Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
1.
J Med Virol ; 93(11): 6116-6123, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34375002

RESUMO

Virus invasion activates the host's innate immune response, inducing the production of numerous cytokines and interferons to eliminate pathogens. Except for viral DNA/RNA, viral proteins are also targets of pattern recognition receptors. Membrane-bound receptors such as Toll-like receptor (TLR)1, TLR2, TLR4, TLR6, and TLR10 relate to the recognition of viral proteins. Distinct TLRs perform both protective and detrimental roles for a specific virus. Here, we review viral proteins serving as pathogen-associated molecular patterns and their corresponding TLRs. These viruses are all enveloped, including respiratory syncytial virus, hepatitis C virus, measles virus, herpesvirus human immunodeficiency virus, and coronavirus, and can encode proteins to activate innate immunity in a TLR-dependent way. The TLR-viral protein relationship plays an important role in innate immunity activation. A detailed understanding of their pathways contributes to a novel direction for vaccine development.


Assuntos
Imunidade Inata , Moléculas com Motivos Associados a Patógenos/metabolismo , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Proteínas Virais/metabolismo , Viroses/imunologia , Vírus/imunologia , Animais , HIV/imunologia , HIV/metabolismo , HIV/patogenicidade , Hepacivirus/imunologia , Hepacivirus/metabolismo , Hepacivirus/patogenicidade , Herpesviridae/imunologia , Herpesviridae/metabolismo , Herpesviridae/patogenicidade , Humanos , Vírus do Sarampo/imunologia , Vírus do Sarampo/metabolismo , Vírus do Sarampo/patogenicidade , Moléculas com Motivos Associados a Patógenos/química , Vírus Sinciciais Respiratórios/imunologia , Vírus Sinciciais Respiratórios/metabolismo , Vírus Sinciciais Respiratórios/patogenicidade , SARS-CoV-2/imunologia , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Proteínas Virais/química , Viroses/virologia , Vírus/metabolismo , Vírus/patogenicidade
2.
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
3.
mBio ; 12(3): e0079921, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34061592

RESUMO

Measles virus (MeV) bearing a single amino acid change in the fusion protein (F)-L454W-was isolated from two patients who died of MeV central nervous system (CNS) infection. This mutation in F confers an advantage over wild-type virus in the CNS, contributing to disease in these patients. Using murine ex vivo organotypic brain cultures and human induced pluripotent stem cell-derived brain organoids, we show that CNS adaptive mutations in F enhance the spread of virus ex vivo. The spread of virus in human brain organoids is blocked by an inhibitory peptide that targets F, confirming that dissemination in the brain tissue is attributable to F. A single mutation in MeV F thus alters the fusion complex to render MeV more neuropathogenic. IMPORTANCE Measles virus (MeV) infection can cause serious complications in immunocompromised individuals, including measles inclusion body encephalitis (MIBE). In some cases, MeV persistence and subacute sclerosing panencephalitis (SSPE), another severe central nervous system (CNS) complication, develop even in the face of a systemic immune response. Both MIBE and SSPE are relatively rare but lethal. It is unclear how MeV causes CNS infection. We introduced specific mutations that are found in MIBE or SSPE cases into the MeV fusion protein to test the hypothesis that dysregulation of the viral fusion complex-comprising F and the receptor binding protein, H-allows virus to spread in the CNS. Using metagenomic, structural, and biochemical approaches, we demonstrate that altered fusion properties of the MeV H-F fusion complex permit MeV to spread in brain tissue.


Assuntos
Encéfalo/virologia , Vírus do Sarampo/genética , Proteínas Virais de Fusão/genética , Substituição de Aminoácidos , Animais , Encéfalo/citologia , Encéfalo/patologia , Doenças do Sistema Nervoso Central/virologia , Chlorocebus aethiops , Feminino , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Pluripotentes Induzidas/virologia , Masculino , Sarampo/virologia , Vírus do Sarampo/patogenicidade , Metagenômica , Camundongos , Neurônios/virologia , Organoides/citologia , Organoides/virologia , Células Vero , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/classificação , Proteínas Virais de Fusão/metabolismo
4.
J Virol ; 95(14): e0052821, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-33910952

RESUMO

Measles virus (MeV), an enveloped RNA virus in the family Paramyxoviridae, is still an important cause of childhood morbidity and mortality worldwide. MeV 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). The disease is fatal, and no effective therapy is currently available. Although transsynaptic cell-to-cell transmission is thought to account for MeV propagation in the brain, neurons do not express the known receptors for MeV. Recent studies have shown that hyperfusogenic changes in the MeV fusion (F) protein play a key role in MeV propagation in the brain. However, how such mutant viruses spread in neurons remains unexplained. Here, we show that cell adhesion molecule 1 (CADM1; also known as IGSF4A, Necl-2, and SynCAM1) and CADM2 (also known as IGSF4D, Necl-3, SynCAM2) are host factors that enable MeV to cause membrane fusion in cells lacking the known receptors and to spread between neurons. During enveloped virus entry, a cellular receptor generally interacts in trans with the attachment protein on the envelope. However, CADM1 and CADM2 interact in cis with the MeV attachment protein on the same cell membrane, causing the fusion protein triggering and membrane fusion. Knockdown of CADM1 and CADM2 inhibits syncytium formation and virus transmission between neurons that are both mediated by hyperfusogenic F proteins. Thus, our results unravel the molecular mechanism (receptor-mimicking cis-acting fusion triggering) by which MeV spreads transsynaptically between neurons, thereby causing SSPE. IMPORTANCE Measles virus (MeV), an enveloped RNA virus, is the causative agent of measles, which is still an important cause of childhood morbidity and mortality worldwide. Persistent MeV infection in the brain causes a fatal progressive neurological disorder, subacute sclerosing panencephalitis (SSPE), several years after acute infection. However, how MeV spreads in neurons, which are mainly affected in SSPE, remains largely unknown. In this study, we demonstrate that cell adhesion molecule 1 (CADM1) and CADM2 are host factors enabling MeV spread between neurons. During enveloped virus entry, a cellular receptor generally interacts in trans with the attachment protein on the viral membrane (envelope). Remarkably, CADM1 and CADM2 interact in cis with the MeV attachment protein on the same membrane, triggering the fusion protein and causing membrane fusion, as viral receptors usually do in trans. Careful screening may lead to more examples of such "receptor-mimicking cis-acting fusion triggering" in other viruses.


Assuntos
Molécula 1 de Adesão Celular/fisiologia , Moléculas de Adesão Celular/fisiologia , Vírus do Sarampo/patogenicidade , Panencefalite Esclerosante Subaguda/virologia , Internalização do Vírus , Animais , Linhagem Celular , Chlorocebus aethiops , Células Gigantes/virologia , Humanos , Camundongos , Células Vero , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/metabolismo
5.
Viruses ; 13(4)2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33916225

RESUMO

Measles virus (MV) can cause severe acute diseases as well as long-lasting clinical deteriorations due to viral-induced immunosuppression and neuronal manifestation. How the virus enters the brain and manages to persist in neuronal tissue is not fully understood. Various mutations in the viral genes were found in MV strains isolated from patient brains. In this study, reverse genetics was used to introduce mutations in the fusion, matrix and polymerase genes of MV. The generated virus clones were characterized in cell culture and used to infect rat brain slice cultures. A mutation in the carboxy-terminal domain of the matrix protein (R293Q) promoted the production of progeny virions. This effect was observed in Vero cells irrespective of the expression of the signaling lymphocyte activation molecule (SLAM). Furthermore, a mutation in the fusion protein (I225M) induced syncytia formation on Vero cells in the absence of SLAM and promoted viral spread throughout the rat brain slices. In this study, a solid ex vivo model was established to elucidate the MV mutations contributing to neural manifestation.


Assuntos
Encéfalo/virologia , Vírus do Sarampo/genética , Mutação , Neurônios/virologia , Proteínas Virais/genética , Tropismo Viral/genética , Animais , Chlorocebus aethiops , Células HEK293 , Humanos , Técnicas In Vitro , Sarampo/virologia , Vírus do Sarampo/patogenicidade , Vírus do Sarampo/fisiologia , Ratos Endogâmicos Lew , Genética Reversa , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Células Vero , Proteínas Virais de Fusão/genética
6.
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
7.
Viruses ; 12(2)2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32033013

RESUMO

Oncolytic viruses, including live attenuated measles virus (MV) vaccine strains, have recently been shown as promising therapeutic agents against human malignancies. In this study, the oncolytic potential of the attenuated vaccine strain Leningrad-16 (L-16) of MV was evaluated in a panel of human metastatic melanoma cell lines. The L-16 measles virus was shown to replicate within melanoma cells mediating direct cell killing of tumor cells, although all melanoma cell lines varied in regard to their ability to respond to L-16 MV infection, as revealed by the different pattern of the Interferon Stimulated Gene expression, cytokine release and mechanisms of cell death. Furthermore, the statistically significant L-16 measles virus related tumor growth inhibition was demonstrated in a melanoma xenograft model. Therefore, L-16 MV represents an appealing oncolytic platform for target delivery of therapeutic genes along with other attenuated measles virus strains.


Assuntos
Vírus do Sarampo/patogenicidade , Melanoma/terapia , Melanoma/virologia , Vírus Oncolíticos/patogenicidade , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Vacina contra Sarampo , Camundongos Endogâmicos BALB C , Camundongos Nus , Terapia Viral Oncolítica/métodos , Vacinas Atenuadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Virol ; 94(2)2020 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-31619560

RESUMO

Measles virus (MeV) is an enveloped RNA virus bearing two envelope glycoproteins, the hemagglutinin (H) and fusion (F) proteins. Upon receptor binding, the H protein triggers conformational changes of the F protein, causing membrane fusion and subsequent virus entry. MeV may persist in the brain, infecting neurons and causing fatal subacute sclerosing panencephalitis (SSPE). Since neurons do not express either of the MeV receptors, signaling lymphocytic activation molecule (SLAM; also called CD150) and nectin-4, how MeV propagates in neurons is unknown. Recent studies have shown that specific substitutions in the F protein found in MeV isolates from SSPE patients are critical for MeV neuropathogenicity by rendering the protein unstable and hyperfusogenic. Recombinant MeVs possessing the F proteins with such substitutions can spread in primary human neurons and in the brains of mice and hamsters and induce cell-cell fusion in cells lacking SLAM and nectin-4. Here, we show that receptor-blind mutant H proteins that have decreased binding affinities to receptors can support membrane fusion mediated by hyperfusogenic mutant F proteins, but not the wild-type F protein, in cells expressing the corresponding receptors. The results suggest that weak interactions of the H protein with certain molecules (putative neuron receptors) trigger hyperfusogenic F proteins in SSPE patients. Notably, where cell-cell contacts are ensured, the weak cis interaction of the H protein with SLAM on the same cell surface also could trigger hyperfusogenic F proteins. Some enveloped viruses may exploit such cis interactions with receptors to infect target cells, especially in cell-to-cell transmission.IMPORTANCE Measles virus (MeV) may persist in the brain, causing incurable subacute sclerosing panencephalitis (SSPE). Because neurons, the main target in SSPE, do not express receptors for wild-type (WT) MeV, how MeV propagates in the brain is a key question for the disease. Recent studies have demonstrated that specific substitutions in the MeV fusion (F) protein are critical for neuropathogenicity. Here, we show that weak cis and trans interactions of the MeV attachment protein with receptors that are not sufficient to trigger the WT MeV F protein can trigger the mutant F proteins from neuropathogenic MeV isolates. Our study not only provides an important clue to understand MeV neuropathogenicity but also reveals a novel viral strategy to expand cell tropism.


Assuntos
Moléculas de Adesão Celular/metabolismo , Hemaglutininas Virais/metabolismo , Vírus do Sarampo/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Panencefalite Esclerosante Subaguda/metabolismo , Proteínas Virais de Fusão/metabolismo , Animais , Moléculas de Adesão Celular/genética , Linhagem Celular , Cricetinae , Hemaglutininas Virais/genética , Humanos , Vírus do Sarampo/genética , Vírus do Sarampo/patogenicidade , Camundongos , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Panencefalite Esclerosante Subaguda/genética , Panencefalite Esclerosante Subaguda/patologia , Proteínas Virais de Fusão/genética
9.
Cancer Lett ; 460: 108-118, 2019 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-31226409

RESUMO

Live-attenuated strain of measles virus (MV) has oncolytic effect. In this study, the antitumor effect of rMV-Hu191, a recombinant Chinese Hu191 MV generated in our laboratory by efficient reverse genetics system, was evaluated in gastric cancer (GC). From our data, rMV-Hu191 induced cytopathic effects and inhibited tumor proliferation both in vitro and in vivo by inducing caspase-dependent apoptosis. In mice bearing GC xenografts, tumor size was reduced and survival was prolonged significantly after intratumoral injections of rMV-Hu191. Furthermore, lipid rafts, a type of membrane microdomain with specific lipid compositions, played an important role in facilitating entry of rMV-Hu191. Integrity of lipid rafts was required for successful viral infection as well as subsequent cell apoptosis, but was not required for viral binding and replication. CD46, a MV membrane receptor, was found to be partially localized in lipid rafts microdomains. This is the first study to demonstrate that Chinese Hu191 MV vaccine strain could be used as a potentially effective therapeutic agent in GC treatment. As part of the underlying cellular mechanism, the integrity of lipid rafts is required for viral entry and to exercise the oncolytic effect.


Assuntos
Apoptose , Vírus do Sarampo/patogenicidade , Microdomínios da Membrana/virologia , Terapia Viral Oncolítica , Vírus Oncolíticos/patogenicidade , Neoplasias Gástricas/terapia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Chlorocebus aethiops , Efeito Citopatogênico Viral , Humanos , Masculino , Vírus do Sarampo/genética , Proteína Cofatora de Membrana/metabolismo , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/patologia , Camundongos Nus , Vírus Oncolíticos/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Neoplasias Gástricas/virologia , Carga Tumoral , Células Vero , Internalização do Vírus , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Am J Dermatopathol ; 41(12): 914-923, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31021834

RESUMO

Despite available vaccination, measles is one of the leading causes of death among young children in developing countries. In clinical practice, the spectrum of differential diagnoses of morbilliform exanthemas associated with fever is wide, and it can be hard to differentiate from other infectious eruptions, especially in adults or in atypical courses in immunocompromised patients. The goal of our study was to identify characteristic histomorphological and immunohistochemical patterns of measles exanthema through the study of 13 skin biopsy specimens obtained from 13 patients with this disease and a review of cases in the literature. Histopathological features of measles exanthema are quite distinctive and characterized by a combination of multinucleated keratinocytes, and individual and clustered necrotic keratinocytes in the epidermis with pronounced folliculosebaceous as well as acrosyringeal involvement. Immunohistochemical staining of skin biopsies with anti-measles virus (MeV) nucleoprotein and anti-MeV phosphoprotein can be of great value in confirming the diagnosis of measles. Both methods can serve as quick additional diagnostic tools for prompt implementation of quarantine measures and for providing medical assistance, even in patients in whom the clinician did not consider measles as a differential diagnosis of the rash due to the rarity of the disease in a putatively vaccinated community.


Assuntos
Exantema/diagnóstico , Imuno-Histoquímica , Vírus do Sarampo/patogenicidade , Sarampo/diagnóstico , Sarampo/patologia , Nucleoproteínas/análise , Fosfoproteínas/análise , Dermatopatias Virais/diagnóstico , Pele , Proteínas Virais/análise , Adolescente , Adulto , Biópsia , Diagnóstico Diferencial , Exantema/metabolismo , Exantema/patologia , Exantema/virologia , Feminino , Humanos , Masculino , Sarampo/metabolismo , Sarampo/virologia , Pessoa de Meia-Idade , Proteínas do Nucleocapsídeo , Valor Preditivo dos Testes , Estudos Retrospectivos , Pele/química , Pele/patologia , Pele/virologia , Dermatopatias Virais/metabolismo , Dermatopatias Virais/patologia , Dermatopatias Virais/virologia , Adulto Jovem
11.
Med Sci Monit ; 25: 903-912, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30705250

RESUMO

BACKGROUND The purpose of this study was to explore the immune mechanism of dendritic cells (DCs) against measles virus (MV), and to identify potential biomarkers to improve measles prevention and treatment. MATERIAL AND METHODS The gene expression profile of GSE980, which comprised 10 DC samples from human blood infected with MV (RNA was isolated at 3, 6, 12, and 24 h post-infection) and 4 normal DC control samples, was obtained from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between the MV-infected DC samples and the control samples were screened using Genevestigator software. Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analyses were performed using GenCLip 2.0 and STRING 10.5 software. The protein-protein interaction (PPI) network was established using Cytoscape 3.4.0. RESULTS The gene expression profiles of MV-infected DCs were obviously changed. Twenty-six common DEGs (0.9%, MV-infected DCs vs. normal DCs) were identified at 4 different time points, including 14 down-regulated and 12 up-regulated genes (P=0.001). GO analysis showed that DEGs were significantly enriched in defense response to virus, type I interferon signaling pathway, et al. ISG15 and CXCL10 were the key genes in the PPI network of the DEGs, and may interact directly with the type I interferon signaling and defense response to virus signaling. CONCLUSIONS The DEGs increased gradually with the duration of MV infection. The type I interferon signaling pathway and the defense response to viral processes can be activated against MV by ISG15 and CXCL10 in DCs. These may provide novel targets for the treatment of MV.


Assuntos
Biologia Computacional/métodos , Células Dendríticas/imunologia , Vírus do Sarampo/imunologia , Biomarcadores , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/genética , Ontologia Genética , Redes Reguladoras de Genes , Humanos , Vírus do Sarampo/patogenicidade , Mapeamento de Interação de Proteínas , Mapas de Interação de Proteínas , Software , Transcriptoma/genética
12.
PLoS Pathog ; 15(2): e1007605, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30768648

RESUMO

Measles virus (MeV) is dual-tropic: it replicates first in lymphatic tissues and then in epithelial cells. This switch in tropism raises the question of whether, and how, intra-host evolution occurs. Towards addressing this question, we adapted MeV either to lymphocytic (Granta-519) or epithelial (H358) cells. We also passaged it consecutively in both human cell lines. Since passaged MeV had different replication kinetics, we sought to investigate the underlying genetic mechanisms of growth differences by performing deep-sequencing analyses. Lymphocytic adaptation reproducibly resulted in accumulation of variants mapping within an 11-nucleotide sequence located in the middle of the phosphoprotein (P) gene. This sequence mediates polymerase slippage and addition of a pseudo-templated guanosine to the P mRNA. This form of co-transcriptional RNA editing results in expression of an interferon antagonist, named V, in place of a polymerase co-factor, named P. We show that lymphocytic-adapted MeV indeed produce minimal amounts of edited transcripts and V protein. In contrast, parental and epithelial-adapted MeV produce similar levels of edited and non-edited transcripts, and of V and P proteins. Raji, another lymphocytic cell line, also positively selects V-deficient MeV genomes. On the other hand, in epithelial cells V-competent MeV genomes rapidly out-compete the V-deficient variants. To characterize the mechanisms of genome re-equilibration we rescued four recombinant MeV carrying individual editing site-proximal mutations. Three mutations interfered with RNA editing, resulting in almost exclusive P protein expression. The fourth preserved RNA editing and a standard P-to-V protein expression ratio. However, it altered a histidine involved in Zn2+ binding, inactivating V function. Thus, the lymphocytic environment favors replication of V-deficient MeV, while the epithelial environment has the opposite effect, resulting in rapid and thorough cyclical quasispecies re-equilibration. Analogous processes may occur in natural infections with other dual-tropic RNA viruses.


Assuntos
Vírus do Sarampo/metabolismo , Fosfoproteínas/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Linfócitos/metabolismo , Linfócitos/virologia , Sarampo , Vírus do Sarampo/patogenicidade , Fosfoproteínas/genética , Quase-Espécies/genética , Quase-Espécies/imunologia , Edição de RNA/genética , RNA Mensageiro/genética , Transcrição Gênica , Proteínas Virais/genética , Replicação Viral/genética
13.
Trends Microbiol ; 27(2): 164-175, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30220445

RESUMO

Measles virus (MeV) may persist in the brain, causing fatal neurodegenerative diseases, subacute sclerosing panencephalitis, and measles inclusion-body encephalitis. However, the mechanism of MeV propagation in the brain remains unexplained because human neurons affected by the diseases do not express the known receptors for MeV. Recent studies have revealed that certain changes in the ectodomain of the MeV fusion (F) protein play a key role in MeV spread in the brain. These changes destabilize the prefusion form of the F protein and render it hyperfusogenic, which in turn allows the virus to propagate in neurons. Based on crystal structures of the F protein, effective fusion inhibitors could be developed to treat these diseases.


Assuntos
Encéfalo/virologia , Vírus do Sarampo/patogenicidade , Sarampo/virologia , Panencefalite Esclerosante Subaguda/virologia , Substituição de Aminoácidos , Animais , Humanos , Sarampo/tratamento farmacológico , Neurônios/virologia , Conformação Proteica , Panencefalite Esclerosante Subaguda/tratamento farmacológico , Proteínas Virais de Fusão/química
14.
J Pharm Biomed Anal ; 161: 73-82, 2018 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-30145452

RESUMO

Protein thermal shift assay (TSA) has been extensively used in investigation of protein stabilization (for protein biopharmaceutics stabilization, protein crystallization studies or screening of recombinant proteins) and drug discovery (screening of ligands or inhibitors). This work aimed to analyze thermal shift assay results in comparison to protein polymerization (multimerization and aggregation) propensity and test the most stabilizing formulations for their stabilization effect on enveloped viruses. Influence of protein concentration, buffer pH and molarity was tested on three proteins (immunoglobulin G, ovalbumin, and albumin) and results showed that each of these factors has an impact on determined shift in protein melting point Tm, and the impact was similar for all three proteins. In case of ovalbumin, molecular dynamics simulations were performed with the goal to understanding molecular basis of protein's thermal stability dependence on pH. Effect of three denaturing agents in a wide concentration range on Tm showed nicely that chemical denaturation occurs only at the highest concentrations. Results showed similar effect on Tm for most formulations on different proteins. Most successful formulations were tested for enveloped virus stabilizing potential using cell culture infectivity assay (CCID50) and results showed lack of correlation with TSA results. Only weak correlation of Tm shift and protein polymerization measured by SEC-HPLC was obtained, meaning that polymerization cannot be predicted from Tm shifts.


Assuntos
Vírus do Sarampo/química , Vírus da Caxumba/química , Estabilidade Proteica , Proteínas do Envelope Viral/química , Albuminas/química , Células Cultivadas , Composição de Medicamentos , Estabilidade de Medicamentos , Guanidina/química , Concentração de Íons de Hidrogênio , Imunoglobulina G/química , Vírus do Sarampo/patogenicidade , Simulação de Dinâmica Molecular , Vírus da Caxumba/patogenicidade , Ovalbumina/química , Polimerização , Cianeto de Potássio/química , Desnaturação Proteica/efeitos dos fármacos , Temperatura de Transição , Ureia/química
15.
Virology ; 518: 210-220, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29525671

RESUMO

The live-attenuated measles virus (MV) vaccine based on the Hu191 strain has played a significant role in controlling measles in China. However, it has considerable adverse effects that may cause public health burden. We hypothesize that the safety and efficacy of MV vaccine can be improved by altering the S-adenosylmethionine (SAM) binding site in the conserved region VI of the large polymerase protein. To test this hypothesis, we established an efficient reverse genetics system for the rMV-Hu191 strain and generated two recombinant MV-Hu191 carrying mutations in the SAM binding site. These two mutants grew to high titer in Vero cells, were genetically stable, and were significantly more attenuated in vitro and in vivo compared to the parental rMV-Hu191 vaccine strain. Importantly, both MV-Hu191 mutants triggered a higher neutralizing antibody than rMV-Hu191 vaccine and provided complete protection against MV challenge. These results demonstrate its potential for an improved MV vaccine candidate.


Assuntos
Vacina contra Sarampo/efeitos adversos , Vacina contra Sarampo/imunologia , Vírus do Sarampo/patogenicidade , Mutação , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , S-Adenosilmetionina/metabolismo , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Sítios de Ligação , China , Chlorocebus aethiops , Feminino , Humanos , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/genética , Vírus do Sarampo/imunologia , Ligação Proteica , Genética Reversa , Sigmodontinae , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Células Vero
16.
Otolaryngol Clin North Am ; 51(2): 305-318, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29502723

RESUMO

Over the past several years, with the evolution of genetic and molecular research, several etiologic factors have been implicated in the pathogenesis of otosclerosis. Overall, current evidence suggests that otosclerosis is a complex disease with a variety of potential pathways contributing to the development of abnormal bone remodeling in the otic capsule. These pathways involved in the pathogenesis of otosclerosis are influenced by both genetic and environmental factors.


Assuntos
Otosclerose/genética , Otosclerose/patologia , Remodelação Óssea/genética , Orelha Interna/patologia , Expressão Gênica/fisiologia , Predisposição Genética para Doença/genética , Humanos , Vírus do Sarampo/patogenicidade , Osteoprotegerina/genética , Otosclerose/etiologia , Ligante RANK/genética , Osso Temporal/patologia
17.
J Virol ; 92(6)2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29298883

RESUMO

Measles virus (MV) usually causes acute infection but in rare cases persists in the brain, resulting in subacute sclerosing panencephalitis (SSPE). Since human neurons, an important target affected in the disease, do not express the known MV receptors (signaling lymphocyte activation molecule [SLAM] and nectin 4), how MV infects neurons and spreads between them is unknown. Recent studies have shown that many virus strains isolated from SSPE patients possess substitutions in the extracellular domain of the fusion (F) protein which confer enhanced fusion activity. Hyperfusogenic viruses with such mutations, unlike the wild-type MV, can induce cell-cell fusion even in SLAM- and nectin 4-negative cells and spread efficiently in human primary neurons and the brains of animal models. We show here that a hyperfusogenic mutant MV, IC323-F(T461I)-EGFP (IC323 with a fusion-enhancing T461I substitution in the F protein and expressing enhanced green fluorescent protein), but not the wild-type MV, spreads in differentiated NT2 cells, a widely used human neuron model. Confocal time-lapse imaging revealed the cell-to-cell spread of IC323-F(T461I)-EGFP between NT2 neurons without syncytium formation. The production of virus particles was strongly suppressed in NT2 neurons, also supporting cell-to-cell viral transmission. The spread of IC323-F(T461I)-EGFP was inhibited by a fusion inhibitor peptide as well as by some but not all of the anti-hemagglutinin antibodies which neutralize SLAM- or nectin-4-dependent MV infection, suggesting the presence of a distinct neuronal receptor. Our results indicate that MV spreads in a cell-to-cell manner between human neurons without causing syncytium formation and that the spread is dependent on the hyperfusogenic F protein, the hemagglutinin, and the putative neuronal receptor for MV.IMPORTANCE Measles virus (MV), in rare cases, persists in the human central nervous system (CNS) and causes subacute sclerosing panencephalitis (SSPE) several years after acute infection. This neurological complication is almost always fatal, and there is currently no effective treatment for it. Mechanisms by which MV invades the CNS and causes the disease remain to be elucidated. We have previously shown that fusion-enhancing substitutions in the fusion protein of MVs isolated from SSPE patients contribute to MV spread in neurons. In this study, we demonstrate that MV bearing the hyperfusogenic mutant fusion protein spreads between human neurons in a cell-to-cell manner. Spread of the virus was inhibited by a fusion inhibitor peptide and antibodies against the MV hemagglutinin, indicating that both the hemagglutinin and hyperfusogenic fusion protein play important roles in MV spread between human neurons. The findings help us better understand the disease process of SSPE.


Assuntos
Hemaglutininas Virais/metabolismo , Vírus do Sarampo/metabolismo , Sarampo/transmissão , Neurônios/metabolismo , Panencefalite Esclerosante Subaguda/transmissão , Proteínas Virais de Fusão/metabolismo , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Chlorocebus aethiops , Hemaglutininas Virais/genética , Humanos , Sarampo/genética , Sarampo/metabolismo , Sarampo/patologia , Vírus do Sarampo/genética , Vírus do Sarampo/patogenicidade , Neurônios/patologia , Neurônios/virologia , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Panencefalite Esclerosante Subaguda/genética , Panencefalite Esclerosante Subaguda/metabolismo , Panencefalite Esclerosante Subaguda/patologia , Células Vero , Proteínas Virais de Fusão/genética
18.
Molecules ; 22(11)2017 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-29088094

RESUMO

Measles virus (MV) infection is re-emerging, despite the availability of an effective vaccine. The mechanism of MV entry into a target cell relies on coordinated action between the MV hemagglutinin (H) receptor binding protein and the fusion envelope glycoprotein (F) which mediates fusion between the viral and cell membranes. Peptides derived from the C-terminal heptad repeat (HRC) of F can interfere with this process, blocking MV infection. As previously described, biophysical properties of HRC-derived peptides modulate their antiviral potency. In this work, we characterized a MV peptide fusion inhibitor conjugated to 25-hydroxycholesterol (25HC), a cholesterol derivative with intrinsic antiviral activity, and evaluated its interaction with membrane model systems and human blood cells. The peptide (MV.


Assuntos
Antivirais/química , Antivirais/farmacologia , Hidroxicolesteróis/química , Vírus do Sarampo/patogenicidade , Proteínas Virais de Fusão/química , Antivirais/metabolismo , Fusão Celular , Difusão Dinâmica da Luz , Eritrócitos/efeitos dos fármacos , Humanos , Ressonância de Plasmônio de Superfície , Lipossomas Unilamelares/metabolismo , Proteínas Virais de Fusão/genética
19.
Indian J Med Res ; 145(4): 536-542, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28862187

RESUMO

BACKGROUND & OBJECTIVES: Measles infection is reported to be more severe, prolonged and associated with a higher complication rate in children with HIV infection. Reports indicate that infants born to HIV-infected women [HIV exposed infants (HEI)] may be more vulnerable to measles. The World Health Organization recommends measles vaccination starting at six months of age in these infants who may be HIV-infected themselves. However, in India, they are given measles vaccination at nine months of age like all other infants. In this study, the seroprevalence of transplacentally acquired measles antibodies was compared in HEI and unexposed infants (HUnI) at six months of age and the proportion of HEI undergoing seroconversion after immunization with measles vaccine was assessed. METHODS: In this prospective longitudinal study, measles IgG antibodies were estimated in serum of 49 HEI and 50 HUnI aged 6-7 months. Measles vaccine was then administered to HEI. Assessment for measles IgG antibodies was repeated 8-12 wk post-immunization. RESULTS: Measles IgG antibodies were detected in two of 49 (4.1%) HEI and 16 of 50 (32%) HUnI. HEI were 11 times more likely to lack measles antibodies as compared to HUnI (odds ratio=11.05, 95% confidence interval=2.989-40.908). Post-vaccination, seroprevalence of measles antibodies increased to 38.5 per cent (PInterpretation & conclusions: Most HEI lacked measles antibodies at six months age and were, therefore, more vulnerable to measles than HUnI. Seroconversion in response to a single dose of measles vaccine administered at six months age was low in these infants, signifying the need of additional dose(s) of measles/measles-containing vaccine.


Assuntos
Anticorpos Antivirais/sangue , Infecções por HIV/sangue , Sarampo/sangue , Anticorpos Antivirais/imunologia , Feminino , HIV/imunologia , HIV/patogenicidade , Infecções por HIV/complicações , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Índia/epidemiologia , Lactente , Masculino , Sarampo/complicações , Sarampo/imunologia , Sarampo/virologia , Vacina contra Sarampo , Vírus do Sarampo/patogenicidade , Estudos Soroepidemiológicos
20.
J Virol ; 91(20)2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28768856

RESUMO

Attenuated measles virus (MV) is one of the most effective and safe vaccines available, making it an attractive candidate vector for preventing other infectious diseases. Yet the great capacity of this vaccine still needs to be understood at the molecular level. MV vaccine strains have different type I interferon (IFN)-inducing abilities that partially depend on the presence of 5' copy-back defective interfering genomes (DI-RNAs). DI-RNAs are pathogen-associated molecular patterns recognized by RIG-I-like receptors (RLRs) (RIG-I, MDA5, and LGP2) that activate innate immune signaling and shape the adaptive immune response. In this study, we characterized the DI-RNAs produced by various modified recombinant MVs (rMVs), including vaccine candidates, as well as wild-type MV. All tested rMVs produced 5' copy-back DI-RNAs that were different in length and nucleotide sequence but still respected the so-called "rule of six." We correlated the presence of DI-RNAs with a larger stimulation of the IFN-ß pathway and compared their immunostimulatory potentials. Importantly, we revealed that encapsidation of DI-RNA molecules within the MV nucleocapsid abolished their immunoactive properties. Furthermore, we identified specific interactions of DI-RNAs with both RIG-I and LGP2 but not MDA5. Our results suggest that DI-RNAs produced by rMV vaccine candidates may indeed strengthen their efficiency by triggering RLR signaling.IMPORTANCE Having been administered to hundreds of millions of children, the live attenuated measles virus (MV) vaccine is the safest and most widely used human vaccine, providing high protection with long-term memory. Additionally, recombinant MVs carrying heterologous antigens are promising vectors for new vaccines. The great capacity of this vaccine still needs to be elucidated at the molecular level. Here we document that recombinant MVs produce defective interfering genomes that have high immunostimulatory properties via their binding to RIG-I and LGP2 proteins, both of which are cytosolic nonself RNA sensors of innate immunity. Defective interfering genome production during viral replication should be considered of great importance due to the immunostimulatory properties of these genomes as intrinsic adjuvants produced by the vector that increase recognition by the innate immune system.


Assuntos
Genoma Viral , Helicase IFIH1 Induzida por Interferon/metabolismo , Vírus do Sarampo/genética , RNA Helicases/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Receptores do Ácido Retinoico/metabolismo , Linhagem Celular , Humanos , Imunidade Inata , Interferon beta/metabolismo , Sarampo/virologia , Vacina contra Sarampo/genética , Vacina contra Sarampo/imunologia , Vírus do Sarampo/patogenicidade , Nucleocapsídeo/metabolismo , RNA Viral/imunologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA