Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33.359
Filtrar
1.
Vopr Virusol ; 69(4): 329-340, 2024 Sep 26.
Artigo em Russo | MEDLINE | ID: mdl-39361927

RESUMO

INTRODUCTION: For four years, SARS-CoV-2, the etiological agent of COVID-19, has been circulating among humans. By the end of the second year, an absence of immunologically naive individuals was observed, attributable to extensive immunization efforts and natural viral exposure. This study focuses on delineating the molecular and biological patterns that facilitate the persistence of SARS-CoV-2, thereby informing predictions on the epidemiological trajectory of COVID-19 toward refining pandemic countermeasures. The aim of this study was to describe the molecular biological patterns identified that contribute to the persistence of the virus in the human population. MATERIALS AND METHODS: For over three years since the beginning of the COVID-19 pandemic, molecular genetic monitoring of SARS-CoV-2 has been conducted, which included the collection of nasopharyngeal swabs from infected individuals, assessment of viral load, and subsequent whole-genome sequencing. RESULTS: We discerned dominant genetic lineages correlated with rising disease incidence. We scrutinized amino acid substitutions across SARS-CoV-2 proteins and quantified viral loads in swab samples from patients with emerging COVID-19 variants. Our findings suggest a model of viral persistence characterized by 1) periodic serotype shifts causing substantial diminutions in serum virus-neutralizing activity (> 10-fold), 2) serotype-specific accrual of point mutations in the receptor-binding domain (RBD) to modestly circumvent neutralizing antibodies and enhance receptor affinity, and 3) a gradually increasing amount of virus being shed in mucosal surfaces within a single serotype. CONCLUSION: This model aptly accounts for the dynamics of COVID-19 incidence in Moscow. For a comprehensive understanding of these dynamics, acquiring population-level data on immune tension and antibody neutralization relative to genetic lineage compositions is essential.


Assuntos
COVID-19 , SARS-CoV-2 , Carga Viral , Humanos , SARS-CoV-2/genética , SARS-CoV-2/imunologia , SARS-CoV-2/isolamento & purificação , COVID-19/epidemiologia , COVID-19/virologia , COVID-19/imunologia , Genoma Viral/genética , Substituição de Aminoácidos , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Pandemias , Filogenia , Nasofaringe/virologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Feminino , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Masculino
2.
Vopr Virusol ; 69(4): 363-376, 2024 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-39361930

RESUMO

INTRODUCTION: Rotavirus infection is the major cause of severe dehydrating diarrhea requiring hospitalization in young children worldwide. Due to their segmented genome, rotaviruses are capable of gene reassortment, which makes the emergence and spread of genetically novel strains possible. The purpose of this study was to search for unusual rotaviruses circulating in Nizhny Novgorod in 2021‒2023 and their molecular genetic characterization based on all genome segments. MATERIALS AND METHODS: Rotavirus-positive stool samples of children were examined by PCR genotyping and electrophoresis in PAAG. cDNA fragments of each of the 11 genes (VP1‒VP4, VP6, VP7, NSP1‒NSP5), 570 to 850 nucleotide pairs in length were sequenced for the selected strains. The phylogenetic analysis was performed in the MEGA X program. RESULTS: In the study period 2021‒2023, 11 G[P] combinations with a predominance of G3P[8] (59.5%) were identified. Six atypical Rotavirus А (RVA) strains were identified: 2 strains of the G2P[4] genotype (G2-P[4]-I2-R2-C2-M2-A3-N2-T3-E2-H3, G2-P[4]-I2-R2-C2-M2-A3-N2-T3-E3-H2) and 4 G3P[9] strains (all strains had the genotype G3-P[9]-I2-R2-C2-M2-A3-N2-T3-E3-H3). Phylogenetic analysis based on all genes showed an evolutionary relationship between rotaviruses similar to rotaviruses of cats and dogs (BA222-like) and unusual strains of the G2P[4] genotype, for which a mixed combination of genotypes was identified and characterized for the first time. DISCUSSION: The results obtained expand the understanding of the diversity of reassortant RVAs, as well as complement the data on the genotypic structure of the rotavirus population in Nizhny Novgorod. CONCLUSION: The wide genetic diversity of reassortant RVA can help rotaviruses overcome the immunological pressure provided by natural and vaccine-induced immunity. In this regard, to control the emergence of new variants and assess changes in the virulence of rotaviruses after reassortment processes, continuous molecular monitoring for circulating RVA is necessary.


Assuntos
Genoma Viral , Genótipo , Filogenia , Infecções por Rotavirus , Rotavirus , Rotavirus/genética , Rotavirus/classificação , Rotavirus/isolamento & purificação , Humanos , Infecções por Rotavirus/virologia , Pré-Escolar , Lactente , Masculino , Fezes/virologia , Feminino , Diarreia/virologia , Criança
3.
Vopr Virusol ; 69(4): 377-389, 2024 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-39361931

RESUMO

RELEVANCE: Influenza A virus is characterized by a segmented single-stranded RNA genome. Such organization of the virus genome determines the possibility of reassortment, which can lead to the emergence of new virus variants. The main natural reservoir of most influenza A virus subtypes are wild waterfowl. Seasonal migrations gather waterfowl from all major migration routes to nesting areas near the northern and southern polar circles. This makes intercontinental spread of influenza A viruses possible. Objective ‒ to conduct molecular genetic monitoring and study the phylogenetic relationships of influenza A virus variants circulating in Antarctica in 2023. MATERIALS AND METHODS: We studied 84 samples of biological material obtained from birds and marine mammals in April‒May 2023 in coastal areas of Antarctica. For 3 samples, sequencing was performed on the Miseq, Illumina platform and phylogenetic analysis of the obtained nucleotide sequences of the influenza A virus genomes was performed. RESULTS: The circulation of avian influenza virus in the Antarctic region was confirmed. Heterogeneity of the pool of circulating variants of the influenza A virus (H3N8, H1N1) was revealed. Full-length genomes of the avian influenza virus were sequenced and posted in the GISAID database (EPI_ISL_19032103, 19174530, 19174467). CONCLUSION: The study of the genetic diversity of influenza A viruses circulating in the polar regions of the Earth and the identification of the conditions for the emergence of new genetic variants is a relevant task for the development of measures to prevent biological threats.


Assuntos
Aves , Genoma Viral , Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A Subtipo H3N8 , Influenza Aviária , Filogenia , Regiões Antárticas , Animais , Aves/virologia , Influenza Aviária/virologia , Influenza Aviária/epidemiologia , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Vírus da Influenza A Subtipo H1N1/classificação , Vírus da Influenza A Subtipo H3N8/genética , Vírus da Influenza A Subtipo H3N8/isolamento & purificação , Vírus da Influenza A Subtipo H3N8/classificação , Humanos , Influenza Humana/virologia , Influenza Humana/epidemiologia , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Orthomyxoviridae/veterinária
4.
Nat Commun ; 15(1): 8482, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39353939

RESUMO

DEV is an obligatory lytic Pseudomonas phage of the N4-like genus, recently reclassified as Schitoviridae. The DEV genome encodes 91 ORFs, including a 3398 amino acid virion-associated RNA polymerase (vRNAP). Here, we describe the complete architecture of DEV, determined using a combination of cryo-electron microscopy localized reconstruction, biochemical methods, and genetic knockouts. We built de novo structures of all capsid factors and tail components involved in host attachment. We demonstrate that DEV long tail fibers are essential for infection of Pseudomonas aeruginosa but dispensable for infecting mutants with a truncated lipopolysaccharide devoid of the O-antigen. We determine that DEV vRNAP is part of a three-gene operon conserved in 191 Schitoviridae genomes. We propose these three proteins are ejected into the host to form a genome ejection motor spanning the cell envelope. We posit that the design principles of the DEV ejection apparatus are conserved in all Schitoviridae.


Assuntos
Microscopia Crioeletrônica , Genoma Viral , Fagos de Pseudomonas , Pseudomonas aeruginosa , Fagos de Pseudomonas/genética , Fagos de Pseudomonas/ultraestrutura , Genoma Viral/genética , Pseudomonas aeruginosa/virologia , Pseudomonas aeruginosa/genética , RNA Polimerases Dirigidas por DNA/metabolismo , RNA Polimerases Dirigidas por DNA/genética , Vírion/ultraestrutura , Vírion/genética , Fases de Leitura Aberta/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Virais/química , Óperon/genética , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/química , Capsídeo/metabolismo , Capsídeo/ultraestrutura
5.
Microbiome ; 12(1): 187, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39354646

RESUMO

BACKGROUND: Metagenomics is a powerful approach to study environmental and human-associated microbial communities and, in particular, the role of viruses in shaping them. Viral genomes are challenging to assemble from metagenomic samples due to their genomic diversity caused by high mutation rates. In the standard de Bruijn graph assemblers, this genomic diversity leads to complex k-mer assembly graphs with a plethora of loops and bulges that are challenging to resolve into strains or haplotypes because variants more than the k-mer size apart cannot be phased. In contrast, overlap assemblers can phase variants as long as they are covered by a single read. RESULTS: Here, we present PenguiN, a software for strain resolved assembly of viral DNA and RNA genomes and bacterial 16S rRNA from shotgun metagenomics. Its exhaustive detection of all read overlaps in linear time combined with a Bayesian model to select strain-resolved extensions allow it to assemble longer viral contigs, less fragmented genomes, and more strains than existing assembly tools, on both real and simulated datasets. We show a 3-40-fold increase in complete viral genomes and a 6-fold increase in bacterial 16S rRNA genes. CONCLUSION: PenguiN is the first overlap-based assembler for viral genome and 16S rRNA assembly from large and complex metagenomic datasets, which we hope will facilitate studying the key roles of viruses in microbial communities. Video Abstract.


Assuntos
Bactérias , Genoma Viral , Metagenômica , RNA Ribossômico 16S , RNA Ribossômico 16S/genética , Genoma Viral/genética , Metagenômica/métodos , Bactérias/genética , Bactérias/classificação , Bactérias/virologia , Software , Humanos , Teorema de Bayes , Vírus/genética , Vírus/classificação , Metagenoma
6.
Nat Commun ; 15(1): 8528, 2024 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-39358425

RESUMO

Usutu virus (USUV) and West Nile virus (WNV) are two closely related emerging mosquito-borne flaviviruses. Their natural hosts are wild birds, but they can also cause severe neurological disorders in humans. Both viruses are efficiently suppressed by type I interferon (IFN), which interferes with viral replication, dissemination, pathogenesis and transmission. Here, we show that the replication of USUV and WNV are inhibited through a common set of IFN-induced genes (ISGs), with the notable exception of ISG20, which USUV is resistant to. Strikingly, USUV was the only virus among all the other tested mosquito-borne flaviviruses that demonstrated resistance to the 3'-5' exonuclease activity of ISG20. Our findings highlight that the intrinsic resistance of the USUV genome, irrespective of the presence of cellular or viral proteins or protective post-transcriptional modifications, relies on a unique sequence present in its 3' untranslated region. Importantly, this genomic region alone can confer ISG20 resistance to a susceptible flavivirus, without compromising its infectivity, suggesting that it could be acquired by other flaviviruses. This study provides new insights into the strategy employed by emerging flaviviruses to overcome host defense mechanisms.


Assuntos
Regiões 3' não Traduzidas , Flavivirus , Replicação Viral , Vírus do Nilo Ocidental , Regiões 3' não Traduzidas/genética , Flavivirus/genética , Flavivirus/fisiologia , Humanos , Animais , Replicação Viral/genética , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/fisiologia , Infecções por Flavivirus/virologia , Exonucleases/metabolismo , Exonucleases/genética , Chlorocebus aethiops , Exorribonucleases/metabolismo , Exorribonucleases/genética , Células HEK293 , Células Vero , Linhagem Celular , Interferon Tipo I/metabolismo , Genoma Viral
7.
Virol J ; 21(1): 242, 2024 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-39358760

RESUMO

Rotavirus group C is an important cause of sporadic cases and outbreaks of gastroenteritis worldwide. Whole-Genome sequences of human rotavirus C (RVC) in public databases are limited. We performed genome sequencing to analyze a RVC outbreak of acute gastroenteritis in China. Samples from 22 patients were screened for pathogens using RT-PCR, and six samples were positive for rotavirus. Whole-Genome sequencing analysis showed that the outbreak strain SJZ217 belongs to the G4-P[2]-I2-R2-C2-M3-A2-N2-T2-E2-H2 genotype and shares almost identical genomic sequences with Chungnam isolated in Korea. Phylogenetic analysis revealed strain SJZ217 also fell into a cluster with rotavirus C strains from Japan and Europe. Reassortment in the VP4 fragment was observed. These results helped to understand the genetic diversity and possible spread of RVC strains.


Assuntos
Surtos de Doenças , Gastroenterite , Genoma Viral , Genótipo , Filogenia , Infecções por Rotavirus , Rotavirus , Humanos , Gastroenterite/virologia , Gastroenterite/epidemiologia , Infecções por Rotavirus/virologia , Infecções por Rotavirus/epidemiologia , China/epidemiologia , Rotavirus/genética , Rotavirus/classificação , Rotavirus/isolamento & purificação , Masculino , Pré-Escolar , RNA Viral/genética , Sequenciamento Completo do Genoma , Feminino , Análise de Sequência de DNA , Lactente , Variação Genética , Vírus Reordenados/genética , Vírus Reordenados/classificação , Vírus Reordenados/isolamento & purificação , Análise por Conglomerados
8.
Arch Virol ; 169(10): 212, 2024 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-39365443

RESUMO

A putative novel virus was identified in Agastache rugosa in China and tentatively named "Agastache rugosa-associated varicosavirus" (ARaVV). The nearly complete genome sequence of ARaVV was obtained through RNA sequencing (RNA-seq) and subsequent RT-PCR. The ARaVV genome consists of two negative-sense single-stranded RNA segments that are 6428 and 3862 nucleotides (nt) in size, respectively. RNA1 encodes a large polymerase (L), and RNA2 encodes a putative nucleocapsid protein (N), protein 2 (P2), and protein 3 (P3). The L protein shared the highest amino acid (aa) sequence identity (51.3%) with Erysimum virus 1 (EryV1, BK061766.1). The N, P2, and P3 shared the highest aa sequence identity (33.1%, 14.0%, and 24.2%) with Leucanthemum virus 1, Raphanus virus 1, and Spinach virus 1, respectively. Phylogenetic analysis based on amino acid sequences of the L protein showed that ARaVV clustered in a clade with the varicosaviruses, indicating that ARaVV is a putative new member of the genus Varicosavirus.


Assuntos
Genoma Viral , Filogenia , Doenças das Plantas , RNA Viral , Proteínas Virais , China , Genoma Viral/genética , Doenças das Plantas/virologia , Proteínas Virais/genética , RNA Viral/genética , Sequência de Aminoácidos , Closteroviridae/genética , Closteroviridae/isolamento & purificação , Closteroviridae/classificação
9.
Front Cell Infect Microbiol ; 14: 1470808, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39359936

RESUMO

A novel goose astrovirus (GAstV) has broken out across China in recent years, causing widespread damage to the poultry industry. In goslings infected with GAstV, the leading cause of death is visceral gout. However, our understanding of the mechanism of gout formation in GAstV infection is largely inadequate. The aim of this study was to examine the pathogenicity of a GAstV strain and explore the molecular mechanisms of visceral gout caused by viral infection in goslings. The virulent GAstV strain HR2105/1 was effectively isolated from the visceral tissue of goslings in gout-affected areas. The whole genome of the HR2105/1 strain was sequenced and analyzed. Subsequently, we established a gosling gout models with experimental GAstV infection. Finally, we conducted a study on the mechanism of GAstV induced acute kidney injury. Phylogenetic analysis of the complete genome sequence showed that it was closely related to the strain circulating in China since 2016, and it was grouped within the GAstV-1 cluster. The clinical signs were reproduced by experimental infection of healthy goslings with the isolated strain and were found to be similar to those reported in clinical cases. Moreover, the virus exhibits strong renal tropism. Infection with the GAstV strain HR2105/1 was found to cause acute kidney injury, as evidenced by increased levels of uric acid and creatinine as well as severe pathological damage. Mechanistic experiments with Masson and Picrosirius Red staining revealed fibrosis in renal tissues after GAstV infection. Furthermore, TUNEL staining revealed that GAstV infection triggered renal cell apoptosis. Additionally, RT-qPCR revealed that GAstV infection caused an excessive inflammatory response by upregulating the expression of IL-1ß, IL-6, IL-10, TGF-ß, and iNOS in renal tissues. Overall, our findings demonstrate that GAstV infection causes renal damage by inducing renal cell apoptosis, fibrosis, and excessive inflammatory response, which subsequently leads to hyperuricemia and lethal visceral gout formation. This is the first systematic study on the etiology of lethal gout in goslings caused by GAstV infection, and we believe that the findings can guide vaccine development and therapeutic targets for GAstV-associated renal diseases.


Assuntos
Injúria Renal Aguda , Infecções por Astroviridae , Gansos , Gota , Filogenia , Doenças das Aves Domésticas , Animais , Gansos/virologia , Gota/virologia , Gota/patologia , Infecções por Astroviridae/veterinária , Infecções por Astroviridae/virologia , Infecções por Astroviridae/patologia , Injúria Renal Aguda/virologia , Injúria Renal Aguda/patologia , China , Doenças das Aves Domésticas/virologia , Doenças das Aves Domésticas/patologia , Rim/patologia , Rim/virologia , Genoma Viral , Avastrovirus/genética , Avastrovirus/isolamento & purificação , Avastrovirus/patogenicidade , Sequenciamento Completo do Genoma , Modelos Animais de Doenças , Astroviridae/genética , Astroviridae/isolamento & purificação , População do Leste Asiático
10.
J Med Virol ; 96(10): e29944, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39360646

RESUMO

Influenza circulation was significantly affected in 2020-21 by the COVID-19 pandemic. During this time, few influenza cases were recorded. However, in the summer of 2021-22, an increase in atypical influenza cases was observed, leading to the resurgence of influenza in the southernmost state of Brazil, Rio Grande do Sul (RS). The present study aimed to identify the circulation of FLUAV, FLUBV and SARS-CoV-2 and characterize the influenza genomes in respiratory samples using high-throughput sequencing technology (HTS). Respiratory samples (n = 694) from patients in RS were selected between July 2021 and August 2022. The samples were typed using reverse transcriptase real-time PCR (RT-qPCR) and showed 32% (223/694) of the samples to be positive for SARS-CoV-2, 7% for FLUAV (H3) (49/694). FLUBV was not detected. RT-qPCR data also resulted in FLUAV and SARS-CoV-2 co-infections in 1.7% (4/223) of samples tested. Whole genome sequencing of FLUAV produced 15 complete genomes of the H3N2 subtype, phylogenetically classified in the 3C.2a1b.2a.2a.3 subclade and revealing the dominance of viruses in the southern region of Brazil. Mutation analysis identified 72 amino acid substitutions in all genes, highlighting ongoing genetic evolution with potential implications for vaccine effectiveness, viral fitness, and pathogenicity. This study underscores limitations in current surveillance systems, advocating for comprehensive data inclusion to enhance understanding of influenza epidemiology in southern Brazil. These findings contribute valuable insights to inform more effective public health responses and underscore the critical need for continuous genomic surveillance.


Assuntos
COVID-19 , Genoma Viral , Influenza Humana , Filogenia , SARS-CoV-2 , Humanos , Brasil/epidemiologia , COVID-19/epidemiologia , COVID-19/virologia , SARS-CoV-2/genética , SARS-CoV-2/classificação , SARS-CoV-2/isolamento & purificação , Influenza Humana/epidemiologia , Influenza Humana/virologia , Pessoa de Meia-Idade , Adulto , Feminino , Genoma Viral/genética , Masculino , Adulto Jovem , Idoso , Adolescente , Surtos de Doenças , Sequenciamento Completo do Genoma , Criança , Pré-Escolar , Lactente , Coinfecção/epidemiologia , Coinfecção/virologia , Sequenciamento de Nucleotídeos em Larga Escala , Idoso de 80 Anos ou mais , Genômica
11.
Vet Q ; 44(1): 1-13, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39350725

RESUMO

Canine parvovirus type 2 (CPV-2) infection in dogs is considered as one of the most common cause of morbidity and mortality in young dogs and continues to occur with high incidence worldwide. Despite a single-stranded DNA virus, CPV-2 possesses a high mutation rate which has led to the development of new variants from time to time. These variants are classically classified based on the amino acid markers present in the VP2 gene. In this study, we examined 20 different cases of CPV-2 infection from seven different states of the North East region (NER) of India. The near-complete genome sequences of all these isolates were subjected to phylodynamic and phylogeographic analysis to evaluate the genetic diversity and geographical spread of CPV-2 variants. Analysis of the deduced amino acid sequences revealed residues characteristic of the 'Asian CPV-2c lineage' in all the 20 sequences confirming it as the dominant strain circulating in NER, India. The phylogenetic analysis based on the whole genome showed that all 20 sequences formed a monophyletic clade together with other Asian CPV-2c sequences. Furthermore, phylogeographic analysis based on the VP2 gene showed the likely introduction of Asian CPV-2c strain to India from China. This study marks the first comprehensive report elucidating the molecular epidemiology of CPV-2 in India.


Assuntos
Doenças do Cão , Infecções por Parvoviridae , Parvovirus Canino , Filogenia , Parvovirus Canino/genética , Parvovirus Canino/classificação , Índia/epidemiologia , Cães , Infecções por Parvoviridae/veterinária , Infecções por Parvoviridae/virologia , Infecções por Parvoviridae/epidemiologia , Doenças do Cão/virologia , Doenças do Cão/epidemiologia , Animais , Filogeografia , Variação Genética , Genoma Viral , Evolução Molecular
12.
Mol Biol (Mosk) ; 58(2): 282-294, 2024.
Artigo em Russo | MEDLINE | ID: mdl-39355885

RESUMO

The tick-borne encephalitis virus (TBEV) strain C11-13 (GenBank acc. no. OQ565596) of the Siberian genotype was previously isolated from the brain of a deceased person. TBEV C11-13 variants obtained at passages 3 and 8 in SPEV cells were inoculated into the brains of white mice for subsequent passages. Full genome sequences of all virus variants were analyzed by high-throughput sequencing. A total of 41 single nucleotide substitutions were found to occur mainly in the genes for the nonstructural proteins NS3 and NS5 (GenBank MF043953, OP902894, and OP902895), and 12 amino acid substitutions were identified in the deduced protein sequences. Reverse nucleotide and amino acid substitutions were detected after three passages through mouse brains. The substitutions restored the primary structures that were characteristic of the isolate C11-13 from a human patient and changed during the eight subsequent passages in SPEV cells. In addition, the 3'-untranslated region (3'-UTR) of the viral genome increased by 306 nt. The Y3 and Y2 3'-UTR elements were found to contain imperfect L and R repeats, which were probably associated with inhibition of cellular XRN1 RNase and thus involved in the formation of subgenomic flaviviral RNAs (sfRNAs). All TBEV variants showed high-level reproduction in both cell cultures and mouse brains. The genomic changes that occurred during successive passages of TBEV are most likely due to its significant genetic variability, which ensures its efficient reproduction in various hosts and its broad distribution in various climatic zones.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos , Genoma Viral , Proteínas não Estruturais Virais , Vírus da Encefalite Transmitidos por Carrapatos/genética , Animais , Camundongos , Humanos , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Regiões 3' não Traduzidas/genética , Encefalite Transmitida por Carrapatos/virologia , Encefalite Transmitida por Carrapatos/genética , Substituição de Aminoácidos , Cultura de Vírus/métodos , Encéfalo/virologia , Encéfalo/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Linhagem Celular , Proteases Virais , Nucleosídeo-Trifosfatase , RNA Helicases DEAD-box
13.
Euro Surveill ; 29(38)2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39301745

RESUMO

Between January and August 2024, mpox cases have been reported in nearly all provinces of the Democratic Republic of the Congo (DRC). Monkeypox virus genome sequences were obtained from 11 mpox cases' samples, collected in July-August 2024 in several health zones of Kinshasa. Characterisation of the sequences showed subclades Ia and Ib co-circulating in the Limete health zone, while phylogenetic analyses suggested multiple introductions of the two subclades in Kinshasa. This illustrates the growing complexity of Clade I mpox outbreaks in DRC.


Assuntos
Surtos de Doenças , Monkeypox virus , Mpox , Filogenia , República Democrática do Congo/epidemiologia , Mpox/epidemiologia , Mpox/virologia , Humanos , Monkeypox virus/genética , Monkeypox virus/isolamento & purificação , Genoma Viral , RNA Viral/genética , Masculino , Análise de Sequência de DNA
14.
Arch Virol ; 169(10): 206, 2024 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-39305367

RESUMO

In this study, we completely sequenced the genome of a new member of the genus Alphaendornavirus, family Endornaviridae, from lima bean (Phaseolus lunatus), for which we propose the name "lima bean endornavirus 1" (LbEV1). The complete genome of LbEV1 consists of 15,265 nucleotides, including a stretch of 12 cytosine residues at its 3' end, and contains a long single open reading frame (ORF) coding for a 4980-aa-long polyprotein. Analysis of the polyprotein sequence revealed the presence of four conserved functional domains (in order from the N- to C-terminus): viral helicase 1, peptidase _C97, glycosyltransferase_GTB-type, and viral RNA-dependent RNA polymerase (RdRP). The LbEV1 polyprotein showed the highest amino acid sequence similarity (63% identity and 98% coverage) to Phaseolus vulgaris endornavirus 3 (PvEV3) and also showed 42% identity (95% coverage) to Geranium carolinianum endornavirus. Phylogenetic analysis based on the viral RdRp domain showed that LbEV1 belongs to a subclade within the genus Alphaendornavirus that includes three other viruses infecting plants of the genus Phaseolus.


Assuntos
Genoma Viral , Fases de Leitura Aberta , Phaseolus , Filogenia , Vírus de RNA , RNA Viral , Genoma Viral/genética , Phaseolus/virologia , Vírus de RNA/genética , Vírus de RNA/classificação , Vírus de RNA/isolamento & purificação , RNA Viral/genética , Proteínas Virais/genética , Sequenciamento Completo do Genoma/métodos , Sequência de Aminoácidos , Poliproteínas/genética , RNA Polimerase Dependente de RNA/genética , Doenças das Plantas/virologia , Sequência de Bases
15.
J Med Virol ; 96(9): e29918, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39311394

RESUMO

Patients with Primary immunodeficiency (PIDs) may be infected by Polioviruses (PVs), especially when vaccinated with live Oral Polio Vaccine before diagnosis. They may establish long-term shedding of divergent strains and may act as reservoirs of PV transmission. This study delved into the effect of the genetic evolution of complete PV genomes, from MHC class II-deficient patients, on the excretion duration and clinical outcomes. Stool samples from three PID patients underwent analysis for PV detection through inoculation on cell culture and real-time PCR, followed by VP1 partial sequencing and full genome sequencing using the Illumina technology. Our findings revealed a low number of mutations for one patient who cleared the virus, while two exhibited a high intra-host diversity favoring the establishment of severe outcomes. Neurovirulence-reverse mutations were detected in two patients, possibly leading to paralysis development. Furthermore, a recombination event, between type 3 Vaccine-Derived Poliovirus and Sabin-like1 (VDPV3/SL1), occurred in one patient. Our findings have suggested an association between intra-host diversity, recombination, prolonged excretion of the virus, and emergence of highly pathogenic strains. Further studies on intra-host diversity are crucial for a better understanding of the virus evolution as well as for the success of the Global Polio Eradication Initiative.


Assuntos
Fezes , Mutação , Poliomielite , Vacina Antipólio Oral , Poliovirus , Recombinação Genética , Eliminação de Partículas Virais , Humanos , Poliovirus/genética , Poliovirus/classificação , Poliovirus/isolamento & purificação , Poliovirus/imunologia , Vacina Antipólio Oral/genética , Vacina Antipólio Oral/efeitos adversos , Poliomielite/virologia , Poliomielite/prevenção & controle , Fezes/virologia , Masculino , Feminino , Genoma Viral/genética , Variação Genética , Doenças da Imunodeficiência Primária/genética , Pré-Escolar , Evolução Molecular , Criança , Lactente , Virulência/genética , Filogenia
16.
J Gen Virol ; 105(9)2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39319430

RESUMO

Fruit bats serve as an important reservoir for many zoonotic pathogens, including Nipah virus, Hendra virus, Marburg virus and Lyssavirus. To gain a deeper insight into the virological characteristics, pathogenicity and zoonotic potential of bat-borne viruses, recovery of infectious viruses from field samples is important. Here, we report the isolation and characterization of a mammalian orthoreovirus (MRV) from a large flying fox (Pteropus vampyrus) in Indonesia, which is the first detection of MRV in Southeast Asia. MRV was recovered from faecal samples of three different P. vampyrus in Central Java. Nucleotide sequence analysis revealed that the genome of the three MRV isolates shared more than 99% nucleotide sequence identity. We tentatively named one isolated strain as MRV12-52 for further analysis and characterization. Among 10 genome segments, MRV12-52 S1 and S4, which encode the cell-attachment protein and outer capsid protein, had 93.6 and 95.1% nucleotide sequence identities with known MRV strains, respectively. Meanwhile, the remaining genome segments of MRV12-52 were divergent with 72.9-80.7 % nucleotide sequence identities. Based on the nucleotide sequence of the S1 segment, MRV12-52 was grouped into serotype 2, and phylogenetic analysis demonstrated evidence of past reassortment events. In vitro characterization of MRV12-52 showed that the virus efficiently replicated in BHK-21, HEK293T and A549 cells. In addition, experimental infection of laboratory mice with MRV12-52 caused severe pneumonia with 75% mortality. This study highlights the presence of pathogenic MRV in Indonesia, which could serve as a potential animal and public health concern.


Assuntos
Quirópteros , Fezes , Genoma Viral , Orthoreovirus de Mamíferos , Filogenia , Infecções por Reoviridae , Animais , Quirópteros/virologia , Indonésia , Infecções por Reoviridae/virologia , Infecções por Reoviridae/veterinária , Camundongos , Fezes/virologia , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/isolamento & purificação , Orthoreovirus de Mamíferos/classificação , Humanos , Análise de Sequência de DNA
17.
Sheng Wu Gong Cheng Xue Bao ; 40(9): 3216-3232, 2024 Sep 25.
Artigo em Chinês | MEDLINE | ID: mdl-39319735

RESUMO

We analyzed the biological and genome characteristics of a phage infecting enteroinvasive Escherichia coli (EIEC), aiming to provide resources and a reference for the prevention and treatment of EIEC. With the EIEC preserved in our laboratory as the host bacterium, one strain of phage was isolated from the effluent sample from a chicken farm in Huzhou, Zhejiang and named ΦEP1. The titer, optimal multiplicity of infection, one-step growth curve, temperature, pH value, chloroform and bile salt sensitivity of ΦEP1 were determined by the double-layer agar plate method. The morphology of the phage was observed by transmission electron microscopy. The biocontrol effects of ΦEP1 in different food matrixes and the protective effect of this phage on Caco-2 cells were tested. The phage ΦEP1 showed the optimal multiplicity of infection of 0.1, the titer of 1.3×1010 PFU/mL, strong tolerance to temperature, pH, chloroform, and bile salt, and a broad host spectrum. Furthermore, it expressed lysis activity against multiple strains of multiple antibiotic-resistant pathogenic E. coli and Shigella with different serotypes. Phage ΦEP1 had an incubation period of 10 min, an outbreak period of 80 min, and an outbreak volume of 48 PFU/cell. According to the morphology observed by transmission electron microscopy, phage ΦEP1 belonged to the order of Caudovirales, and it had a good protective effect on Caco-2 cells. Phage ΦEP1 had a genome of 87 182 bp with the GC content of 39.80%, 128 putative open reading frames, and no antibiotic resistance genes or virulence genes. ΦEP1 inhibited the growth of EIEC in artificially contaminated milk and beef and eliminated EIEC in cell protection experiments. It significantly increased the survival rate of Caco-2 cells and down-regulated the expression of interleukin (IL)-6 and IL-1ß to reduce inflammation. We obtained an EIEC-targeting phage ΦEP1 with a high titer and strong tolerance to the environment, which provided a basis for the application of phages in food preservation and other fields.


Assuntos
Escherichia coli , Escherichia coli/virologia , Escherichia coli/genética , Humanos , Células CACO-2 , Animais , Genoma Viral , Especificidade de Hospedeiro , Bacteriófagos/genética , Bacteriófagos/isolamento & purificação , Galinhas/microbiologia
18.
PLoS One ; 19(9): e0310862, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39302958

RESUMO

INTRODUCTION: Emerging tick-borne viruses of medical and veterinary importance are increasingly being reported globally. This resurgence emphasizes the need for sustained surveillance to provide insights into tick-borne viral diversity and associated potential public health risks. We report on a virus tentatively designated Kinna virus (KIV) in the family Phenuiviridae and genus Bandavirus. The virus was isolated from a pool of Amblyomma gemma ticks from Kinna in Isiolo County, Kenya. High throughput sequencing of the virus isolate revealed close relatedness to the Guertu virus. The virus genome is consistent with the described genomes of other members of the genus Bandavirus, with nucleotides lengths of 6403, 3332 and 1752 in the Large (L), Medium (M) and Small (S) segments respectively. Phylogenetic analysis showed that the virus clustered with Guertu virus although it formed a distinct and well supported branch. The RdRp amino acid sequence had a 93.3% identity to that of Guertu virus, an indication that the virus is possibly novel. Neutralizing antibodies were detected in 125 (38.6%, 95% CI 33.3-44.1%) of the human sera from the communities in this region. In vivo experiments showed that the virus was lethal to mice with death occurring 6-9 days post-infection. The virus infected mammalian cells (Vero cells) but had reduced infectivity in the mosquito cell line (C636) tested. CONCLUSION: Isolation of this novel virus with the potential to cause disease in human and animal populations necessitates the need to evaluate its public health significance and contribution to disease burden in the affected regions. This also points to the need for continuous monitoring of vector and human populations in high-risk ecosystems to update pathogen diversity.


Assuntos
Amblyomma , Filogenia , Animais , Quênia/epidemiologia , Humanos , Amblyomma/virologia , Camundongos , Genoma Viral , Prevalência , Feminino , Masculino
19.
Mem Inst Oswaldo Cruz ; 119: e230214, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39319873

RESUMO

BACKGROUND: The human immunodeficiency virus 1 (HIV-1) infections in Brazil are predominantly caused by two subtypes, B and C. OBJECTIVES: Here we present the characterisation of a novel HIV-1 recombinant form, indicating a new Brazilian CRF_BC, named CRF146_BC. METHODS: RDP, JphMM and Simplot recombination tools were used to evaluate the mosaic pattern. FINDINGS: In this work, we identified three HIV-1 nucleotide sequences previously classified as unique recombinant forms (URFs), plus one new partial genome sharing the same BC recombination pattern. The mosaic genome is almost entirely represented by the subtype C sequence, with a small subtype B recombination region in the pol gene, at the Integrase level. The phylogenetic analyses strongly indicate a common origin between the strains, which were isolated in Rio Grande do Sul, Rio de Janeiro and Bahia states. MAIN CONCLUSIONS: Thus, the new HIV-1 CRF146_BC is circulating in three different Brazilian regions: South, Southeast and Northeast.


Assuntos
Infecções por HIV , HIV-1 , Filogenia , Recombinação Genética , HIV-1/genética , HIV-1/classificação , Humanos , Brasil/epidemiologia , Infecções por HIV/virologia , Genótipo , RNA Viral/genética , Genoma Viral/genética
20.
Gigascience ; 132024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-39320317

RESUMO

BACKGROUND: Antimicrobial resistance is a serious threat to global health. Due to the stagnant antibiotic discovery pipeline, bacteriophages (phages) have been proposed as an alternative therapy for the treatment of infections caused by multidrug-resistant pathogens. Genomic features play an important role in phage pharmacology. However, our knowledge of phage genomics is sparse, and the use of existing bioinformatic pipelines and tools requires considerable bioinformatic expertise. These challenges have substantially limited the clinical translation of phage therapy. FINDINGS: We have developed PhageGE (Phage Genome Explorer), a user-friendly graphical interface application for the interactive analysis of phage genomes. PhageGE enables users to perform key analyses, including phylogenetic analysis, visualization of phylogenetic trees, prediction of phage life cycle, and comparative analysis of phage genome annotations. The new R Shiny web server, PhageGE, integrates existing R packages and combines them with several newly developed functions to facilitate these analyses. Additionally, the web server provides interactive visualization capabilities and allows users to directly export publication-quality images. CONCLUSIONS: PhageGE is a valuable tool that simplifies the analysis of phage genome data and may expedite the development and clinical translation of phage therapy. PhageGE is publicly available at https://jason-zhao.shinyapps.io/PhageGE_Update/.


Assuntos
Bacteriófagos , Genoma Viral , Software , Bacteriófagos/genética , Genômica/métodos , Biologia Computacional/métodos , Internet , Filogenia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA