Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 465
Filtrar
1.
Nature ; 626(7998): 392-400, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38086420

RESUMO

An ideal vaccine both attenuates virus growth and disease in infected individuals and reduces the spread of infections in the population, thereby generating herd immunity. Although this strategy has proved successful by generating humoral immunity to measles, yellow fever and polio, many respiratory viruses evolve to evade pre-existing antibodies1. One approach for improving the breadth of antiviral immunity against escape variants is through the generation of memory T cells in the respiratory tract, which are positioned to respond rapidly to respiratory virus infections2-6. However, it is unknown whether memory T cells alone can effectively surveil the respiratory tract to the extent that they eliminate or greatly reduce viral transmission following exposure of an individual to infection. Here we use a mouse model of natural parainfluenza virus transmission to quantify the extent to which memory CD8+ T cells resident in the respiratory tract can provide herd immunity by reducing both the susceptibility of acquiring infection and the extent of transmission, even in the absence of virus-specific antibodies. We demonstrate that protection by resident memory CD8+ T cells requires the antiviral cytokine interferon-γ (IFNγ) and leads to altered transcriptional programming of epithelial cells within the respiratory tract. These results suggest that tissue-resident CD8+ T cells in the respiratory tract can have important roles in protecting the host against viral disease and limiting viral spread throughout the population.


Assuntos
Linfócitos T CD8-Positivos , Memória Imunológica , Células T de Memória , Infecções por Paramyxoviridae , Sistema Respiratório , Animais , Camundongos , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Imunidade Coletiva/imunologia , Memória Imunológica/imunologia , Interferon gama/imunologia , Células T de Memória/imunologia , Paramyxoviridae/imunologia , Paramyxoviridae/fisiologia , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Infecções por Paramyxoviridae/transmissão , Infecções por Paramyxoviridae/virologia , Sistema Respiratório/citologia , Sistema Respiratório/imunologia , Sistema Respiratório/virologia , Transcrição Gênica , Humanos
2.
mBio ; 14(5): e0139123, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37737607

RESUMO

IMPORTANCE: Genetically diverse paramyxoviruses are united in their presentation of a receptor-binding protein (RBP), which works in concert with the fusion protein to facilitate host-cell entry. The C-terminal head region of the paramyxoviral RBP, a primary determinant of host-cell tropism and inter-species transmission potential, forms structurally distinct classes dependent upon protein and glycan receptor specificity. Here, we reveal the architecture of the C-terminal head region of the RBPs from Nariva virus (NarV) and Mossman virus (MosV), two archetypal rodent-borne paramyxoviruses within the recently established genus Narmovirus, family Paramyxoviridae. Our analysis reveals that while narmoviruses retain the general architectural features associated with paramyxoviral RBPs, namely, a six-bladed ß-propeller fold, they lack the structural motifs associated with known receptor-mediated host-cell entry pathways. This investigation indicates that the RBPs of narmoviruses exhibit pathobiological features that are distinct from those of other paramyxoviruses.


Assuntos
Proteínas de Transporte , Paramyxovirinae , Proteínas de Transporte/metabolismo , Paramyxoviridae , Proteínas Virais de Fusão/metabolismo , Ligação Proteica , Internalização do Vírus
3.
Front Immunol ; 13: 970750, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36045682

RESUMO

Retinoic acid-inducible gene I (RIG-I) is a receptor that senses viral RNA and interacts with mitochondrial antiviral signaling (MAVS) protein, leading to the production of type I interferons and inflammatory cytokines to establish an antiviral state. This signaling axis is initiated by the K63-linked RIG-I ubiquitination, mediated by E3 ubiquitin ligases such as TRIM25. However, many viruses, including several members of the family Paramyxoviridae and human respiratory syncytial virus (HRSV), a member of the family Pneumoviridae, escape the immune system by targeting RIG-I/TRIM25 signaling. In this study, we screened human metapneumovirus (HMPV) open reading frames (ORFs) for their ability to block RIG-I signaling reconstituted in HEK293T cells by transfection with TRIM25 and RIG-I CARD (an N-terminal CARD domain that is constitutively active in RIG-I signaling). HMPV M2-2 was the most potent inhibitor of RIG-I/TRIM25-mediated interferon (IFN)-ß activation. M2-2 silencing induced the activation of transcription factors (IRF and NF-kB) downstream of RIG-I signaling in A549 cells. Moreover, M2-2 inhibited RIG-I ubiquitination and CARD-dependent interactions with MAVS. Immunoprecipitation revealed that M2-2 forms a stable complex with RIG-I CARD/TRIM25 via direct interaction with the SPRY domain of TRIM25. Similarly, HRSV NS1 also formed a stable complex with RIG-I CARD/TRIM25 and inhibited RIG-I ubiquitination. Notably, the inhibitory actions of HMPV M2-2 and HRSV NS1 are similar to those of V proteins of several members of the Paramyxoviridae family. In this study, we have identified a novel mechanism of immune escape by HMPV, similar to that of Pneumoviridae and Paramyxoviridae family members.


Assuntos
Interferon Tipo I , Metapneumovirus , Infecções por Paramyxoviridae/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Antivirais , Proteína DEAD-box 58/metabolismo , Células HEK293 , Humanos , Imunidade Inata , Interferon Tipo I/metabolismo , Interferon beta/metabolismo , Paramyxoviridae , Infecções por Paramyxoviridae/virologia , Receptores Imunológicos/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
4.
PLoS One ; 16(12): e0260360, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34855795

RESUMO

Recent emergence of SARS-CoV-2 and associated COVID-19 pandemic have posed a great challenge for the scientific community. In this study, we performed bioinformatic analyses on SARS-CoV-2 protein sequences, trying to unravel potential molecular similarities between this newly emerged pathogen with non-coronavirus ssRNA viruses. Comparing the proteins of SARS-CoV-2 with non-coronavirus positive and negative strand ssRNA viruses revealed multiple sequence similarities between SARS-CoV-2 and non-coronaviruses, including similarities between RNA-dependent RNA-polymerases and helicases (two highly-conserved proteins). We also observed similarities between SARS-CoV-2 surface (i.e. spike) protein with paramyxovirus fusion proteins. This similarity was restricted to a segment of spike protein S2 subunit which is involved in cell fusion. We next analyzed spike proteins from SARS-CoV-2 "variants of concern" (VOCs) and "variants of interests" (VOIs) and found that some of these variants show considerably higher spike-fusion similarity with paramyxoviruses. The 'spike-fusion' similarity was also observed for some pathogenic coronaviruses other than SARS-CoV-2. Epitope analysis using experimentally verified data deposited in Immune Epitope Database (IEDB) revealed that several B cell epitopes as well as T cell and MHC binding epitopes map within the spike-fusion similarity region. These data indicate that there might be a degree of convergent evolution between SARS-CoV-2 and paramyxovirus surface proteins which could be of pathogenic and immunological importance.


Assuntos
SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , Proteínas Virais de Fusão/genética , Epitopos/genética , Humanos , Paramyxoviridae/genética , Filogenia , Estrutura Terciária de Proteína , Glicoproteína da Espícula de Coronavírus/química
5.
Virology ; 562: 40-49, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34256243

RESUMO

Paramyxoviruses harbored by multiple natural reservoirs pose a potential threat to public health. Jeilongvirus has been proposed as a novel paramyxovirus genus found in rodents, bats, and cats. Paramyxovirus RNA was detected in 108/824 (13.1%) Apodemus agrarius captured at 14 trapping sites in the Republic of Korea. We first present two genetically distinct novel paramyxoviruses, Paju Apodemus paramyxovirus 1 (PAPV-1) and 2 (PAPV-2). The disparity between PAPV-1 (19,716 nucleotides) and -2 (17,475 nucleotides) revealed the presence of the SH gene and length of the G gene in the genome organization. The phylogeny of PAPV-1 and -2 belonged to distinct genetic lineages of Jeilongvirus, respectively, even though these viruses were originated from A. agrarius. PAPV-1 infected human epithelial and endothelial cells, facilitating the induction of type I/III interferons, interferon-stimulated genes, and pro-inflammatory cytokines. Therefore, this study provides insights into the molecular epidemiology, genetic diversity, and virus-host interactions of novel rodent-borne paramyxoviruses.


Assuntos
Murinae/virologia , Paramyxoviridae/classificação , Paramyxoviridae/genética , Animais , Citocinas/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Genoma Viral/genética , Humanos , Filogenia , RNA Viral/genética , República da Coreia , Especificidade da Espécie , Proteínas Virais/genética , Replicação Viral
6.
J Med Virol ; 93(11): 6140-6147, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34160830

RESUMO

To investigate the presence of respiratory viruses in the middle ear cavity of the individuals with a healthy middle ear and the children with otitis media with effusion (OME). A total of 72 middle ear samples were collected from 25 children with OME (Group 1) and 47 individuals with no middle ear disease (Group 2). Multiplex real-time polymerase chain reaction was used to investigate the presence of 20 different respiratory viruses. Virus results were compared with bacteriomes of the same populations. At least one respiratory virus was detected in 56% of the patients in Group 1 and 12.8% of the individuals in Group 2. The viral co-infection rate for Group 1 and 2 was 8% and 2.1%, respectively. In Group 1, adenovirus was the most frequently detected virus with a rate of 24%, either alone (16%) or concurrent with other viruses (8%), followed by influenza B (12%), rhinovirus, and bocavirus (8%) each. Parainfluenza 4, coronavirus OC43, and RSV A/B were detected in 4% of the sample each. In Group 2, rhinovirus was detected in two samples (4.3%) followed by adenovirus, coronavirus OC43, coronavirus E299, and coronavirus NL63 with a rate of 2.1% each. The detection rate of respiratory viruses was significantly higher in children aged 6 to 11 years. There was no positive association between virus and bacteria found in the middle ear cavity. The current study has provided comprehensive data indicating the presence of diverse respiratory viruses in the healthy middle ear cavity. Our results also suggest that respiratory viruses might have a contribution to OME pathogenesis.


Assuntos
Orelha Média/virologia , Otite Média com Derrame/virologia , Vírus/isolamento & purificação , Adenovírus Humanos/isolamento & purificação , Bactérias/isolamento & purificação , Criança , Pré-Escolar , Coinfecção , Coronavirus/isolamento & purificação , Feminino , Bocavirus Humano/isolamento & purificação , Humanos , Lactente , Masculino , Orthomyxoviridae/isolamento & purificação , Otite Média com Derrame/microbiologia , Paramyxoviridae/isolamento & purificação , Rhinovirus/isolamento & purificação , Viroses/virologia
7.
PLoS Pathog ; 16(10): e1008877, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33035269

RESUMO

The antigenic and genomic stability of paramyxoviruses remains a mystery. Here, we evaluate the genetic plasticity of Sendai virus (SeV) and mumps virus (MuV), sialic acid-using paramyxoviruses that infect mammals from two Paramyxoviridae subfamilies (Orthoparamyxovirinae and Rubulavirinae). We performed saturating whole-genome transposon insertional mutagenesis, and identified important commonalities: disordered regions in the N and P genes near the 3' genomic end were more tolerant to insertional disruptions; but the envelope glycoproteins were not, highlighting structural constraints that contribute to the restricted antigenic drift in paramyxoviruses. Nonetheless, when we applied our strategy to a fusion-defective Newcastle disease virus (Avulavirinae subfamily), we could select for F-revertants and other insertants in the 5' end of the genome. Our genome-wide interrogation of representative paramyxovirus genomes from all three Paramyxoviridae subfamilies provides a family-wide context in which to explore specific variations within and among paramyxovirus genera and species.


Assuntos
Elementos de DNA Transponíveis/genética , Genoma Viral , Mutagênese Insercional , Mutação , Infecções por Paramyxoviridae/virologia , Paramyxoviridae/genética , Proteínas Virais de Fusão/genética , Humanos , Paramyxoviridae/classificação
8.
Arch Virol ; 165(10): 2165-2176, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32740830

RESUMO

The PI3K/Akt signalling pathway is a crucial signalling cascade that regulates transcription, protein translation, cell growth, proliferation, cell survival, and metabolism. During viral infection, viruses exploit a variety of cellular pathways, including the well-known PI3K/Akt signalling pathway. Conversely, cells rely on this pathway to stimulate an antiviral response. The PI3K/Akt pathway is manipulated by a number of viruses, including DNA and RNA viruses and retroviruses. The aim of this review is to provide up-to-date information about the role of the PI3K-Akt pathway in infection with members of five different families of negative-sense ssRNA viruses. This pathway is hijacked for viral entry, regulation of endocytosis, suppression of premature apoptosis, viral protein expression, and replication. Although less common, the PI3K/Akt pathway can be downregulated as an immunomodulatory strategy or as a mechanism for inducing autophagy. Moreover, the cell activates this pathway as an antiviral strategy for interferon and cytokine production, among other strategies. Here, we present new data concerning the role of this pathway in infection with the paramyxovirus Newcastle disease virus (NDV). Our data seem to indicate that NDV uses the PI3K/Akt pathway to delay cell death and increase cell survival as a means of improving its replication. The interference of negative-sense ssRNA viruses with this essential pathway might have implications for the development of antiviral therapies.


Assuntos
Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Fosfatidilinositol 3-Quinase/genética , Proteínas Proto-Oncogênicas c-akt/genética , Infecções por Vírus de RNA/genética , Apoptose/genética , Autofagia/genética , Autofagia/imunologia , Citocinas/genética , Citocinas/imunologia , Endocitose/genética , Endocitose/imunologia , Filoviridae/genética , Filoviridae/metabolismo , Filoviridae/patogenicidade , Interações Hospedeiro-Patógeno/imunologia , Interferons/genética , Interferons/imunologia , Orthomyxoviridae/genética , Orthomyxoviridae/metabolismo , Orthomyxoviridae/patogenicidade , Paramyxoviridae/genética , Paramyxoviridae/metabolismo , Paramyxoviridae/patogenicidade , Fosfatidilinositol 3-Quinase/imunologia , Pneumovirinae/genética , Pneumovirinae/metabolismo , Pneumovirinae/patogenicidade , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas c-akt/imunologia , Infecções por Vírus de RNA/imunologia , Infecções por Vírus de RNA/virologia , Rhabdoviridae/genética , Rhabdoviridae/metabolismo , Rhabdoviridae/patogenicidade , Transdução de Sinais , Proteínas Virais/genética , Proteínas Virais/imunologia , Internalização do Vírus , Replicação Viral
9.
J Virol ; 94(18)2020 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-32581107

RESUMO

Wild birds are major natural reservoirs and potential dispersers of a variety of infectious diseases. As such, it is important to determine the diversity of viruses they carry and use this information to help understand the potential risks of spillover to humans, domestic animals, and other wildlife. We investigated the potential viral causes of paresis in long-standing, but undiagnosed, disease syndromes in wild Australian birds. RNA from diseased birds was extracted and pooled based on tissue type, host species, and clinical manifestation for metagenomic sequencing. Using a bulk and unbiased metatranscriptomic approach, combined with clinical investigation and histopathology, we identified a number of novel viruses from the families Astroviridae, Adenoviridae, Picornaviridae, Polyomaviridae, Paramyxoviridae, Parvoviridae, and Circoviridae in common urban wild birds, including Australian magpies, magpie larks, pied currawongs, Australian ravens, and rainbow lorikeets. In each case, the presence of the virus was confirmed by reverse transcription (RT)-PCR. These data revealed a number of candidate viral pathogens that may contribute to coronary, skeletal muscle, vascular, and neuropathology in birds of the Corvidae and Artamidae families and neuropathology in members of the Psittaculidae The existence of such a diverse virome in urban avian species highlights the importance and challenges in elucidating the etiology and ecology of wildlife pathogens in urban environments. This information will be increasingly important for managing disease risks and conducting surveillance for potential viral threats to wildlife, livestock, and human health.IMPORTANCE Wildlife naturally harbor a diverse array of infectious microorganisms and can be a source of novel diseases in domestic animals and human populations. Using unbiased RNA sequencing, we identified highly diverse viruses in native birds from Australian urban environments presenting with paresis. This research included the clinical investigation and description of poorly understood recurring syndromes of unknown etiology: clenched claw syndrome and black and white bird disease. As well as identifying a range of potentially disease-causing viral pathogens, this study describes methods that can effectively and efficiently characterize emergent disease syndromes in free-ranging wildlife and promotes further surveillance for specific pathogens of potential conservation and zoonotic concern.


Assuntos
Animais Selvagens/virologia , Doenças das Aves/epidemiologia , Aves/virologia , Infecções por Vírus de DNA/veterinária , Metagenoma , Infecções por Vírus de RNA/veterinária , Transcriptoma , Adenoviridae/classificação , Adenoviridae/genética , Adenoviridae/isolamento & purificação , Animais , Astroviridae/classificação , Astroviridae/genética , Astroviridae/isolamento & purificação , Austrália/epidemiologia , Doenças das Aves/virologia , Circoviridae/classificação , Circoviridae/genética , Circoviridae/isolamento & purificação , Cidades , Infecções por Vírus de DNA/epidemiologia , Infecções por Vírus de DNA/virologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Paramyxoviridae/classificação , Paramyxoviridae/genética , Paramyxoviridae/isolamento & purificação , Parvoviridae/classificação , Parvoviridae/genética , Parvoviridae/isolamento & purificação , Filogenia , Picornaviridae/classificação , Picornaviridae/genética , Picornaviridae/isolamento & purificação , Polyomaviridae/classificação , Polyomaviridae/genética , Polyomaviridae/isolamento & purificação , Infecções por Vírus de RNA/epidemiologia , Infecções por Vírus de RNA/virologia
10.
J Vet Diagn Invest ; 32(3): 429-434, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32194003

RESUMO

We screened 104 snakes with respiratory disease, collected from 52 snake farms in Guangxi Province, China, for pathogens. Ferlaviruses were detected in 70 of 104 lung samples by reverse-transcription PCR; 34 of 52 of the snake farms were positive for ferlaviruses. No reovirus, adenovirus, sunshine virus, or nidovirus was detected in any of the snakes. We obtained 96 bacterial isolates from snake organs, of which the most commonly isolated species were Salmonella (18) and Proteus (16). Sequence analysis, based on 27 partial RNA-dependent RNA polymerase gene (L) sequences, revealed that ferlaviruses from Guangxi and the known GenBank strains clustered together and formed 3 genogroups. The nucleotide and deduced amino acid homologies of ferlaviruses were 84.3-100% and 95.0-100% within groups, respectively, and 77.0-81.6% and 90.4-95.2% between groups, respectively. Ferlaviruses from Guangxi had close genetic relationships with the known GenBank strains. Our results indicate that ferlaviruses are common in snakes with respiratory disease on the farms of Guangxi that we sampled, and that ferlavirus molecular epidemiology is both diverse and complex.


Assuntos
Epidemiologia Molecular , Infecções por Paramyxoviridae/veterinária , Paramyxoviridae/genética , Paramyxoviridae/isolamento & purificação , Serpentes/virologia , Animais , China/epidemiologia , Genótipo , Infecções por Paramyxoviridae/epidemiologia , Infecções por Paramyxoviridae/virologia , Filogenia , Reação em Cadeia da Polimerase/veterinária , Doenças Respiratórias/veterinária
11.
Viruses ; 12(2)2020 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-32019182

RESUMO

Paramyxovirus (PMV) entry requires the coordinated action of two envelope glycoproteins, the receptor binding protein (RBP) and fusion protein (F). The sequence of events that occurs during the PMV entry process is tightly regulated. This regulation ensures entry will only initiate when the virion is in the vicinity of a target cell membrane. Here, we review recent structural and mechanistic studies to delineate the entry features that are shared and distinct amongst the Paramyxoviridae. In general, we observe overarching distinctions between the protein-using RBPs and the sialic acid- (SA-) using RBPs, including how their stalk domains differentially trigger F. Moreover, through sequence comparisons, we identify greater structural and functional conservation amongst the PMV fusion proteins, as compared to the RBPs. When examining the relative contributions to sequence conservation of the globular head versus stalk domains of the RBP, we observe that, for the protein-using PMVs, the stalk domains exhibit higher conservation and find the opposite trend is true for SA-using PMVs. A better understanding of conserved and distinct features that govern the entry of protein-using versus SA-using PMVs will inform the rational design of broader spectrum therapeutics that impede this process.


Assuntos
Paramyxoviridae/genética , Proteínas Virais de Fusão/química , Internalização do Vírus , Proteínas de Transporte , Humanos , Ácido N-Acetilneuramínico/metabolismo , Paramyxoviridae/classificação , Paramyxoviridae/fisiologia , Ligação Proteica , Proteínas Virais de Fusão/genética , Ligação Viral
12.
Antiviral Res ; 175: 104712, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31935422

RESUMO

Nipah virus (NiV) is a highly pathogenic zoonotic paramyxovirus that continues to cause outbreaks in humans characterized by high mortality and significant clinical sequelae in survivors. Currently, no therapeutics are approved for use in humans against NiV infection. Here, we report that 4'-chloromethyl-2'-deoxy-2'-fluorocytidine (ALS-8112) inhibits NiV. ALS-8112 is the parent nucleoside of lumicitabine, which has been evaluated in phase I and II clinical trials to treat pediatric and adult respiratory syncytial virus infection. In this study, we tested ALS-8112 against NiV and other major human respiratory pneumo- and paramyxoviruses in 2 human lung epithelial cell lines, and demonstrated the ability of ALS-8112 to reduce infectious wild-type NiV yield by over 6 orders of magnitude with no apparent cytotoxicity. However, further cytotoxicity testing in primary cells and bone marrow progenitor cells indicated cytotoxicity at higher concentrations of ALS-8112. Our results warrant the evaluation of lumicitabine against NiV infection in relevant animal models.


Assuntos
Antivirais/farmacologia , Desoxicitidina/análogos & derivados , Vírus Nipah/efeitos dos fármacos , Antivirais/química , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Desoxicitidina/química , Desoxicitidina/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Células HeLa , Humanos , Pulmão/citologia , Nucleosídeos/química , Nucleosídeos/farmacologia , Paramyxoviridae/efeitos dos fármacos
13.
J Med Virol ; 92(8): 971-981, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31769525

RESUMO

Active surveillance for zoonotic respiratory viruses is essential to inform the development of appropriate interventions and outbreak responses. Here we target individuals with a high frequency of animal exposure in Vietnam. Three-year community-based surveillance was conducted in Vietnam during 2013-2016. We enrolled a total of 581 individuals (animal-raising farmers, slaughterers, animal-health workers, and rat traders), and utilized reverse transcription-polymerase chain reaction to detect 15 common respiratory viruses in pooled nasal-throat swabs collected at baseline or acute respiratory disease episodes. A respiratory virus was detected in 7.9% (58 of 732) of baseline samples, and 17.7% (136 of 770) of disease episode samples (P < .001), with enteroviruses (EVs), rhinoviruses and influenza A virus being the predominant viruses detected. There were temporal and spatial fluctuations in the frequencies of the detected viruses over the study period, for example, EVs and influenza A viruses were more often detected during rainy seasons. We reported the detection of common respiratory viruses in individuals with a high frequency of animal exposure in Vietnam, an emerging infectious disease hotspot. The results show the value of baseline/control sampling in delineating the causative relationships and have revealed important insights into the ecological aspects of EVs, rhinoviruses and influenza A and their contributions to the burden posed by respiratory infections in Vietnam.


Assuntos
Exposição Ocupacional , Infecções Respiratórias/epidemiologia , Infecções Respiratórias/virologia , Viroses/epidemiologia , Viroses/virologia , Adenoviridae/isolamento & purificação , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Coronavirus/isolamento & purificação , Fazendeiros , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Orthomyxoviridae/isolamento & purificação , Paramyxoviridae/isolamento & purificação , Picornaviridae/isolamento & purificação , Estações do Ano , Vietnã/epidemiologia , Zoonoses Virais/epidemiologia , Adulto Jovem
14.
mBio ; 10(3)2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31088929

RESUMO

Paramyxoviruses and pneumoviruses have similar life cycles and share the respiratory tract as a point of entry. In comparative genome-scale siRNA screens with wild-type-derived measles, mumps, and respiratory syncytial viruses in A549 cells, a human lung adenocarcinoma cell line, we identified vesicular transport, RNA processing pathways, and translation as the top pathways required by all three viruses. As the top hit in the translation pathway, ABCE1, a member of the ATP-binding cassette transporters, was chosen for further study. We found that ABCE1 supports replication of all three viruses, confirming its importance for viruses of both families. More detailed characterization revealed that ABCE1 is specifically required for efficient viral but not general cellular protein synthesis, indicating that paramyxoviral and pneumoviral mRNAs exploit specific translation mechanisms. In addition to providing a novel overview of cellular proteins and pathways that impact these important pathogens, this study highlights the role of ABCE1 as a host factor required for efficient paramyxovirus and pneumovirus translation.IMPORTANCE The Paramyxoviridae and Pneumoviridae families include important human and animal pathogens. To identify common host factors, we performed genome-scale siRNA screens with wild-type-derived measles, mumps, and respiratory syncytial viruses in the same cell line. A comparative bioinformatics analysis yielded different members of the coatomer complex I, translation factors ABCE1 and eIF3A, and several RNA binding proteins as cellular proteins with proviral activity for all three viruses. A more detailed characterization of ABCE1 revealed its essential role for viral protein synthesis. Taken together, these data sets provide new insight into the interactions between paramyxoviruses and pneumoviruses and host cell proteins and constitute a starting point for the development of broadly effective antivirals.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Interações entre Hospedeiro e Microrganismos/genética , Paramyxoviridae/patogenicidade , Pneumovirus/patogenicidade , Células A549 , Biologia Computacional , Expressão Gênica , Humanos , RNA Mensageiro , RNA Interferente Pequeno , Proteínas de Ligação a RNA/genética
15.
PLoS One ; 14(4): e0214227, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30969980

RESUMO

Bats are implicated as natural reservoirs for a wide range of zoonotic viruses including SARS and MERS coronaviruses, Ebola, Marburg, Nipah, Hendra, Rabies and other lyssaviruses. Accordingly, many One Health surveillance and viral discovery programs have focused on bats. In this report we present viral metagenomic data from bats collected in the Kingdom of Saudi Arabia [KSA]. Unbiased high throughput sequencing of fecal samples from 72 bat individuals comprising four species; lesser mouse-tailed bat (Rhinopoma hardwickii), Egyptian tomb bat (Taphozous perforatus), straw-colored fruit bat (Eidolon helvum), and Egyptian fruit bat (Rousettus aegyptiacus) revealed molecular evidence of a diverse set of viral families: Picornaviridae (hepatovirus, teschovirus, parechovirus), Reoviridae (rotavirus), Polyomaviridae (polyomavirus), Papillomaviridae (papillomavirus), Astroviridae (astrovirus), Caliciviridae (sapovirus), Coronaviridae (coronavirus), Adenoviridae (adenovirus), Paramyxoviridae (paramyxovirus), and unassigned mononegavirales (chuvirus). Additionally, we discovered a bastro-like virus (Middle East Hepe-Astrovirus), with a genomic organization similar to Hepeviridae. However, since it shared homology with Hepeviridae and Astroviridae at ORF1 and in ORF2, respectively, the newly discovered Hepe-Astrovirus may represent a phylogenetic bridge between Hepeviridae and Astroviridae.


Assuntos
Quirópteros/virologia , Metagenoma/genética , Metagenômica , Filogenia , Animais , Caliciviridae/genética , Caliciviridae/isolamento & purificação , Quirópteros/genética , Egito , Fezes/virologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Mamíferos/virologia , Oriente Médio , Coronavírus da Síndrome Respiratória do Oriente Médio , Paramyxoviridae/genética , Paramyxoviridae/isolamento & purificação , Picornaviridae/genética , Picornaviridae/isolamento & purificação , Vírus de RNA/genética , Rotavirus/genética , Rotavirus/isolamento & purificação , Arábia Saudita
16.
Viruses ; 11(3)2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30871070

RESUMO

Bats are unique mammals, exhibit distinctive life history traits and have unique immunological approaches to suppression of viral diseases upon infection. High-throughput next-generation sequencing has been used in characterizing the virome of different bat species. The cave nectar bat, Eonycteris spelaea, has a broad geographical range across Southeast Asia, India and southern China, however, little is known about their involvement in virus transmission. Here we investigate the diversity and abundance of viral communities from a colony of Eonycteris spelaea residing in Singapore. Our results detected 47 and 22 different virus families from bat fecal and urine samples, respectively. Among these, we identify a large number of virus families including Adenoviridae, Flaviviridae, Reoviridae, Papillomaviridae, Paramyxoviridae, Parvoviridae, Picornaviridae, and Polyomaviridae. In most cases, viral sequences from Eonycteris spelaea are genetically related to a group of bat viruses from other bat genera (e.g., Eidolon, Miniopterus, Rhinolophus and Rousettus). The results of this study improve our knowledge of the host range, spread and evolution of several important viral pathogens. More significantly, our findings provide a baseline to study the temporal patterns of virus shedding and how they correlate with bat phenological trends.


Assuntos
Quirópteros/virologia , Evolução Molecular , Variação Genética , Vírus/genética , Vírus/patogenicidade , Adenoviridae/genética , Animais , Sudeste Asiático/epidemiologia , Fezes/virologia , Genoma Viral , Herpesviridae/genética , Sequenciamento de Nucleotídeos em Larga Escala , Paramyxoviridae/genética , Filogenia , Viroses/epidemiologia , Viroses/urina , Viroses/veterinária
17.
Front Immunol ; 10: 2861, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31921130

RESUMO

Due to their overall immunocompromised state, lung transplant recipients (LTRs) are at increased risk for the development of viral respiratory infections compared to the general population. Such respiratory infections often lead to poor transplant outcomes. We performed a systematic review of the last 30 years of medical literature to summarize the impact of specific respiratory viruses on LTRs. After screening 2,150 articles for potential inclusion, 39 manuscripts were chosen for final review. We found evidence for an association of respiratory viruses including respiratory syncytial virus (RSV), parainfluenza virus, and influenza viruses with increased morbidity following transplant. Through the literature search, we also documented associations of RSV and adenovirus infections with increased mortality among LTRs. We posit that the medical literature supports aggressive surveillance for respiratory viruses among this population.


Assuntos
Hospedeiro Imunocomprometido , Transplante de Pulmão , Orthomyxoviridae/imunologia , Paramyxoviridae/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Infecções Respiratórias , Humanos , Infecções Respiratórias/imunologia , Infecções Respiratórias/mortalidade , Infecções Respiratórias/virologia
18.
J Virol ; 93(6)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30567988

RESUMO

The acute antiviral response is mediated by a family of interferon-stimulated genes (ISGs), providing cell-intrinsic immunity. Mutations in genes encoding these proteins are often associated with increased susceptibility to viral infections. One family of ISGs with antiviral function is the interferon-inducible transmembrane proteins (IFITMs), of which IFITM3 has been studied extensively. In contrast, IFITM1 has not been studied in detail. Since IFITM1 can localize to the plasma membrane, we investigated its function with a range of enveloped viruses thought to infect cells by fusion with the plasma membrane. Overexpression of IFITM1 prevented infection by a number of Paramyxoviridae and Pneumoviridae, including respiratory syncytial virus (RSV), mumps virus, and human metapneumovirus (HMPV). IFITM1 also restricted infection with an enveloped DNA virus that can enter via the plasma membrane, herpes simplex virus 1 (HSV-1). To test the importance of plasma membrane localization for IFITM1 function, we identified blocks of amino acids in the conserved intracellular loop (CIL) domain that altered the subcellular localization of the protein and reduced antiviral activity. By screening reported data sets, 12 rare nonsynonymous single nucleotide polymorphisms (SNPs) were identified in human IFITM1, some of which are in the CIL domain. Using an Ifitm1-/- mouse, we show that RSV infection was more severe, thereby extending the range of viruses restricted in vivo by IFITM proteins and suggesting overall that IFITM1 is broadly antiviral and that this antiviral function is associated with cell surface localization.IMPORTANCE Host susceptibility to viral infection is multifactorial, but early control of viruses not previously encountered is predominantly mediated by the interferon-stimulated gene (ISG) family. There are upwards of 300 of these genes, the majority of which do not have a clearly defined function or mechanism of action. The cellular location of these proteins may have an important effect on their function. One ISG located at the plasma membrane is interferon-inducible transmembrane protein 1 (IFITM1). Here we demonstrate that IFITM1 can inhibit infection with a range of viruses that enter via the plasma membrane. Mutant IFITM1 proteins that were unable to localize to the plasma membrane did not restrict viral infection. We also observed for the first time that IFITM1 plays a role in vivo, and Ifitm1-/- mice were more susceptible to viral lung infection. These data contribute to our understanding of how ISGs prevent viral infections.


Assuntos
Antígenos de Diferenciação/metabolismo , Membrana Celular/virologia , Paramyxoviridae/efeitos dos fármacos , Pneumovirinae/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Células A549 , Sequência de Aminoácidos , Animais , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Humanos , Interferons/farmacologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polimorfismo de Nucleotídeo Único/efeitos dos fármacos , Células Vero
19.
J Zoo Wildl Med ; 49(4): 983-995, 2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30592933

RESUMO

Between June and October 2013, 26 snakes of six viperid species kept in two adjoining rooms died ( n = 16) or were euthanized on medical (1) or welfare grounds (9). Two were from the main zoo collection, but the other 24 had been imported and quarantined for a minimum of 6 mo. Four of those that died and the single snake euthanized on medical grounds showed minor signs of respiratory disease prior to death, and five were weak, lethargic, and/or poor feeders. Frequent postmortem findings among all snakes were poor body condition (18) and respiratory disease (13). Seventeen cases were examined histologically, and pneumonia, sometimes with air sacculitis and/or tracheitis, was present in 15 individuals. Lung samples from 24 snakes were ferlavirus polymerase chain reaction (PCR) positive, and one of the two snakes for which only liver was available was also positive. The negative liver sample was from a snake that died of sepsis following anesthesia for surgical removal of a spindle cell sarcoma. Correlation with antemortem PCR testing of glottal and cloacal swabs in five cases was poor (sensitivity = 40%). Immunohistochemistry (IHC) for ferlaviruses on the tissues of 13 PCR-positive cases showed positive labeling in 7 only. Tissues samples from 22 ferlavirus PCR-positive snakes were examined for Chlamydia species by PCR, and 9 were positive, although DNA sequencing only confirmed two of three tested as Chlamydia pneumoniae. Immunohistochemistry for Chlamydia pneumoniae of seven cases (two Chlamydiales PCR positive, one of which was sequenced as C. pneumoniae, plus five negative) confirmed the Chlamydia PCR results. These two Chlamydiales PCR and IHC positive snakes were ferlavirus PCR positive, but IHC negative suggesting that, even though a ferlavirus was the predominant cause of the outbreak, in a few cases death may have been due to chlamydiosis with ferlavirus present, but not acting as the primary pathogen.


Assuntos
Surtos de Doenças/veterinária , Infecções por Paramyxoviridae/veterinária , Paramyxoviridae/isolamento & purificação , Viperidae , Animais , Animais de Zoológico , Feminino , Londres/epidemiologia , Masculino , Infecções por Paramyxoviridae/epidemiologia , Infecções por Paramyxoviridae/mortalidade , Infecções por Paramyxoviridae/virologia
20.
J Virol ; 92(20)2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30068647

RESUMO

J paramyxovirus (JPV) was first isolated from moribund mice with hemorrhagic lung lesions in Australia in 1972. It is a paramyxovirus classified under the newly proposed genus Jeilongvirus JPV has a genome of 18,954 nucleotides, consisting of eight genes in the order 3'-N-P/V/C-M-F-SH-TM-G-L-5'. JPV causes little cytopathic effect (CPE) in tissue culture cells but severe disease in mice. The small hydrophobic (SH) protein is an integral membrane protein encoded by many paramyxoviruses, such as mumps virus (MuV) and respiratory syncytial virus (RSV). However, the function of SH has not been defined in a suitable animal model. In this work, the functions of SH of JPV, MuV, and RSV have been examined by generating recombinant JPV lacking the SH protein (rJPV-ΔSH) or replacing SH of JPV with MuV SH (rJPV-MuVSH) or RSV SH (rJPV-RSVSH). rJPV-ΔSH, rJPV-MuVSH, and rJPV-RSVSH were viable and had no growth defect in tissue culture cells. However, more tumor necrosis factor alpha (TNF-α) was produced during rJPV-ΔSH infection, confirming the role of SH in inhibiting TNF-α production. rJPV-ΔSH induced more apoptosis in tissue culture cells than rJPV, rJPV-MuVSH, and rJPV-RSVSH, suggesting that SH plays a role in blocking apoptosis. Furthermore, rJPV-ΔSH was attenuated in mice compared to rJPV, rJPV-MuVSH, and rJPV-RSVSH, indicating that the SH protein plays an essential role in virulence. The results indicate that the functions of MuV SH and RSV SH are similar to that of JPV SH even though they have no sequence homology.IMPORTANCE Paramyxoviruses are associated with many devastating diseases in animals and humans. J paramyxovirus (JPV) was isolated from moribund mice in Australia in 1972. Newly isolated viruses, such as Beilong virus (BeiPV) and Tailam virus (TlmPV), have genome structures similar to that of JPV. A new paramyxovirus genus, Jeilongvirus, which contains JPV, BeiPV, and TlmPV, has been proposed. Small hydrophobic (SH) protein is present in many paramyxoviruses. Our present study investigates the role of SH protein of JPV in pathogenesis in its natural host. Understanding the pathogenic mechanism of Jeilongvirus is important to control and prevent potential diseases that may emerge from this group of viruses.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Infecções por Paramyxoviridae/patologia , Paramyxoviridae/crescimento & desenvolvimento , Proteínas Oncogênicas de Retroviridae/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fatores de Virulência/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Modelos Animais de Doenças , Deleção de Genes , Teste de Complementação Genética , Humanos , Camundongos , Viabilidade Microbiana , Vírus da Caxumba/genética , Vírus da Caxumba/fisiologia , Infecções por Paramyxoviridae/virologia , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/fisiologia , Proteínas Oncogênicas de Retroviridae/genética , Virulência , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA