Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 875
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
PLoS Pathog ; 20(9): e1012569, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39283943

RESUMO

Activation of the DNA-sensing STING axis by RNA viruses plays a role in antiviral response through mechanisms that remain poorly understood. Here, we show that the STING pathway regulates Nipah virus (NiV) replication in vivo in mice. Moreover, we demonstrate that following both NiV and measles virus (MeV) infection, IFNγ-inducible protein 16 (IFI16), an alternative DNA sensor in addition to cGAS, induces the activation of STING, leading to the phosphorylation of NF-κB p65 and the production of IFNß and interleukin 6. Finally, we found that paramyxovirus-induced syncytia formation is responsible for loss of mitochondrial membrane potential and leakage of mitochondrial DNA in the cytoplasm, the latter of which is further detected by both cGAS and IFI16. These results contribute to improve our understanding about NiV and MeV immunopathogenesis and provide potential paths for alternative therapeutic strategies.


Assuntos
Células Gigantes , Vírus do Sarampo , Proteínas de Membrana , Vírus Nipah , Animais , Vírus do Sarampo/fisiologia , Camundongos , Células Gigantes/virologia , Células Gigantes/metabolismo , Proteínas de Membrana/metabolismo , Vírus Nipah/fisiologia , Sarampo/virologia , Sarampo/metabolismo , Sarampo/imunologia , Humanos , Replicação Viral/fisiologia , Infecções por Henipavirus/virologia , Infecções por Henipavirus/metabolismo , Infecções por Henipavirus/imunologia , Fosfoproteínas/metabolismo , Proteínas Nucleares/metabolismo , Camundongos Endogâmicos C57BL
2.
J Virol ; 98(8): e0075824, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39041786

RESUMO

Measles is a highly transmissible systemic viral infection associated with substantial mortality primarily due to secondary infections. Measles induces lifelong immunity to reinfection but loss of immunity to other pathogens. An attenuated live virus vaccine is highly effective, but lapses in delivery have resulted in increasing cases worldwide. Although the primary cause of failure to control measles is failure to vaccinate, waning vaccine-induced immunity and the possible emergence of more virulent virus strains may also contribute.


Assuntos
Vacina contra Sarampo , Vírus do Sarampo , Sarampo , Sarampo/prevenção & controle , Sarampo/imunologia , Sarampo/virologia , Humanos , Vacina contra Sarampo/imunologia , Vírus do Sarampo/imunologia , Vacinação , Vacinas Atenuadas/imunologia
3.
J Virol ; 98(9): e0102024, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39194235

RESUMO

Some negative-sense RNA viruses, including measles virus (MeV), share the characteristic that during their infection cycle, cytoplasmic inclusion bodies (IBs) are formed where components of the viral replication machinery are concentrated. As a foci of viral replication, how IBs act to enhance the efficiency of infection by affecting virus-host interactions remains an important topic of investigation. We previously established that upon MeV infection, the epigenetic host protein, WD repeat-containing protein 5 (WDR5), translocates to cytoplasmic viral IBs and facilitates MeV replication. We now show that WDR5 is recruited to IBs by forming a complex with IB-associated MeV phosphoprotein via a conserved binding motif located on the surface of WDR5. Furthermore, we provide evidence that WDR5 promotes viral replication by suppressing a major innate immune response pathway, the double-stranded RNA-mediated activation of protein kinase R and integrated stress response. IMPORTANCE: MeV is a pathogen that remains a global concern, with an estimated 9 million measles cases and 128,000 measles deaths in 2022 according to the World Health Organization. A large population of the world still has inadequate access to the effective vaccine against the exceptionally transmissible MeV. Measles disease is characterized by a high morbidity in children and in immunocompromised individuals. An important area of research for negative-sense RNA viruses, including MeV, is the characterization of the complex interactome between virus and host occurring at cytoplasmic IBs where viral replication occurs. Despite the progress made in understanding IB structures, little is known regarding the virus-host interactions within IBs and the role of these interactions in promoting viral replication and antagonizing host innate immunity. Herein we provide evidence suggesting a model by which MeV IBs utilize the host protein WDR5 to suppress the protein kinase R-integrated stress response pathway.


Assuntos
Imunidade Inata , Vírus do Sarampo , Sarampo , Replicação Viral , Vírus do Sarampo/fisiologia , Vírus do Sarampo/genética , Humanos , Sarampo/virologia , Sarampo/metabolismo , Corpos de Inclusão Viral/metabolismo , Interações Hospedeiro-Patógeno , eIF-2 Quinase/metabolismo , eIF-2 Quinase/genética , Células HEK293 , Estresse Fisiológico , RNA de Cadeia Dupla/metabolismo , Proteínas Virais/metabolismo , Proteínas Virais/genética , Animais
4.
J Virol ; 98(3): e0187423, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38329336

RESUMO

Subacute sclerosing panencephalitis (SSPE) is a rare but fatal late neurological complication of measles, caused by persistent measles virus (MeV) infection of the central nervous system. There are no drugs approved for the treatment of SSPE. Here, we followed the clinical progression of a 5-year-old SSPE patient after treatment with the nucleoside analog remdesivir, conducted a post-mortem evaluation of the patient's brain, and characterized the MeV detected in the brain. The quality of life of the patient transiently improved after the first two courses of remdesivir, but a third course had no further clinical effect, and the patient eventually succumbed to his condition. Post-mortem evaluation of the brain displayed histopathological changes including loss of neurons and demyelination paired with abundant presence of MeV RNA-positive cells throughout the brain. Next-generation sequencing of RNA isolated from the brain revealed a complete MeV genome with mutations that are typically detected in SSPE, characterized by a hypermutated M gene. Additional mutations were detected in the polymerase (L) gene, which were not associated with resistance to remdesivir. Functional characterization showed that mutations in the F gene led to a hyperfusogenic phenotype predominantly mediated by N465I. Additionally, recombinant wild-type-based MeV with the SSPE-F gene or the F gene with the N465I mutation was no longer lymphotropic but instead efficiently disseminated in neural cultures. Altogether, this case encourages further investigation of remdesivir as a potential treatment of SSPE and highlights the necessity to functionally understand SSPE-causing MeV.IMPORTANCEMeasles virus (MeV) causes acute, systemic disease and remains an important cause of morbidity and mortality in humans. Despite the lack of known entry receptors in the brain, MeV can persistently infect the brain causing the rare but fatal neurological disorder subacute sclerosing panencephalitis (SSPE). SSPE-causing MeVs are characterized by a hypermutated genome and a hyperfusogenic F protein that facilitates the rapid spread of MeV throughout the brain. No treatment against SSPE is available, but the nucleoside analog remdesivir was recently demonstrated to be effective against MeV in vitro. We show that treatment of an SSPE patient with remdesivir led to transient clinical improvement and did not induce viral escape mutants, encouraging the future use of remdesivir in SSPE patients. Functional characterization of the viral proteins sheds light on the shared properties of SSPE-causing MeVs and further contributes to understanding how those viruses cause disease.


Assuntos
Monofosfato de Adenosina , Alanina , Vírus do Sarampo , Sarampo , Panencefalite Esclerosante Subaguda , Proteínas Virais , Pré-Escolar , Humanos , Monofosfato de Adenosina/administração & dosagem , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/uso terapêutico , Alanina/administração & dosagem , Alanina/análogos & derivados , Alanina/uso terapêutico , Autopsia , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/virologia , Progressão da Doença , Evolução Fatal , Genoma Viral/genética , Sequenciamento de Nucleotídeos em Larga Escala , Sarampo/complicações , Sarampo/tratamento farmacológico , Sarampo/virologia , Vírus do Sarampo/efeitos dos fármacos , Vírus do Sarampo/genética , Vírus do Sarampo/metabolismo , Proteínas Mutantes/análise , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Qualidade de Vida , RNA Viral/análise , RNA Viral/genética , Panencefalite Esclerosante Subaguda/tratamento farmacológico , Panencefalite Esclerosante Subaguda/etiologia , Panencefalite Esclerosante Subaguda/virologia , Proteínas Virais/análise , Proteínas Virais/genética , Proteínas Virais/metabolismo
5.
Emerg Infect Dis ; 30(5): 926-933, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38579738

RESUMO

We investigated clinically suspected measles cases that had discrepant real-time reverse transcription PCR (rRT-PCR) and measles-specific IgM test results to determine diagnoses. We performed rRT-PCR and measles-specific IgM testing on samples from 541 suspected measles cases. Of the 24 IgM-positive and rRT-PCR--negative cases, 20 were among children who received a measles-containing vaccine within the previous 6 months; most had low IgG relative avidity indexes (RAIs). The other 4 cases were among adults who had an unknown previous measles history, unknown vaccination status, and high RAIs. We detected viral nucleic acid for viruses other than measles in 15 (62.5%) of the 24 cases with discrepant rRT-PCR and IgM test results. Measles vaccination, measles history, and contact history should be considered in suspected measles cases with discrepant rRT-PCR and IgM test results. If in doubt, measles IgG avidity and PCR testing for other febrile exanthematous viruses can help confirm or refute the diagnosis.


Assuntos
Anticorpos Antivirais , Imunoglobulina M , Vírus do Sarampo , Sarampo , Humanos , Imunoglobulina M/sangue , Sarampo/diagnóstico , Sarampo/epidemiologia , Sarampo/virologia , Sarampo/imunologia , Anticorpos Antivirais/sangue , Japão/epidemiologia , Criança , Pré-Escolar , Vírus do Sarampo/imunologia , Vírus do Sarampo/genética , Masculino , Adulto , Feminino , Lactente , Adolescente , Imunoglobulina G/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Vacina contra Sarampo/imunologia , Adulto Jovem , Reação em Cadeia da Polimerase em Tempo Real/métodos
6.
J Virol ; 97(10): e0105123, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37732787

RESUMO

IMPORTANCE: For many years, measles virus (MeV) was assumed to first enter the host via the apical surface of airway epithelial cells and subsequently spread systemically. We and others reported that MeV has an overwhelming preference for entry at the basolateral surface of airway epithelial cells, which led to a fundamental new understanding of how MeV enters a human host. This unexpected observation using well-differentiated primary cultures of airway epithelia from human donors contradicted previous studies using immortalized cultured cells. Here, we show that appropriate differentiation and cell morphology of primary human airway epithelial cells are critical to recapitulate MeV infection patterns and pathogenesis of the in vivo airways. By simply culturing primary cells in media containing serum or passaging primary cultures, erroneous results quickly emerge. These results have broad implications for data interpretation related to respiratory virus infection, spread, and release from human airway epithelial cells.


Assuntos
Células Cultivadas , Células Epiteliais , Vírus do Sarampo , Sarampo , Sistema Respiratório , Humanos , Células Epiteliais/virologia , Epitélio , Sarampo/virologia , Sistema Respiratório/citologia
7.
J Med Virol ; 96(9): e29892, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39210621

RESUMO

In line with European trends, since 2023 Lombardy (Northern Italy) is experiencing a resurgence of measles and an increased number of reported cases of fever and rash. Measles discarded cases observed in our region within the context of measles and rubella surveillance from the first few months of 2024 (N = 30) were investigated for parvovirus B19 (B19V) and other rash-associated viruses. Thirteen cases tested positive for B19V DNA, representing a significant increase from previous years (on average 3 cases per year, p < 0.001) and ~40% of all B19V DNA-positive patients we detected since 2017. In 2024, B19V DNA-positive subjects spanned all ages, and the virus was predominant among adolescents and adults (84.6%). Two B19V infected patients were hospitalised, and likely cross-reacting anti-measles virus IgM were found in both. Our data align with the recent reports from the ECDC and various European countries, which are experiencing a surge in B19V infections, and underline the importance of comprehensive measles and rubella surveillance systems that can adapt to changing epidemiological trends.


Assuntos
Sarampo , Parvovirus B19 Humano , Rubéola (Sarampo Alemão) , Humanos , Itália/epidemiologia , Parvovirus B19 Humano/genética , Parvovirus B19 Humano/isolamento & purificação , Parvovirus B19 Humano/imunologia , Sarampo/epidemiologia , Sarampo/diagnóstico , Sarampo/virologia , Rubéola (Sarampo Alemão)/epidemiologia , Rubéola (Sarampo Alemão)/diagnóstico , Rubéola (Sarampo Alemão)/virologia , Feminino , Masculino , Adulto , Adolescente , Adulto Jovem , Criança , Diagnóstico Diferencial , Pré-Escolar , Infecções por Parvoviridae/epidemiologia , Infecções por Parvoviridae/diagnóstico , Anticorpos Antivirais/sangue , Pessoa de Meia-Idade , Lactente , DNA Viral/genética , Imunoglobulina M/sangue
8.
Bull Math Biol ; 86(7): 85, 2024 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-38853189

RESUMO

How viral infections develop can change based on the number of viruses initially entering the body. The understanding of the impacts of infection doses remains incomplete, in part due to challenging constraints, and a lack of research. Gaining more insights is crucial regarding the measles virus (MV). The higher the MV infection dose, the earlier the peak of acute viremia, but the magnitude of the peak viremia remains almost constant. Measles is highly contagious, causes immunosuppression such as lymphopenia, and contributes substantially to childhood morbidity and mortality. This work investigated mechanisms underlying the observed wild-type measles infection dose responses in cynomolgus monkeys. We fitted longitudinal data on viremia using maximum likelihood estimation, and used the Akaike Information Criterion (AIC) to evaluate relevant biological hypotheses and their respective model parameterizations. The lowest AIC indicates a linear relationship between the infection dose, the initial viral load, and the initial number of activated MV-specific T cells. Early peak viremia is associated with high initial number of activated MV-specific T cells. Thus, when MV infection dose increases, the initial viremia and associated immune cell stimulation increase, and reduce the time it takes for T cell killing to be sufficient, thereby allowing dose-independent peaks for viremia, MV-specific T cells, and lymphocyte depletion. Together, these results suggest that the development of measles depends on virus-host interactions at the start and the efficiency of viral control by cellular immunity. These relationships are additional motivations for prevention, vaccination, and early treatment for measles.


Assuntos
Macaca fascicularis , Conceitos Matemáticos , Vírus do Sarampo , Sarampo , Carga Viral , Viremia , Sarampo/imunologia , Sarampo/transmissão , Sarampo/prevenção & controle , Sarampo/virologia , Sarampo/epidemiologia , Animais , Viremia/imunologia , Viremia/virologia , Vírus do Sarampo/imunologia , Vírus do Sarampo/patogenicidade , Vírus do Sarampo/fisiologia , Funções Verossimilhança , Humanos , Modelos Imunológicos , Modelos Biológicos , Linfócitos T/imunologia , Ativação Linfocitária
9.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33903248

RESUMO

Measles virus (MeV) is resurgent and caused >200,000 deaths in 2019. MeV infection can establish a chronic latent infection of the brain that can recrudesce months to years after recovery from the primary infection. Recrudescent MeV leads to fatal subacute sclerosing panencephalitis (SSPE) or measles inclusion body encephalitis (MIBE) as the virus spreads across multiple brain regions. Most clinical isolates of SSPE/MIBE strains show mutations in the fusion (F) gene that result in a hyperfusogenic phenotype in vitro and allow for efficient spread in primary human neurons. Wild-type MeV receptor-binding protein is indispensable for manifesting these mutant F phenotypes, even though neurons lack canonical MeV receptors (CD150/SLAMF1 or nectin-4). How such hyperfusogenic F mutants are selected and whether they confer a fitness advantage for efficient neuronal spread is unresolved. To better understand the fitness landscape that allows for the selection of such hyperfusogenic F mutants, we conducted a screen of ≥3.1 × 105 MeV-F point mutants in their genomic context. We rescued and amplified our genomic MeV-F mutant libraries in BSR-T7 cells under conditions in which MeV-F-T461I (a known SSPE mutant), but not wild-type MeV, can spread. We recovered known SSPE mutants but also characterized at least 15 hyperfusogenic F mutations with an SSPE phenotype. Structural mapping of these mutants onto the prefusion MeV-F trimer confirm and extend our understanding of the F regulatory domains in MeV-F. Our list of hyperfusogenic F mutants is a valuable resource for future studies into MeV neuropathogenesis and the regulation of paramyxovirus F.


Assuntos
Vírus do Sarampo/genética , Sarampo/genética , Panencefalite Esclerosante Subaguda/genética , Proteínas Virais de Fusão/genética , Substituição de Aminoácidos/genética , Animais , Encéfalo/patologia , Encéfalo/virologia , Chlorocebus aethiops , Humanos , Sarampo/patologia , Sarampo/virologia , Vírus do Sarampo/patogenicidade , Mutação/genética , Neurônios/patologia , Neurônios/virologia , Panencefalite Esclerosante Subaguda/patologia , Panencefalite Esclerosante Subaguda/virologia , Células Vero
10.
Euro Surveill ; 29(28)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38994600

RESUMO

We investigated a variant of measles virus that encodes three mismatches to the reverse priming site for a widely used diagnostic real-time RT-PCR assay; reduction of sensitivity was hypothesised. We examined performance of the assay in context of the variant using in silico data, synthetic RNA templates and clinical specimens. Sensitivity was reduced observed at low copy numbers for templates encoding the variant sequence. We designed and tested an alternate priming strategy, rescuing the sensitivity of the assay.


Assuntos
Vírus do Sarampo , Sarampo , RNA Viral , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade , Humanos , Sarampo/diagnóstico , Sarampo/virologia , Vírus do Sarampo/genética , Vírus do Sarampo/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , RNA Viral/genética
11.
Lancet ; 399(10325): 678-690, 2022 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-35093206

RESUMO

Measles is a highly contagious, potentially fatal, but vaccine-preventable disease caused by measles virus. Symptoms include fever, maculopapular rash, and at least one of cough, coryza, or conjunctivitis, although vaccinated individuals can have milder or even no symptoms. Laboratory diagnosis relies largely on the detection of specific IgM antibodies in serum, dried blood spots, or oral fluid, or the detection of viral RNA in throat or nasopharyngeal swabs, urine, or oral fluid. Complications can affect many organs and often include otitis media, laryngotracheobronchitis, pneumonia, stomatitis, and diarrhoea. Neurological complications are uncommon but serious, and can occur during or soon after the acute disease (eg, acute disseminated encephalomyelitis) or months or even years later (eg, measles inclusion body encephalitis and subacute sclerosing panencephalitis). Patient management mainly involves supportive therapy, such as vitamin A supplementation, monitoring for and treatment of secondary bacterial infections with antibiotics, and rehydration in the case of severe diarrhoea. There is no specific antiviral therapy for the treatment of measles, and disease control largely depends on prevention. However, despite the availability of a safe and effective vaccine, measles is still endemic in many countries and causes considerable morbidity and mortality, especially among children in resource-poor settings. The low case numbers reported in 2020, after a worldwide resurgence of measles between 2017 and 2019, have to be interpreted cautiously, owing to the effect of the COVID-19 pandemic on disease surveillance. Disrupted vaccination activities during the pandemic increase the potential for another resurgence of measles in the near future, and effective, timely catch-up vaccination campaigns, strong commitment and leadership, and sufficient resources will be required to mitigate this threat.


Assuntos
COVID-19/epidemiologia , Doenças Endêmicas/prevenção & controle , Vacinação em Massa/organização & administração , Vacina contra Sarampo/administração & dosagem , Sarampo/prevenção & controle , COVID-19/prevenção & controle , Controle de Doenças Transmissíveis/organização & administração , Controle de Doenças Transmissíveis/normas , Doenças Endêmicas/estatística & dados numéricos , Humanos , Vacinação em Massa/normas , Vacinação em Massa/estatística & dados numéricos , Sarampo/epidemiologia , Sarampo/imunologia , Sarampo/virologia , Vírus do Sarampo/imunologia , Vírus do Sarampo/patogenicidade , Pandemias/prevenção & controle
12.
J Virol ; 96(3): e0194921, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34788082

RESUMO

Measles virus (MeV), an enveloped RNA virus in the family Paramyxoviridae, usually causes acute febrile illness with skin rash but in rare cases persists in the brain, causing a progressive neurological disorder, subacute sclerosing panencephalitis (SSPE). MeV bears two envelope glycoproteins, the hemagglutinin (H) and fusion (F) proteins. The H protein possesses a head domain that initially mediates receptor binding and a stalk domain that subsequently transmits the fusion-triggering signal to the F protein. We recently showed that cell adhesion molecule 1 (CADM1; also known as IGSF4A, Necl-2, and SynCAM1) and CADM2 (also known as IGSF4D, Necl-3, and SynCAM2) are host factors enabling cell-cell membrane fusion mediated by hyperfusogenic F proteins of neuropathogenic MeVs as well as MeV spread between neurons lacking the known receptors. CADM1 and CADM2 interact in cis with the H protein on the same cell membrane, triggering hyperfusogenic F protein-mediated membrane fusion. Multiple isoforms of CADM1 and CADM2 containing various lengths of their stalk regions are generated by alternative splicing. Here, we show that only short-stalk isoforms of CADM1 and CADM2 predominantly expressed in the brain induce hyperfusogenic F protein-mediated membrane fusion. While the known receptors interact in trans with the H protein through its head domain, these isoforms can interact in cis even with the H protein lacking the head domain and trigger membrane fusion, presumably through its stalk domain. Thus, our results unveil a new mechanism of viral fusion triggering by host factors. IMPORTANCE Measles, an acute febrile illness with skin rash, is still an important cause of childhood morbidity and mortality worldwide. Measles virus (MeV), the causative agent of measles, may also cause a progressive neurological disorder, subacute sclerosing panencephalitis (SSPE), several years after acute infection. The disease is fatal, and no effective therapy is available. Recently, we reported that cell adhesion molecule 1 (CADM1) and CADM2 are host factors enabling MeV cell-to-cell spread in neurons. These molecules interact in cis with the MeV attachment protein on the same cell membrane, triggering the fusion protein and causing membrane fusion. CADM1 and CADM2 are known to exist in multiple splice isoforms. In this study, we report that their short-stalk isoforms can induce membrane fusion by interacting in cis with the viral attachment protein independently of its receptor-binding head domain. This finding may have important implications for cis-acting fusion triggering by host factors.


Assuntos
Molécula 1 de Adesão Celular/metabolismo , Células Gigantes/virologia , Hemaglutininas Virais/metabolismo , Interações Hospedeiro-Patógeno , Vírus do Sarampo/fisiologia , Sarampo/metabolismo , Sarampo/virologia , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Molécula 1 de Adesão Celular/genética , Células Cultivadas , Cricetinae , Modelos Biológicos , Ligação Proteica , Isoformas de Proteínas , Proteínas Virais de Fusão/metabolismo
13.
J Virol ; 96(22): e0131922, 2022 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-36300942

RESUMO

Many negative-sense RNA viruses, including measles virus (MeV), are thought to carry out much of their viral replication in cytoplasmic membraneless foci known as inclusion bodies (IBs). The mechanisms by which IBs facilitate efficient viral replication remain largely unknown but may involve an intricate network of regulation at the host-virus interface. Viruses are able to modulate such interactions by a variety of strategies including adaptation of their genomes and "hijacking" of host proteins. The latter possibility broadens the molecular reservoir available for a virus to enhance its replication and/or antagonize host antiviral responses. Here, we show that the cellular 5'-3' exoribonuclease, XRN1, is a host protein hijacked by MeV. We found that upon MeV infection, XRN1 is translocated to cytoplasmic IBs where it acts in a proviral manner by preventing the accumulation of double-stranded RNA (dsRNA) within the IBs. This leads to the suppression of the dsRNA-induced innate immune responses mediated via the protein kinase R (PKR)-integrated stress response (ISR) pathway. IMPORTANCE Measles virus remains a major global health threat due to its high transmissibility and significant morbidity in children and immunocompromised individuals. Although there is an effective vaccine against MeV, a large population in the world remains without access to the vaccine, contributing to more than 7,000,000 measles cases and 60,000 measles deaths in 2020 (CDC). For negative-sense RNA viruses including MeV, one active research area is the exploration of virus-host interactions occurring at cytoplasmic IBs where viral replication takes place. In this study we present evidence suggesting a model in which MeV IBs antagonize host innate immunity by recruiting XRN1 to reduce dsRNA accumulation and subsequent PKR kinase activation/ISR induction. In the absence of XRN1, the increased dsRNA level acts as a potent activator of the antiviral PKR/ISR pathway leading to suppression of global cap-dependent mRNA translation and inhibition of viral replication.


Assuntos
Exorribonucleases , Sarampo , Proteínas Associadas aos Microtúbulos , Replicação Viral , Humanos , eIF-2 Quinase/metabolismo , Exorribonucleases/genética , Exorribonucleases/metabolismo , Sarampo/genética , Sarampo/virologia , Vírus do Sarampo/genética , Vírus do Sarampo/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Quinases/metabolismo , Provírus/genética , RNA de Cadeia Dupla , Corpos de Inclusão Viral
14.
PLoS Pathog ; 17(8): e1009458, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34383863

RESUMO

Measles virus (MeV) is the most contagious human virus. Unlike most respiratory viruses, MeV does not directly infect epithelial cells upon entry in a new host. MeV traverses the epithelium within immune cells that carry it to lymphatic organs where amplification occurs. Infected immune cells then synchronously deliver large amounts of virus to the airways. However, our understanding of MeV replication in airway epithelia is limited. To model it, we use well-differentiated primary cultures of human airway epithelial cells (HAE) from lung donors. In HAE, MeV spreads directly cell-to-cell forming infectious centers that grow for ~3-5 days, are stable for a few days, and then disappear. Transepithelial electrical resistance remains intact during the entire course of HAE infection, thus we hypothesized that MeV infectious centers may dislodge while epithelial function is preserved. After documenting by confocal microscopy that infectious centers progressively detach from HAE, we recovered apical washes and separated cell-associated from cell-free virus by centrifugation. Virus titers were about 10 times higher in the cell-associated fraction than in the supernatant. In dislodged infectious centers, ciliary beating persisted, and apoptotic markers were not readily detected, suggesting that they retain functional metabolism. Cell-associated MeV infected primary human monocyte-derived macrophages, which models the first stage of infection in a new host. Single-cell RNA sequencing identified wound healing, cell growth, and cell differentiation as biological processes relevant for infectious center dislodging. 5-ethynyl-2'-deoxyuridine (EdU) staining located proliferating cells underneath infectious centers. Thus, cells located below infectious centers divide and differentiate to repair the dislodged infected epithelial patch. As an extension of these studies, we postulate that expulsion of infectious centers through coughing and sneezing could contribute to MeV's strikingly high reproductive number by allowing the virus to survive longer in the environment and by delivering a high infectious dose to the next host.


Assuntos
Células Epiteliais/virologia , Macrófagos/virologia , Vírus do Sarampo/patogenicidade , Sarampo/virologia , Sistema Respiratório/virologia , Internalização do Vírus , Replicação Viral , Células Cultivadas , Células Epiteliais/metabolismo , Humanos , Macrófagos/metabolismo , Sarampo/genética , Sarampo/metabolismo , RNA-Seq , Sistema Respiratório/metabolismo , Análise de Célula Única , Transcriptoma
15.
PLoS Pathog ; 17(2): e1009371, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33621266

RESUMO

Morbilliviruses, such as measles virus (MeV) and canine distemper virus (CDV), are highly infectious members of the paramyxovirus family. MeV is responsible for major morbidity and mortality in non-vaccinated populations. ERDRP-0519, a pan-morbillivirus small molecule inhibitor for the treatment of measles, targets the morbillivirus RNA-dependent RNA-polymerase (RdRP) complex and displayed unparalleled oral efficacy against lethal infection of ferrets with CDV, an established surrogate model for human measles. Resistance profiling identified the L subunit of the RdRP, which harbors all enzymatic activity of the polymerase complex, as the molecular target of inhibition. Here, we examined binding characteristics, physical docking site, and the molecular mechanism of action of ERDRP-0519 through label-free biolayer interferometry, photoaffinity cross-linking, and in vitro RdRP assays using purified MeV RdRP complexes and synthetic templates. Results demonstrate that unlike all other mononegavirus small molecule inhibitors identified to date, ERDRP-0519 inhibits all phosphodiester bond formation in both de novo initiation of RNA synthesis at the promoter and RNA elongation by a committed polymerase complex. Photocrosslinking and resistance profiling-informed ligand docking revealed that this unprecedented mechanism of action of ERDRP-0519 is due to simultaneous engagement of the L protein polyribonucleotidyl transferase (PRNTase)-like domain and the flexible intrusion loop by the compound, pharmacologically locking the polymerase in pre-initiation conformation. This study informs selection of ERDRP-0519 as clinical candidate for measles therapy and identifies a previously unrecognized druggable site in mononegavirus L polymerase proteins that can silence all synthesis of viral RNA.


Assuntos
Antivirais/farmacologia , Inibidores Enzimáticos/farmacologia , Vírus do Sarampo/efeitos dos fármacos , Sarampo/tratamento farmacológico , Morfolinas/farmacologia , Piperidinas/farmacologia , Pirazóis/farmacologia , RNA Viral/biossíntese , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Chlorocebus aethiops , Sarampo/metabolismo , Sarampo/virologia , Vírus do Sarampo/enzimologia , Mutação , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Células Vero
16.
PLoS Pathog ; 16(10): e1008253, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33031460

RESUMO

Measles is characterized by fever and a maculopapular skin rash, which is accompanied by immune clearance of measles virus (MV)-infected cells. Histopathological analyses of skin biopsies from humans and non-human primates (NHPs) with measles rash have identified MV-infected keratinocytes and mononuclear cells in the epidermis, around hair follicles and near sebaceous glands. Here, we address the pathogenesis of measles skin rash by combining data from experimentally infected NHPs, ex vivo infection of human skin sheets and in vitro infection of primary human keratinocytes. Analysis of NHP skin samples collected at different time points following MV inoculation demonstrated that infection in the skin precedes onset of rash by several days. MV infection was detected in lymphoid and myeloid cells in the dermis before dissemination to the epidermal leukocytes and keratinocytes. These data were in good concordance with ex vivo MV infections of human skin sheets, in which dermal cells were more targeted than the epidermal cells. To address viral dissemination to the epidermis and to determine whether the dissemination is receptor-dependent, we performed experimental infections of primary keratinocytes collected from healthy donors. These experiments demonstrated that MV infection of keratinocytes is mainly nectin-4-dependent, and differentiated keratinocytes, which express higher levels of nectin-4, are more susceptible to MV infection than proliferating keratinocytes. Based on these data, we propose a model to explain measles skin rash: migrating MV-infected lymphocytes initiate the infection of dermal skin-resident CD150+ immune cells. The infection is subsequently disseminated from the dermal papillae to nectin-4+ keratinocytes in the basal epidermis. Lateral spread of MV infection is observed in the superficial epidermis, most likely due to the higher level of nectin-4 expression on differentiated keratinocytes. Finally, MV-infected cells are cleared by infiltrating immune cells, causing hyperemia and edema, which give the appearance of morbilliform skin rash.


Assuntos
Derme/virologia , Epiderme/virologia , Queratinócitos/virologia , Linfócitos/virologia , Sarampo/virologia , Células Mieloides/virologia , Pele/virologia , Animais , Células Cultivadas , Derme/patologia , Epiderme/patologia , Humanos , Queratinócitos/patologia , Linfócitos/patologia , Macaca fascicularis , Sarampo/patologia , Vírus do Sarampo/isolamento & purificação , Células Mieloides/patologia , Pele/patologia
17.
J Med Virol ; 94(2): 521-530, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34761827

RESUMO

Measles is one of the most infectious diseases of humans. It is caused by the measles virus (MeV) and can lead to serious illness, lifelong complications, and even death. Whole-genome sequencing (WGS) is now available to study molecular epidemiology and identify MeV transmission pathways. In the present study, WGS of 23 MeV strains of genotype H1, collected in Mainland China between 2006 and 2018, were generated and compared to 31 WGSs from the public domain to analyze genomic characteristics, evolutionary rates and date of emergence of H1 genotype. The noncoding region between M and F protein genes (M/F NCR) was the most variable region throughout the genome. Although the nucleotide substitution rate of H1 WGS was around 0.75 × 10-3 substitution per site per year, the M/F NCR had an evolutionary rate three times higher, with 2.44 × 10-3 substitution per site per year. Phylogenetic analysis identified three distinct genetic groups. The Time of the Most Recent Common Ancestor (TMRCA) of H1 genotype was estimated at approximately 1988, while the first genetic group appeared around 1995 followed by two other genetic groups in 1999-2002. Bayesian skyline plot showed that the genetic diversity of the H1 genotype remained stable even though the number of MeV cases decreased 50 times between 2014 (52 628) and 2020 (993). The current coronavirus disease 2019 (COVID-19) pandemic might have some effect on the measles epidemic and further studies will be necessary to assess the genetic diversity of the H1 genotype in a post-COVID area.


Assuntos
Evolução Molecular , Genoma Viral/genética , Vírus do Sarampo/genética , China/epidemiologia , Genes Virais/genética , Variação Genética , Genômica , Genótipo , Humanos , Sarampo/epidemiologia , Sarampo/virologia , Vírus do Sarampo/classificação , Filogenia , RNA Viral/genética
18.
J Virol ; 94(4)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31748390

RESUMO

Measles virus (MeV), like all viruses of the order Mononegavirales, utilizes a complex consisting of genomic RNA, nucleoprotein, the RNA-dependent RNA polymerase, and a polymerase cofactor, the phosphoprotein (P), for transcription and replication. We previously showed that a recombinant MeV that does not express another viral protein, C, has severe transcription and replication deficiencies, including a steeper transcription gradient than the parental virus and generation of defective interfering RNA. This virus is attenuated in vitro and in vivo However, how the C protein operates and whether it is a component of the replication complex remained unclear. Here, we show that C associates with the ribonucleocapsid and forms a complex that can be purified by immunoprecipitation or ultracentrifugation. In the presence of detergent, the C protein is retained on purified ribonucleocapsids less efficiently than the P protein and the polymerase. The C protein is recruited to the ribonucleocapsid through its interaction with the P protein, as shown by immunofluorescence microscopy of cells expressing different combinations of viral proteins and by split luciferase complementation assays. Forty amino-terminal C protein residues are dispensable for the interaction with P, and the carboxyl-terminal half of P is sufficient for the interaction with C. Thus, the C protein, rather than being an "accessory" protein as qualified in textbooks so far, is a ribonucleocapsid-associated protein that interacts with P, thereby increasing replication accuracy and processivity of the polymerase complex.IMPORTANCE Replication of negative-strand RNA viruses relies on two components: a helical ribonucleocapsid and an RNA-dependent RNA polymerase composed of a catalytic subunit, the L protein, and a cofactor, the P protein. We show that the measles virus (MeV) C protein is an additional component of the replication complex. We provide evidence that the C protein is recruited to the ribonucleocapsid by the P protein and map the interacting segments of both C and P proteins. We conclude that the primary function of MeV C is to improve polymerase processivity and accuracy, rather than uniquely to antagonize the type I interferon response. Since most viruses of the Paramyxoviridae family express C proteins, their primary function may be conserved.


Assuntos
Vírus do Sarampo/metabolismo , Nucleoproteínas/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/genética , Animais , Proteínas de Transporte , Linhagem Celular , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Sarampo/virologia , Vírus do Sarampo/genética , Proteínas do Nucleocapsídeo , Nucleoproteínas/metabolismo , Fosfoproteínas/metabolismo , Ligação Proteica , RNA Polimerase Dependente de RNA/metabolismo , Células Vero , Proteínas não Estruturais Virais/fisiologia , Proteínas Virais/metabolismo , Ativação Viral/genética , Replicação Viral/genética
19.
J Med Virol ; 93(8): 5141-5145, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33527448

RESUMO

The increase of the evolutionary pressure will cause phylodynamics changes of viruses. In post-vaccine coverage era, measles viruses face more immune pressure than ever before. Vice versa, the phylodynamic changes may reflect herd immunity level provided by vaccination. In this study, we analyzed phylodynamic characteristics of measles viruses isolated from 2005 to 2017 in Jiangsu province of China using nucleoprotein gene sequences of measles viruses. The phylogenetic tree was constructed with Markov chain Monte Carlo algorithm. The mean gene distance within each group was computed with MEGA7.0 software. Our results showed that a decline trend is observed in the gene distance of nucleoprotein gene with time as well as incidence of measles from epidemic surveillance system. Two clusters of H1a genotype show cocirculation of multiple variants in early years and the disappearance of most variants with time. We explore the phylodynamic of measles virus under high immune pressure. Our findings highlight that phylodynamic of measles viruses is a helpful tool to assess the effectiveness of epidemic control.


Assuntos
Vacina contra Sarampo/imunologia , Vírus do Sarampo/genética , Sarampo/epidemiologia , Sarampo/virologia , China/epidemiologia , Genótipo , Humanos , Incidência , Sarampo/prevenção & controle , Vírus do Sarampo/classificação , Vírus do Sarampo/imunologia , Epidemiologia Molecular , Proteínas do Nucleocapsídeo/genética , Filogenia
20.
J Infect Dis ; 221(5): 721-728, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-31580436

RESUMO

BACKGROUND: Levels of antibodies induced by the measles virus-containing vaccine have been shown to decline over time, but there is no formal recommendation about testing immunized subjects (in particular, healthcare workers [HCWs]) to investigate the persistence of measles immunoglobulin G (IgG). METHODS: This study aims to evaluate the long-term immunogenicity of measles vaccine in a sample of medical students and residents of the University of Bari who attended the Hygiene Department for a biological risk assessment (April 2014-June 2018). RESULTS: Two thousand immunized (2 doses of measles-mumps-rubella [MMR] vaccine) students and residents were tested; 305 of these (15%) did not show protective anti-measles IgG. This proportion was higher among subjects who received vaccination at ≤15 months (20%) than in those who received vaccination at 16-23 months (17%) and at ≥24 months (10%) (P < .0001). After an MMR vaccine booster dose, we noted a seroconversion of 74% of seronegative HCWs. The overall seroconversion rate after a second dose (booster) was 93%. No serious adverse events were noted after the booster doses. CONCLUSIONS: An important proportion of subjects immunized for measles do not show a protective IgG titer in the 10 years after vaccination. Our management strategy seems consistent with the purpose of evidencing immunological memory.


Assuntos
Imunogenicidade da Vacina , Vacina contra Sarampo-Caxumba-Rubéola/imunologia , Sarampo/prevenção & controle , Morbillivirus/imunologia , Vírus da Caxumba/imunologia , Caxumba/prevenção & controle , Vírus da Rubéola/imunologia , Rubéola (Sarampo Alemão)/prevenção & controle , Vacinação , Adolescente , Adulto , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Feminino , Humanos , Esquemas de Imunização , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Itália , Masculino , Sarampo/virologia , Caxumba/virologia , Estudos Retrospectivos , Rubéola (Sarampo Alemão)/virologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA