Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
mSphere ; 8(4): e0008223, 2023 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-37377421

RESUMO

Canine distemper virus (CDV) causes systemic infection resulting in severe and often fatal disease in a large spectrum of animal host species. The virus is closely related to measles virus and targets myeloid, lymphoid, and epithelial cells, but CDV is more virulent and the infection spreads more rapidly within the infected host. Here, we aimed to study the pathogenesis of wild-type CDV infection by experimentally inoculating ferrets with recombinant CDV (rCDV) based on an isolate directly obtained from a naturally infected raccoon. The recombinant virus was engineered to express a fluorescent reporter protein, facilitating assessment of viral tropism and virulence. In ferrets, this wild type-based rCDV infected myeloid, lymphoid, and epithelial cells, and the infection resulted in systemic dissemination to multiple tissues and organs, especially those of the lymphatic system. High infection percentages in immune cells resulted in depletion of these cells both from circulation and from lymphoid tissues. The majority of CDV-infected ferrets reached their humane endpoints within 20 d and had to be euthanized. In that period, the virus also reached the central nervous system in several ferrets, but we did not observe the development of neurological complications during the study period of 23 d. Two out of 14 ferrets survived CDV infection and developed neutralizing antibodies. We show for the first time the pathogenesis of a non-adapted wild type-based rCDV in ferrets. IMPORTANCE Infection of ferrets with recombinant canine distemper virus (rCDV) expressing a fluorescent reporter protein has been used as proxy to understand measles pathogenesis and immune suppression in humans. CDV and measles virus use the same cellular receptors, but CDV is more virulent, and infection is often associated with neurological complications. rCDV strains in current use have complicated passage histories, which may have affected their pathogenesis. Here, we studied the pathogenesis of the first wild type-based rCDV in ferrets. We used macroscopic fluorescence to identify infected cells and tissues; multicolor flow cytometry to determine viral tropism in immune cells; and histopathology and immunohistochemistry to characterize infected cells and lesions in tissues. We conclude that CDV often overwhelmed the immune system, resulting in viral dissemination to multiple tissues in the absence of a detectable neutralizing antibody response. This virus is a promising tool to study the pathogenesis of morbillivirus infections.


Assuntos
Vírus da Cinomose Canina , Cinomose , Humanos , Cães , Animais , Vírus da Cinomose Canina/genética , Furões , Cinomose/patologia , Células Epiteliais/patologia , Vírus do Sarampo/genética , Anticorpos Neutralizantes , Sistema Imunitário/patologia
2.
PLoS Biol ; 21(2): e3001922, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36780432

RESUMO

A universal taxonomy of viruses is essential for a comprehensive view of the virus world and for communicating the complicated evolutionary relationships among viruses. However, there are major differences in the conceptualisation and approaches to virus classification and nomenclature among virologists, clinicians, agronomists, and other interested parties. Here, we provide recommendations to guide the construction of a coherent and comprehensive virus taxonomy, based on expert scientific consensus. Firstly, assignments of viruses should be congruent with the best attainable reconstruction of their evolutionary histories, i.e., taxa should be monophyletic. This fundamental principle for classification of viruses is currently included in the International Committee on Taxonomy of Viruses (ICTV) code only for the rank of species. Secondly, phenotypic and ecological properties of viruses may inform, but not override, evolutionary relatedness in the placement of ranks. Thirdly, alternative classifications that consider phenotypic attributes, such as being vector-borne (e.g., "arboviruses"), infecting a certain type of host (e.g., "mycoviruses," "bacteriophages") or displaying specific pathogenicity (e.g., "human immunodeficiency viruses"), may serve important clinical and regulatory purposes but often create polyphyletic categories that do not reflect evolutionary relationships. Nevertheless, such classifications ought to be maintained if they serve the needs of specific communities or play a practical clinical or regulatory role. However, they should not be considered or called taxonomies. Finally, while an evolution-based framework enables viruses discovered by metagenomics to be incorporated into the ICTV taxonomy, there are essential requirements for quality control of the sequence data used for these assignments. Combined, these four principles will enable future development and expansion of virus taxonomy as the true evolutionary diversity of viruses becomes apparent.


Assuntos
Bacteriófagos , Vírus , Humanos , Metagenômica , Filogenia , Vírus/genética
3.
J Virol ; 97(2): e0008923, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36700640

RESUMO

Viruses have brought humanity many challenges: respiratory infection, cancer, neurological impairment and immunosuppression to name a few. Virology research over the last 60+ years has responded to reduce this disease burden with vaccines and antivirals. Despite this long history, the COVID-19 pandemic has brought unprecedented attention to the field of virology. Some of this attention is focused on concern about the safe conduct of research with human pathogens. A small but vocal group of individuals has seized upon these concerns - conflating legitimate questions about safely conducting virus-related research with uncertainties over the origins of SARS-CoV-2. The result has fueled public confusion and, in many instances, ill-informed condemnation of virology. With this article, we seek to promote a return to rational discourse. We explain the use of gain-of-function approaches in science, discuss the possible origins of SARS-CoV-2 and outline current regulatory structures that provide oversight for virological research in the United States. By offering our expertise, we - a broad group of working virologists - seek to aid policy makers in navigating these controversial issues. Balanced, evidence-based discourse is essential to addressing public concern while maintaining and expanding much-needed research in virology.


Assuntos
Pesquisa , Virologia , Viroses , Humanos , COVID-19/prevenção & controle , Disseminação de Informação , Pandemias/prevenção & controle , Formulação de Políticas , Pesquisa/normas , Pesquisa/tendências , SARS-CoV-2 , Virologia/normas , Virologia/tendências , Viroses/prevenção & controle , Viroses/virologia , Vírus
4.
mBio ; 14(1): e0018823, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36700642

RESUMO

Viruses have brought humanity many challenges: respiratory infection, cancer, neurological impairment and immunosuppression to name a few. Virology research over the last 60+ years has responded to reduce this disease burden with vaccines and antivirals. Despite this long history, the COVID-19 pandemic has brought unprecedented attention to the field of virology. Some of this attention is focused on concern about the safe conduct of research with human pathogens. A small but vocal group of individuals has seized upon these concerns - conflating legitimate questions about safely conducting virus-related research with uncertainties over the origins of SARS-CoV-2. The result has fueled public confusion and, in many instances, ill-informed condemnation of virology. With this article, we seek to promote a return to rational discourse. We explain the use of gain-of-function approaches in science, discuss the possible origins of SARS-CoV-2 and outline current regulatory structures that provide oversight for virological research in the United States. By offering our expertise, we - a broad group of working virologists - seek to aid policy makers in navigating these controversial issues. Balanced, evidence-based discourse is essential to addressing public concern while maintaining and expanding much-needed research in virology.


Assuntos
COVID-19 , Infecções Respiratórias , Vírus , Humanos , COVID-19/prevenção & controle , SARS-CoV-2 , Pandemias/prevenção & controle , Vírus/genética
5.
mSphere ; 8(2): e0003423, 2023 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-36700653

RESUMO

Viruses have brought humanity many challenges: respiratory infection, cancer, neurological impairment and immunosuppression to name a few. Virology research over the last 60+ years has responded to reduce this disease burden with vaccines and antivirals. Despite this long history, the COVID-19 pandemic has brought unprecedented attention to the field of virology. Some of this attention is focused on concern about the safe conduct of research with human pathogens. A small but vocal group of individuals has seized upon these concerns - conflating legitimate questions about safely conducting virus-related research with uncertainties over the origins of SARS-CoV-2. The result has fueled public confusion and, in many instances, ill-informed condemnation of virology. With this article, we seek to promote a return to rational discourse. We explain the use of gain-of-function approaches in science, discuss the possible origins of SARS-CoV-2 and outline current regulatory structures that provide oversight for virological research in the United States. By offering our expertise, we - a broad group of working virologists - seek to aid policy makers in navigating these controversial issues. Balanced, evidence-based discourse is essential to addressing public concern while maintaining and expanding much-needed research in virology.


Assuntos
COVID-19 , Vírus , Humanos , COVID-19/prevenção & controle , SARS-CoV-2 , Pandemias/prevenção & controle , Antivirais
6.
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
7.
J Feline Med Surg ; 21(1): 5-11, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30472918

RESUMO

PRACTICAL RELEVANCE: New technologies capable of sequencing the genetic material in any given biological sample, combined with computer-based algorithms for sequence assembly and analysis, have revolutionised infectious disease research. The rate at which novel viruses are being discovered now exceeds our understanding of their clinical relevance. Novel viruses may contribute to diseases that are major causes of feline morbidity and mortality, including cancer and chronic kidney disease. The identification of new viral pathogens raises the prospect of not only improved patient outcomes through specific treatment but even disease prevention through viral control measures. CLINICAL CHALLENGES: It can be difficult to determine the role of a novel virus in disease development. Disease may be an occasional outcome, often years after infection. A high prevalence of infection in the general population can make disease associations harder to identify and almost impossible to rule out. Host cofactors such as immune dysfunction, genetic background or coinfections may be required for manifestation of disease, and one virus species may be linked to a range of pathological sequelae. Establishing causality relies on evaluating accumulating evidence from multiple investigations, which is often hard to access by practitioners. GLOBAL IMPORTANCE: The worldwide distribution of gammaherpesvirus and morbillivirus infections in domestic cats underlines the potential of these viruses to negatively impact feline health and welfare globally. EVIDENCE BASE: This review relies on grade la-III evidence.


Assuntos
Doenças do Gato/virologia , Infecções por Herpesviridae/veterinária , Infecções por Morbillivirus/veterinária , Animais , Doenças do Gato/diagnóstico , Gatos , Gammaherpesvirinae/genética , Gammaherpesvirinae/patogenicidade , Infecções por Herpesviridae/complicações , Infecções por Herpesviridae/epidemiologia , Morbillivirus/genética , Morbillivirus/patogenicidade , Infecções por Morbillivirus/complicações , Infecções por Morbillivirus/epidemiologia , Filogenia , Prevalência , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/veterinária
8.
mSphere ; 3(3)2018.
Artigo em Inglês | MEDLINE | ID: mdl-29743202

RESUMO

Characterization of human measles cases is essential in order to better assess the data generated in model systems of morbillivirus infection. To this end, we collected formalin-fixed tissue samples from 23 natural measles cases from different areas in the world and different phases of disease ranging from prodromal and acute measles to a persistent infection in an immunocompromised subject. We show that the vast majority of measles virus (MV)-infected cells in epithelia were intraepithelial immune cells that were, in most cases, positive for the CD11c myeloid cell marker. Small numbers of measles virus-infected cytokeratin-positive epithelial cells were also detected in bronchial and appendix epithelia. Dissolution and disruption of uninfected and MV-infected alveolar and bronchial epithelia were prominent features of the measles cases, especially in the established and late phases of the disease. In some instances, this was associated with the formation of MV-infected multinucleated giant cells which expressed CD11c and/or macrophage cell marker 68, a pathological feature also prominently observed in closely associated mucosa-associated lymphoid tissue. Collectively, these data show that resident and inflammatory infiltrating immune cells alter the architecture of respiratory tract epithelia and highlight the necessity for additional research into the function(s) and expression of nectin-4 in human tissues.IMPORTANCE We have brought together a unique collection of 23 human cases of measles infection and studied the types of cells that are infected. This work has not been done with modern technologies such as double labeling with antibodies and confocal microscopy in human cases primarily due to the fact that it is difficult to obtain the material because, fortunately, measles is fatal in only a very small fraction of infected patients. During the past decades, the receptors for measles virus have been elucidated and monkey models have been developed. We found that, in most cases, independently of whether the tissues were obtained early or later in the infection, the primary cell types that were infected were those of the immune system such as lymphocytes, macrophages, and dendritic cells. A very small number of epithelial cells were also found to be infected.


Assuntos
Células Dendríticas/virologia , Células Gigantes/virologia , Macrófagos/virologia , Sarampo/virologia , Morbillivirus/crescimento & desenvolvimento , Mucosa Respiratória/virologia , Adolescente , Idoso , Antígeno CD11c/análise , Criança , Pré-Escolar , Células Dendríticas/química , Feminino , Células Gigantes/química , Humanos , Lactente , Macrófagos/química , Masculino , Sarampo/patologia , Mucosa Respiratória/patologia
9.
Nat Commun ; 8: 16060, 2017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28699636

RESUMO

In 2012, cases of lethal pneumonia among Chinese miners prompted the isolation of a rat-borne henipavirus (HNV), Mòjiang virus (MojV). Although MojV is genetically related to highly pathogenic bat-borne henipaviruses, the absence of a conserved ephrin receptor-binding motif in the MojV attachment glycoprotein (MojV-G) indicates a differing host-cell recognition mechanism. Here we find that MojV-G displays a six-bladed ß-propeller fold bearing limited similarity to known paramyxoviral attachment glycoproteins, in particular at host receptor-binding surfaces. We confirm the inability of MojV-G to interact with known paramyxoviral receptors in vitro, indicating an independence from well-characterized ephrinB2/B3, sialic acid and CD150-mediated entry pathways. Furthermore, we find that MojV-G is antigenically distinct, indicating that MojV would less likely be detected in existing large-scale serological screening studies focused on well-established HNVs. Altogether, these data indicate a unique host-cell entry pathway for this emerging and potentially pathogenic HNV.


Assuntos
Paramyxoviridae/fisiologia , Proteínas Virais de Fusão/fisiologia , Ligação Viral , Animais , Efrina-B2/metabolismo , Efrina-B3/metabolismo , Células HEK293 , Humanos , Camundongos Endogâmicos BALB C , Ácido N-Acetilneuramínico/metabolismo , Paramyxoviridae/química , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Proteínas Virais de Fusão/química
10.
Acta Neuropathol ; 133(1): 139-147, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27770235

RESUMO

Routine childhood vaccination against measles, mumps and rubella has virtually abolished virus-related morbidity and mortality. Notwithstanding this, we describe here devastating neurological complications associated with the detection of live-attenuated mumps virus Jeryl Lynn (MuVJL5) in the brain of a child who had undergone successful allogeneic transplantation for severe combined immunodeficiency (SCID). This is the first confirmed report of MuVJL5 associated with chronic encephalitis and highlights the need to exclude immunodeficient individuals from immunisation with live-attenuated vaccines. The diagnosis was only possible by deep sequencing of the brain biopsy. Sequence comparison of the vaccine batch to the MuVJL5 isolated from brain identified biased hypermutation, particularly in the matrix gene, similar to those found in measles from cases of SSPE. The findings provide unique insights into the pathogenesis of paramyxovirus brain infections.


Assuntos
Encéfalo/virologia , Encefalite Viral/virologia , Vacina contra Caxumba/efeitos adversos , Vírus da Caxumba/isolamento & purificação , Biópsia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Doença Crônica , Encefalite Viral/complicações , Encefalite Viral/diagnóstico por imagem , Encefalite Viral/terapia , Evolução Fatal , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Masculino , Vírus da Caxumba/genética , Imunodeficiência Combinada Severa/complicações , Imunodeficiência Combinada Severa/diagnóstico por imagem , Imunodeficiência Combinada Severa/terapia
11.
Viruses ; 8(7)2016 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-27455307

RESUMO

Effective inactivation of biosafety level 4 (BSL-4) pathogens is vital in order to study these agents safely. Gamma irradiation is a commonly used method for the inactivation of BSL-4 viruses, which among other advantages, facilitates the study of inactivated yet morphologically intact virions. The reported values for susceptibility of viruses to inactivation by gamma irradiation are sometimes inconsistent, likely due to differences in experimental protocols. We analyzed the effects of common sample attributes on the inactivation of a recombinant vesicular stomatitis virus expressing the Zaire ebolavirus glycoprotein and green fluorescent protein. Using this surrogate virus, we found that sample volume and protein content of the sample modulated viral inactivation by gamma irradiation but that air volume within the sample container and the addition of external disinfectant surrounding the sample did not. These data identify several factors which alter viral susceptibility to inactivation and highlight the usefulness of lower biosafety level surrogate viruses for such studies. Our results underscore the need to validate inactivation protocols of BSL-4 pathogens using "worst-case scenario" procedures to ensure complete sample inactivation.


Assuntos
Raios gama , Vesiculovirus/efeitos da radiação , Inativação de Vírus , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas Recombinantes/genética , Vesiculovirus/genética , Proteínas do Envelope Viral/genética
13.
J Virol ; 89(4): 2192-200, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25473055

RESUMO

UNLABELLED: Although live-attenuated measles virus (MV) vaccines have been used successfully for over 50 years, the target cells that sustain virus replication in vivo are still unknown. We generated a reverse genetics system for the live-attenuated MV vaccine strain Edmonston-Zagreb (EZ), allowing recovery of recombinant (r)MV(EZ). Three recombinant viruses were generated that contained the open reading frame encoding enhanced green fluorescent protein (EGFP) within an additional transcriptional unit (ATU) at various positions within the genome. rMV(EZ)EGFP(1), rMV(EZ)EGFP(3), and rMV(EZ)EGFP(6) contained the ATU upstream of the N gene, following the P gene, and following the H gene, respectively. The viruses were compared in vitro by growth curves, which indicated that rMV(EZ)EGFP(1) was overattenuated. Intratracheal infection of cynomolgus macaques with these recombinant viruses revealed differences in immunogenicity. rMV(EZ)EGFP(1) and rMV(EZ)EGFP(6) did not induce satisfactory serum antibody responses, whereas both in vitro and in vivo rMV(EZ)EGFP(3) was functionally equivalent to the commercial MV(EZ)-containing vaccine. Intramuscular vaccination of macaques with rMV(EZ)EGFP(3) resulted in the identification of EGFP(+) cells in the muscle at days 3, 5, and 7 postvaccination. Phenotypic characterization of these cells demonstrated that muscle cells were not infected and that dendritic cells and macrophages were the predominant target cells of live-attenuated MV. IMPORTANCE: Even though MV strain Edmonston-Zagreb has long been used as a live-attenuated vaccine (LAV) to protect against measles, nothing is known about the primary cells in which the virus replicates in vivo. This is vital information given the push to move toward needle-free routes of vaccination, since vaccine virus replication is essential for vaccination efficacy. We have generated a number of recombinant MV strains expressing enhanced green fluorescent protein. The virus that best mimicked the nonrecombinant vaccine virus was formulated according to protocols for production of commercial vaccine virus batches, and was subsequently used to assess viral tropism in nonhuman primates. The virus primarily replicated in professional antigen-presenting cells, which may explain why this LAV is so immunogenic and efficacious.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Macrófagos/imunologia , Macrófagos/virologia , Vacina contra Sarampo/imunologia , Vírus do Sarampo/imunologia , Músculos/imunologia , Animais , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Macaca fascicularis , Masculino , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/genética , Coloração e Rotulagem , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
14.
J Pathol ; 235(2): 242-52, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25229387

RESUMO

Mumps is caused by the mumps virus (MuV), a member of the Paramyxoviridae family of enveloped, non-segmented, negative-sense RNA viruses. Mumps is characterized by painful inflammatory symptoms, such as parotitis and orchitis. The virus is highly neurotropic, with laboratory evidence of central nervous system (CNS) infection in approximately half of cases. Symptomatic CNS infection occurs less frequently; nonetheless, prior to the introduction of routine vaccination, MuV was a leading cause of aseptic meningitis and viral encephalitis in many developed countries. Despite being one of the oldest recognized diseases, with a worldwide distribution, surprisingly little attention has been given to its study. Cases of aseptic meningitis associated with some vaccine strains and a global resurgence of cases, including in highly vaccinated populations, has renewed interest in the virus, particularly in its pathogenesis and the need for development of clinically relevant models of disease. In this review we summarize the current state of knowledge on the virus, its pathogenesis and its clinical and pathological outcomes.


Assuntos
Vírus da Caxumba/patogenicidade , Caxumba/patologia , Caxumba/virologia , Patologia Molecular/métodos , Animais , Biópsia , Modelos Animais de Doenças , Genótipo , Interações Hospedeiro-Patógeno , Humanos , Caxumba/epidemiologia , Caxumba/prevenção & controle , Vacina contra Caxumba/uso terapêutico , Vírus da Caxumba/genética , Valor Preditivo dos Testes , Prognóstico , Virologia/métodos , Virulência
15.
Cell Host Microbe ; 16(1): 19-30, 2014 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-25011105

RESUMO

The cytosolic sensor MDA5 is crucial for antiviral innate immune defense against various RNA viruses including measles virus; as such, many viruses have evolved strategies to antagonize the antiviral activity of MDA5. Here, we show that measles virus escapes MDA5 detection by targeting the phosphatases PP1α and PP1γ, which regulate MDA5 activity by removing an inhibitory phosphorylation mark. The V proteins of measles virus and the related paramyxovirus Nipah virus interact with PP1α/γ, preventing PP1-mediated dephosphorylation of MDA5 and thereby its activation. The PP1 interaction with the measles V protein is mediated by a conserved PP1-binding motif in the C-terminal region of the V protein. A recombinant measles virus expressing a mutant V protein deficient in PP1 binding is unable to antagonize MDA5 and is growth impaired due to its inability to suppress interferon induction. This identifies PP1 antagonism as a mechanism employed by paramyxoviruses for evading innate immune recognition.


Assuntos
RNA Helicases DEAD-box/metabolismo , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Vírus do Sarampo/imunologia , Vírus do Sarampo/fisiologia , Fosfoproteínas/metabolismo , Proteína Fosfatase 1/antagonistas & inibidores , Proteínas Virais/metabolismo , Linhagem Celular , Humanos , Helicase IFIH1 Induzida por Interferon , Vírus Nipah/imunologia , Vírus Nipah/fisiologia , Proteínas Estruturais Virais/metabolismo
16.
Cell Host Microbe ; 16(1): 31-42, 2014 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-25011106

RESUMO

Dendritic cells (DCs) are targets of measles virus (MV) and play central roles in viral dissemination. However, DCs express the RIG-I-like receptors (RLRs) RIG-I and Mda5 that sense MV and induce type I interferon (IFN) production. Given the potency of this antiviral response, RLRs are tightly regulated at various steps, including dephosphorylation by PP1 phosphatases, which induces their activation. We demonstrate that MV suppresses RIG-I and Mda5 by activating the C-type lectin DC-SIGN and inducing signaling that prevents RLR dephosphorylation. MV binding to DC-SIGN leads to activation of the kinase Raf-1, which induces the association of PP1 inhibitor I-1 with GADD34-PP1 holoenzymes, thereby inhibiting phosphatase activity. Consequently, GADD34-PP1 holoenzymes are unable to dephosphorylate RIG-I and Mda5, hence suppressing type I IFN responses and enhancing MV replication. Blocking DC-SIGN signaling allows RLR activation and suppresses MV infection of DCs. Thus, MV subverts DC-SIGN to control RLR activation and escape antiviral responses.


Assuntos
Moléculas de Adesão Celular/metabolismo , RNA Helicases DEAD-box/metabolismo , Células Dendríticas/imunologia , Interações Hospedeiro-Patógeno , Lectinas Tipo C/metabolismo , Vírus do Sarampo/imunologia , Proteína Fosfatase 1/antagonistas & inibidores , Receptores de Superfície Celular/metabolismo , Linhagem Celular , Proteína DEAD-box 58 , Células Dendríticas/virologia , Humanos , Evasão da Resposta Imune , Vírus do Sarampo/fisiologia , Receptores Imunológicos
17.
J Gen Virol ; 94(Pt 9): 1933-1944, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23784446

RESUMO

Measles virus (MV), a member of the family Paramyxoviridae, remains a major cause of morbidity and mortality in the developing world. MV is spread by aerosols but the mechanism(s) responsible for the high transmissibility of MV are largely unknown. We previously infected macaques with enhanced green fluorescent protein-expressing recombinant MV and euthanized them at a range of time points. In this study a comprehensive pathological analysis has been performed of tissues from the respiratory tract around the peak of virus replication. Isolation of virus from nose and throat swab samples showed that high levels of both cell-associated and cell-free virus were present in the upper respiratory tract. Analysis of tissue sections from lung and primary bronchus revealed localized infection of epithelial cells, concomitant infiltration of MV-infected immune cells into the epithelium and localized shedding of cells or cell debris into the lumen. While high numbers of MV-infected cells were present in the tongue, these were largely encapsulated by intact keratinocyte cell layers that likely limit virus transmission. In contrast, the integrity of tonsillar and adenoidal epithelia was disrupted with high numbers of MV-infected epithelial cells and infiltrating immune cells present throughout epithelial cell layers. Disruption was associated with large numbers of MV-infected cells or cell debris 'spilling' from epithelia into the respiratory tract. The coughing and sneezing response induced by disruption of the ciliated epithelium, leading to the expulsion of MV-infected cells, cell debris and cell-free virus, contributes to the highly infectious nature of MV.


Assuntos
Vírus do Sarampo/patogenicidade , Sarampo/virologia , Infecções Respiratórias/virologia , Animais , Modelos Animais de Doenças , Tecido Linfoide/virologia , Macaca , Sarampo/patologia , Vírus do Sarampo/isolamento & purificação , Mucosa Respiratória/virologia , Sistema Respiratório/patologia , Sistema Respiratório/virologia , Infecções Respiratórias/patologia , Carga Viral
18.
Curr Opin Virol ; 2(3): 248-55, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22483507

RESUMO

Measles is an important cause of childhood morbidity and mortality in developing countries. Measles virus (MV) is transmitted via the respiratory route and causes systemic disease. Over the last decade, identification of new cellular receptors and studies in animal models have challenged the historic concepts of measles pathogenesis. It is thought that MV enters the host by infection of alveolar macrophages and/or dendritic cells in the airways, and is amplified in local lymphoid tissues. Viremia mediated by infected CD150+ lymphocytes results in systemic dissemination. Infection of lymphocytes and dendritic cells in the respiratory submucosa facilitates basolateral infection of epithelial cells via the newly identified receptor Nectin-4. Concomitant and extensive epithelial damage may contribute to efficient transmission to the next host.


Assuntos
Vírus do Sarampo/patogenicidade , Sarampo/patologia , Sarampo/virologia , Animais , Células Dendríticas/virologia , Modelos Animais de Doenças , Células Epiteliais/virologia , Humanos , Linfonodos/virologia , Linfócitos/virologia , Macrófagos/virologia , Receptores Virais/metabolismo , Sistema Respiratório/virologia , Viremia , Internalização do Vírus
19.
Biosci Rep ; 32(3): 333-43, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22385277

RESUMO

Protein interactions play key roles throughout all subcellular compartments. In the present paper, we report the visualization of protein interactions throughout living mammalian cells using two oligomerizing MV (measles virus) transmembrane glycoproteins, the H (haemagglutinin) and the F (fusion) glycoproteins, which mediate MV entry into permissive cells. BiFC (bimolecular fluorescence complementation) has been used to examine the dimerization of these viral glycoproteins. The H glycoprotein is a type II membrane-receptor-binding homodimeric glycoprotein and the F glycoprotein is a type I disulfide-linked membrane glycoprotein which homotrimerizes. Together they co-operate to allow the enveloped virus to enter a cell by fusing the viral and cellular membranes. We generated a pair of chimaeric H glycoproteins linked to complementary fragments of EGFP (enhanced green fluorescent protein)--haptoEGFPs--which, on association, generate fluorescence. Homodimerization of H glycoproteins specifically drives this association, leading to the generation of a fluorescent signal in the ER (endoplasmic reticulum), the Golgi and at the plasma membrane. Similarly, the generation of a pair of corresponding F glycoprotein-haptoEGFP chimaeras also produced a comparable fluorescent signal. Co-expression of H and F glycoprotein chimaeras linked to complementary haptoEGFPs led to the formation of fluorescent fusion complexes at the cell surface which retained their biological activity as evidenced by cell-to-cell fusion.


Assuntos
Proteínas de Fluorescência Verde/biossíntese , Fusão de Membrana , Proteínas de Membrana/metabolismo , Mapeamento de Interação de Proteínas/métodos , Multimerização Proteica , Animais , Chlorocebus aethiops , Proteínas de Fluorescência Verde/genética , Microscopia Confocal , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/genética , Ligação Proteica , Proteínas Recombinantes de Fusão/biossíntese , Análise de Célula Única , Células Vero , Proteínas Virais de Fusão/biossíntese
20.
Virus Res ; 162(1-2): 47-62, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22001568

RESUMO

This review focuses on new concepts important for the understanding of the pathogenesis of measles virus. First the requirement for specific entry receptors restricts the cell types that measles can enter during the initial stages of infection in the human host. Recently, the paradigm for measles has shifted from an epithelial infection similar to that caused in the respiratory tract by other members of the paramyxoviruses to one which displays more similarity to the infection of the immune system by HIV-1, though the route of infection is different. Secondly we review the role of host proteins that support viral replication as well as those that modify the cellular environment in order to promote measles virus replication. The role of specific virus proteins in the anti-antiviral response is also reviewed. Measles virus counteracts all pathways known to induce interferon synthesis as well as signalling by interferons, exemplifying the importance of these in the virulence/attenuation of the virus. We conclude that only studies in relevant animal model systems or humans or in vitro or ex vivo studies of relevant cell types and tissues will bring us closer to an understanding of the pathogenesis of the virus, factors that have often been overlooked in past studies.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Evasão da Resposta Imune , Imunidade Inata , Interferons/imunologia , Vírus do Sarampo/imunologia , Sarampo , Proteínas Virais , Adenosina Desaminase/genética , Adenosina Desaminase/imunologia , Adenosina Desaminase/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/imunologia , Proteínas de Ciclo Celular/metabolismo , Humanos , Interferons/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Sarampo/imunologia , Sarampo/virologia , Vírus do Sarampo/metabolismo , Vírus do Sarampo/patogenicidade , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas de Ligação a RNA , Receptores Virais/genética , Receptores Virais/imunologia , Receptores Virais/metabolismo , Transdução de Sinais/imunologia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Proteínas Virais/genética , Proteínas Virais/imunologia , Proteínas Virais/metabolismo , Replicação Viral/genética , Replicação Viral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA