cão/diagnóstico , Doenças do Cão/epidemiologia , Animais Domésticos
2.
Arch Virol ; 167(12): 2457-2481, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36098802

RESUMO

Hepatitis E virus (HEV) is a leading cause of acute hepatitis worldwide. Hepatitis E is an enterically transmitted zoonotic disease that causes large waterborne epidemic outbreaks in developing countries and has become an increasing public-health concern in industrialized countries. In this setting, the infection is usually acute and self-limiting in immunocompetent individuals, although chronic cases in immunocompromised patients have been reported, frequently associated with several extrahepatic manifestations. Moreover, extrahepatic manifestations have also been reported in immunocompetent individuals with acute HEV infection. HEV belongs to the alphavirus-like supergroup III of single-stranded positive-sense RNA viruses, and its genome contains three partially overlapping open reading frames (ORFs). ORF1 encodes a nonstructural protein with eight domains, most of which have not been extensively characterized: methyltransferase, Y domain, papain-like cysteine protease, hypervariable region, proline-rich region, X domain, Hel domain, and RNA-dependent RNA polymerase. ORF2 and ORF3 encode the capsid protein and a multifunctional protein believed to be involved in virion release, respectively. The novel ORF4 is only expressed in HEV genotype 1 under endoplasmic reticulum stress conditions, and its exact function has not yet been elucidated. Despite important advances in recent years, the biological and molecular processes underlying HEV replication remain poorly understood, primarily due to a lack of detailed information about the functions of the viral proteins and the mechanisms involved in host-pathogen interactions. This review summarizes the current knowledge concerning HEV proteins and their biological properties, providing updated detailed data describing their function and focusing in detail on their structural characteristics. Furthermore, we review some unclear aspects of the four proteins encoded by the ORFs, highlighting the current key information gaps and discussing potential novel experimental strategies for shedding light on those issues.


Assuntos
Vírus da Hepatite E , Hepatite E , Animais , Humanos , Vírus da Hepatite E/genética , Fases de Leitura Aberta , Proteínas Virais/genética , Proteínas Virais/metabolismo , Zoonoses/epidemiologia
3.
J Med Virol ; 93(6): 4010-4014, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32592500

RESUMO

Hepatitis E virus (HEV) infection is considered a neglected disease of major concern in developed countries. Clinically, HEV occurs as an acute and self-limited disease, though chronic cases mostly associated to HEV-3 are now being commonly reported in immunocompromised individuals and solid organ transplant recipients. Transmission of HEV through blood and derivatives have been increasingly described in the last years, highlighting the importance of including this agent on the screening programs. Since 2010 both acute and chronic hepatitis E cases have been frequently reported in Uruguay. However, updated prevalence data among different population groups are lacking and HEV is not currently screened in blood banks. Herein, we report a seroprevalence and molecular survey of HEV in 400 plasma samples from blood donors. Overall, our results showed an HEV seroprevalence rate of 10% (40/400); almost 10-fold higher than 20 years ago. Total anti-HEV immunoglobulin antibodies were found to increase with age. Moreover, we reported an RNA detection rate of at least 0.75%, and two strains were sequenced. Phylogenetic analysis grouped them with human and swine HEV-3 strains from Uruguay. Data presented here should prompt public health policies of HEV screening in blood banks to minimize the risk of transfusion-transmitted hepatitis E.


Assuntos
Doadores de Sangue , Vírus da Hepatite E/genética , Vírus da Hepatite E/imunologia , Hepatite E/epidemiologia , Hepatite E/imunologia , Adolescente , Adulto , Fatores Etários , Doadores de Sangue/estatística & dados numéricos , Transfusão de Sangue , Anticorpos Anti-Hepatite/sangue , Hepatite E/sangue , Hepatite E/transmissão , Humanos , Pessoa de Meia-Idade , Filogenia , Prevalência , RNA Viral/sangue , RNA Viral/genética , Estudos Soroepidemiológicos , Transplantados/estatística & dados numéricos , Uruguai/epidemiologia , Adulto Jovem
4.
Virus Res ; 323: 198974, 2023 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-36272542

RESUMO

Torque Teno Virus (TTV) was initially associated with post-transfusion hepatitis, but growing evidence of its ubiquity in humans is compatible to no apparent clinical significance. TTV is a small non-enveloped virus with a circular single-negative-stranded DNA genome, belonging to the Anelloviridae family. Currently, TTVs are divided in seven phylogenetic groups and are further classified into 21 species. Studies about diversity of TTV in different conditions are receiving increasing interest and in this sense, sequencing of whole genomes for better genetic characterization becomes even more important. Since its discovery in 1997, few TTV complete genomes have been reported worldwide. This is probably due, among other reasons, to the great genetic heterogeneity among TTV strains that prevents its amplification and sequencing by conventional PCR and cloning methods. In addition, although metagenomics approach is useful in these cases, it remains a challenging tool for viromic analysis. With the aim of contributing to the expansion of the TTV whole genomes dataset and to study intra-host variants, we employed a methodology that combined a rolling-circle amplification approach followed by EcoRI digestion, generating a DNA fragment of ∼4Kb consistent with TTV genome length which was sequenced by Illumina next generation sequencing. A genogroup 3 full-length consensus TTV genome was obtained and co-infection with other species (at least those with a single EcoRI cleavage site) was not identified. Additionally, bioinformatics analysis allowed to identify the spectrum of TTV intra-host variants which provides evidence of a complex evolution dynamics of these DNA circular viruses, similarly to what occurs with RNA viruses.

5.
J Biomol Struct Dyn ; 41(2): 705-721, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-34861797

RESUMO

Hepatitis E Virus (HEV) infection is an emergent zoonotic disease, where chronic hepatitis E associated to solid organ transplant (SOT) recipients, related to genotype 3, is the clinical manifestation of major concern. In this setting, ribavirin (RBV) treatment is the only available therapy, though drug-resistant variants could emerge leading to a therapeutic failure. Crystallographic structures have not been reported for most of the HEV proteins, including the RNA-polymerase (RdRp). Therefore, the mechanism of action of RBV against HEV and the molecular interactions between this drug and RdRp are largely unknown. In this work, we aimed to model in silico the 3 D structure of a novel HEV3 RdRp (HEV_C1_Uy) from a chronically HEV infected-SOT recipient treated with RBV and to perform a molecular docking simulation between RBV triphosphate (RBVT), 7-methyl-guanosine-5'-triphosphate and the modelled protein. The models were generated using I-TASSER server and validated with multiple bioinformatics tools. The docking analysis were carried out with AutoDock Vina and LeDock software. We obtained a suitable model for HEV_C1_Uy (C-Score=-1.33, RMSD = 10.4 ± 4.6 Å). RBVT displayed a binding affinity of -7.6 ± 0.2 Kcal/mol by molecular docking, mediated by 6 hydrogen-bonds (Q195-O14, S198-O11, E257-O13, S260-O2, O3, S311-O11) between the finger's-palm-domains and a free binding energy of 31.26 ± 16.81 kcal/mol by molecular dynamics simulations. We identified the possible HEV RdRp interacting region for incoming nucleotides or analogs and provide novel insights that will contribute to better understand the molecular interactions of RBV and the enzyme and the mechanism of action of this antiviral drug.Communicated by Ramaswamy H. Sarma.


Assuntos
Vírus da Hepatite E , Hepatite E , Humanos , Ribavirina/farmacologia , Ribavirina/uso terapêutico , Vírus da Hepatite E/genética , Simulação de Acoplamento Molecular , RNA Polimerase Dependente de RNA/genética , Antivirais/farmacologia , Antivirais/uso terapêutico , Hepatite E/tratamento farmacológico , Genótipo
6.
Food Environ Virol ; 15(4): 281-291, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37642917

RESUMO

Hepatitis E caused by hepatitis E virus (HEV) is considered an emerging foodborne zoonosis in industrialized, non-endemic countries. Domestic pigs and wild boars are considered the main reservoir of HEV. However, HEV can also infect an ever-expanding host range of animals, but they exact role in transmitting the virus to other species or humans is mostly unknown. In this work, we investigated the spread of HEV in free-living and captive spotted deer (Axis axis) from Uruguay in a 2-year period (2020-2022) and examined the role of this invasive species as a new potential reservoir of the virus. In addition, with the aim to gain new insights into viral ecology in the context of One Health, by using camera trapping, we identified and quantified temporal and spatial coexistence of spotted deer, wild boars, and cattle. In free-living animals, we detected an anti-HEV seropositivity of 11.1% (6/54). HEV infection and viral excretion in feces were assessed by RT-PCR. Thirteen of 19 samples (68.4%) had HEV RNA. Six samples were amplified using a broadly reactive RT-PCR and sequenced. No captive animal showed evidence of HEV infection. Additionally, HEV RNA was detected in a freshwater pond shared by these species. Phylogenetic and p-distance analysis revealed that zoonotic HEV genotype 3 strains circulate together with unclassified variants related to moose HEV whose potential risk of transmission to humans and other domestic and wild animals is unknown. The data presented here suggest that spotted deer (A. axis) may be a novel host for zoonotic HEV strains.


Assuntos
Cervos , Vírus da Hepatite E , Hepatite E , Suínos , Humanos , Bovinos , Animais , Vírus da Hepatite E/genética , Filogenia , Uruguai , Sus scrofa , RNA Viral/genética , Genótipo
7.
PLoS One ; 18(4): e0284483, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37083889

RESUMO

SARS-CoV-2 surveillance of viral populations in wastewater samples is recognized as a useful tool for monitoring epidemic waves and boosting health preparedness. Next generation sequencing of viral RNA isolated from wastewater is a convenient and cost-effective strategy to understand the molecular epidemiology of SARS-CoV-2 and provide insights on the population dynamics of viral variants at the community level. However, in low- and middle-income countries, isolated groups have performed wastewater monitoring and data has not been extensively shared in the scientific community. Here we report the results of monitoring the co-circulation and abundance of variants of concern (VOCs) of SARS-CoV-2 in Uruguay, a small country in Latin America, between November 2020-July 2021 using wastewater surveillance. RNA isolated from wastewater was characterized by targeted sequencing of the Receptor Binding Domain region within the spike gene. Two computational approaches were used to track the viral variants. The results of the wastewater analysis showed the transition in the overall predominance of viral variants in wastewater from No-VOCs to successive VOCs, in agreement with clinical surveillance from sequencing of nasal swabs. The mutations K417T, E484K and N501Y, that characterize the Gamma VOC, were detected as early as December 2020, several weeks before the first clinical case was reported. Interestingly, a non-synonymous mutation described in the Delta VOC, L452R, was detected at a very low frequency since April 2021 when using a recently described sequence analysis tool (SAM Refiner). Wastewater NGS-based surveillance of SARS-CoV-2 is a reliable and complementary tool for monitoring the introduction and prevalence of VOCs at a community level allowing early public health decisions. This approach allows the tracking of symptomatic and asymptomatic individuals, who are generally under-reported in countries with limited clinical testing capacity. Our results suggests that wastewater-based epidemiology can contribute to improving public health responses in low- and middle-income countries.


Assuntos
COVID-19 , Águas Residuárias , Humanos , SARS-CoV-2/genética , Vigilância Epidemiológica Baseada em Águas Residuárias , COVID-19/epidemiologia , Genômica , Sequenciamento de Nucleotídeos em Larga Escala
8.
Viruses ; 15(10)2023 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-37896784

RESUMO

Hepatitis E Virus (HEV) infection is an emergent zoonotic disease of increasing concern in developed regions. HEV genotype 3 (HEV-3) is mainly transmitted through consumption of contaminated food in high-income countries and is classified into at least 13 subtypes (3a-3n), based on p-distance values from complete genomes. In Latin America, HEV epidemiology studies are very scant. Our group has previously detected HEV3 in clinical cases, swine, wild boars, captive white-collared peccaries, and spotted deer from Uruguay. Herein, we aimed to provide novel insights and an updated overview of the molecular epidemiology of zoonotic HEV in Uruguay, including data from wastewater-based surveillance studies. A thorough analysis of HEV whole genomes and partial ORF2 sequences from Uruguayan human and domestic pig strains showed that they formed a separate monophyletic cluster with high nucleotide identity and exhibited p-distance values over the established cut-off (0.093) compared with reference subtypes' sequences. Furthermore, we found an overall prevalence of 10.87% (10/92) in wastewater, where two samples revealed a close relationship with humans, and animal reservoirs/hosts isolates from Uruguay. In conclusion, a single, new HEV-3 subtype currently circulates in different epidemiological settings in Uruguay, and we propose its designation as 3o along with its reference sequence.


Assuntos
Cervos , Vírus da Hepatite E , Hepatite E , Doenças dos Suínos , Suínos , Animais , Humanos , Vírus da Hepatite E/genética , Hepatite E/epidemiologia , Hepatite E/veterinária , Uruguai/epidemiologia , Filogenia , Genótipo , Cervos/genética , Sus scrofa/genética , Monitoramento Ambiental , RNA Viral/genética
9.
Microbiol Resour Announc ; 10(22): e0036721, 2021 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-34080907

RESUMO

Hepatitis E virus (HEV) is a leading cause of acute viral hepatitis worldwide. We report the full-length genome sequence of an HEV-3 strain obtained from a chronically infected patient from Uruguay. This strain shared only 86% nucleotide sequence identity with the most closely related reference strain belonging to subtype 3m.

10.
Transbound Emerg Dis ; 68(3): 1040-1045, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-32799421

RESUMO

Hepatitis E virus (HEV) infection is a major cause of acute hepatitis worldwide. Clinical presentation of hepatitis E mainly occurs as an acute and self-limited disease, though chronic cases are now being commonly reported in immunocompromised individuals. In high-income developed areas and non-endemic regions, HEV is mainly transmitted by the zoonotic route through direct contact with infected animals or by consumption of contaminated meat products. Although pigs and wild boars are the main reservoirs of the disease, HEV can also infect deer, camels, and rats and seems to have an ever-expanding host range. Peccaries (Tayassuidae family, superfamily Suoidea), the 'new world pigs', share susceptibility to several pathogens with domestic pigs and wild boars. Herein, we performed a serological and molecular survey of two captive populations of white-collared peccaries (Pecari tajacu) from Uruguay, with the aim to assess the role of the species as an HEV reservoir. One-hundred and one serum samples were analysed for anti-HEV antibodies. Further evidences of active HEV infection were investigated in stool by RT-nested PCR. Animals from both wildlife reserves were exposed to HEV with an overall prevalence of 24.7%. Moreover, HEV RNA could be detected in peccaries' stool samples from one of the reserves. Phylogenetic analysis clustered the strains within HEV-3, closely related to both human and swine isolates. Our work provides the first evidences supporting the notion that white-collared peccaries are susceptible to HEV. However, these data should not be overinterpreted. Further research is needed concerning the role of peccaries in the transmission of HEV.


Assuntos
Artiodáctilos , Vírus da Hepatite E/isolamento & purificação , Hepatite E/veterinária , Animais , Animais de Zoológico , Genótipo , Anticorpos Anti-Hepatite/análise , Hepatite E/epidemiologia , Hepatite E/virologia , Vírus da Hepatite E/classificação , Filogenia , Prevalência , Estudos Soroepidemiológicos , Uruguai/epidemiologia
11.
Mem. Inst. Oswaldo Cruz ; 117: e220177, 2022. tab, graf
Artigo em Inglês | LILACS-Express | LILACS | ID: biblio-1422138

RESUMO

BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections in domestic animals have occurred from the beginning of the pandemic to the present time. Therefore, from the perspective of One Health, investigating this topic is of global scientific and public interest. OBJECTIVES The present study aimed to determine the presence of SARS-CoV-2 in domestic animals whose owners had coronavirus disease 2019 (COVID-19). METHODS Nasopharyngeal and faecal samples were collected in Uruguay. Using quantitative polymerase chain reaction (qPCR), we analysed the presence of the SARS-CoV-2 genome. Complete genomes were obtained using ARTIC enrichment and Illumina sequencing. Sera samples were used for virus neutralisation assays. FINDINGS SARS-CoV-2 was detected in an asymptomatic dog and a cat. Viral genomes were identical and belonged to the P.6 Uruguayan SARS-CoV-2 lineage. Only antiserum from the infected cat contained neutralising antibodies against the ancestral SARS-CoV-2 strain and showed cross-reactivity against the Delta but not against the B.A.1 Omicron variant. MAIN CONCLUSIONS Domestic animals and the human SARS-CoV-2 P.6 variant comparison evidence a close relationship and gene flow between them. Different SARS-CoV-2 lineages infect dogs and cats, and no specific variants are adapted to domestic animals. This first record of SARS-CoV-2 in domestic animals from Uruguay supports regular surveillance of animals close to human hosts.

12.
Infect Genet Evol ; 44: 501-506, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27510954

RESUMO

Torque Teno Virus (TTV), member of Anelloviridae family, is considered a worldwide distributed emergent virus and is currently classified into seven genogroups. Interestingly, the pathogenicity of TTV remains unclear. However, it has been constantly associated to hepatitis cases of unknown etiology (HUE) as well as extensively studied in concurrent infections with Hepatitis B Virus (HBV), Hepatitis C Virus (HCV) and Human Immunodeficiency Virus type 1 (HIV-1). In South America, TTV epidemiological data is scant, involving some studies from Brazil, Venezuela, Colombia and Bolivia. The aim of this work was to investigate for the first time in Uruguay the presence of TTV by a nested-PCR system in 85 human serum samples infected with HBV and/or HCV and/or HIV-1 and in HUE cases. Overall, our results reported a TTV infection rate of 79% (67/85). Furthermore, the molecular characterization of Uruguayan strains revealed that one of them clustered in genogroup 1, while the remaining ones formed separate clusters closely related to genogroup 3, which should be confirmed by complete genome sequencing. Further investigation about TTV circulation in Uruguayan population is needed in order to provide additional information about the genetic variability and TTV epidemiology in South America.


Assuntos
Infecções por Vírus de DNA/epidemiologia , Infecções por Vírus de DNA/virologia , Torque teno virus/genética , Genótipo , Humanos , Filogenia , Análise de Sequência de DNA , Torque teno virus/classificação , Uruguai/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA