Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell ; 184(25): 6037-6051.e14, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34852237

RESUMEN

RNA viruses generate defective viral genomes (DVGs) that can interfere with replication of the parental wild-type virus. To examine their therapeutic potential, we created a DVG by deleting the capsid-coding region of poliovirus. Strikingly, intraperitoneal or intranasal administration of this genome, which we termed eTIP1, elicits an antiviral response, inhibits replication, and protects mice from several RNA viruses, including enteroviruses, influenza, and SARS-CoV-2. While eTIP1 replication following intranasal administration is limited to the nasal cavity, its antiviral action extends non-cell-autonomously to the lungs. eTIP1 broad-spectrum antiviral effects are mediated by both local and distal type I interferon responses. Importantly, while a single eTIP1 dose protects animals from SARS-CoV-2 infection, it also stimulates production of SARS-CoV-2 neutralizing antibodies that afford long-lasting protection from SARS-CoV-2 reinfection. Thus, eTIP1 is a safe and effective broad-spectrum antiviral generating short- and long-term protection against SARS-CoV-2 and other respiratory infections in animal models.


Asunto(s)
Proteínas de la Cápside/genética , Virus Interferentes Defectuosos/metabolismo , Replicación Viral/efectos de los fármacos , Administración Intranasal , Animales , Antivirales/farmacología , Anticuerpos ampliamente neutralizantes/inmunología , Anticuerpos ampliamente neutralizantes/farmacología , COVID-19 , Proteínas de la Cápside/metabolismo , Línea Celular , Virus Interferentes Defectuosos/patogenicidad , Modelos Animales de Enfermedad , Genoma Viral/genética , Humanos , Gripe Humana , Interferones/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Poliovirus/genética , Poliovirus/metabolismo , Infecciones del Sistema Respiratorio/virología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad
2.
J Virol ; 97(5): e0034023, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37166307

RESUMEN

Measles virus (MeV), the causative agent of measles, is an enveloped RNA virus of the family Paramyxoviridae, which remains an important cause of childhood morbidity and mortality. MeV has two envelope glycoproteins, the hemagglutinin (H) and fusion (F) proteins. During viral entry or virus-mediated fusion between infected cells and neighboring susceptible cells, the head domain of the H protein initially binds to its receptors, signaling lymphocytic activation molecule family member 1 (SLAM) and nectin-4, and then the stalk region of the H protein transmits the fusion-triggering signal to the F protein. MeV may persist in the human brain and cause a fatal neurodegenerative disease, subacute sclerosing panencephalitis (SSPE). Recently, we showed, using in vitro cell culture, that cell adhesion molecule (CADM) 1 and CADM2 are host factors that trigger hyperfusogenic mutant F proteins, causing cell-to-cell fusion and the transfer of the MeV genome between neurons. Unlike conventional receptors, CADM1 and CADM2 interact in cis (on the same membrane) with the H protein and then trigger membrane fusion. Here, we show that alanine substitutions in part of the stalk region (positions 171-175) abolish the ability of the H protein to mediate membrane fusion triggered by CADM1 and CADM2, but not by SLAM. The recombinant hyperfusogenic MeV carrying this mutant H protein loses its ability to spread in primary mouse neurons as well as its neurovirulence in experimentally infected suckling hamsters. These results indicate that CADM1 and CADM2 are key molecules for MeV propagation in the brain and its neurovirulence in vivo. IMPORTANCE Measles is an acute febrile illness with skin rash. Despite the availability of highly effective vaccines, measles is still an important cause of childhood morbidity and mortality in many countries. The World Health Organization estimates that more than 120,000 people died from measles worldwide in 2021. Measles virus (MeV), the causative agent of measles, can also cause a fatal progressive neurological disorder, subacute sclerosing panencephalitis (SSPE), several years after acute infection. There is currently no effective treatment for this disease. In this study, using recombinant MeVs with altered receptor usage patterns, we show that cell adhesion molecule (CADM) 1 and CADM2 are host factors critical for MeV spread in neurons and its neurovirulence. These findings further our understanding of the molecular mechanism of MeV neuropathogenicity.


Asunto(s)
Sarampión , Enfermedades Neurodegenerativas , Panencefalitis Esclerosante Subaguda , Cricetinae , Humanos , Ratones , Animales , Virus del Sarampión/fisiología , Panencefalitis Esclerosante Subaguda/genética , Hemaglutininas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Proteínas Recombinantes/metabolismo , Neuronas , Molécula 1 de Adhesión Celular/metabolismo
3.
J Virol ; 96(3): e0194921, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-34788082

RESUMEN

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.


Asunto(s)
Molécula 1 de Adhesión Celular/metabolismo , Células Gigantes/virología , Hemaglutininas Virales/metabolismo , Interacciones Huésped-Patógeno , Virus del Sarampión/fisiología , Sarampión/metabolismo , Sarampión/virología , Animales , Encéfalo/metabolismo , Encéfalo/virología , Molécula 1 de Adhesión Celular/genética , Células Cultivadas , Cricetinae , Modelos Biológicos , Unión Proteica , Isoformas de Proteínas , Proteínas Virales de Fusión/metabolismo
4.
PLoS Pathog ; 17(9): e1009277, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34570820

RESUMEN

During replication, RNA viruses accumulate genome alterations, such as mutations and deletions. The interactions between individual variants can determine the fitness of the virus population and, thus, the outcome of infection. To investigate the effects of defective interfering genomes (DI) on wild-type (WT) poliovirus replication, we developed an ordinary differential equation model, which enables exploring the parameter space of the WT and DI competition. We also experimentally examined virus and DI replication kinetics during co-infection, and used these data to infer model parameters. Our model identifies, and our experimental measurements confirm, that the efficiencies of DI genome replication and encapsidation are two most critical parameters determining the outcome of WT replication. However, an equilibrium can be established which enables WT to replicate, albeit to reduced levels.


Asunto(s)
Coinfección/virología , Virus Defectuosos , Modelos Teóricos , Poliovirus , Replicación Viral/fisiología , Virus Defectuosos/fisiología , Humanos , Poliovirus/fisiología
5.
J Virol ; 95(14): e0052821, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-33910952

RESUMEN

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.


Asunto(s)
Molécula 1 de Adhesión Celular/fisiología , Moléculas de Adhesión Celular/fisiología , Virus del Sarampión/patogenicidad , Panencefalitis Esclerosante Subaguda/virología , Internalización del Virus , Animales , Línea Celular , Chlorocebus aethiops , Células Gigantes/virología , Humanos , Ratones , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales de Fusión/metabolismo
6.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31619560

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Hemaglutininas Virales/metabolismo , Virus del Sarampión/metabolismo , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Panencefalitis Esclerosante Subaguda/metabolismo , Proteínas Virales de Fusión/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Línea Celular , Cricetinae , Hemaglutininas Virales/genética , Humanos , Virus del Sarampión/genética , Virus del Sarampión/patogenicidad , Ratones , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Panencefalitis Esclerosante Subaguda/genética , Panencefalitis Esclerosante Subaguda/patología , Proteínas Virales de Fusión/genética
7.
J Virol ; 94(12)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32295904

RESUMEN

Mumps virus (MuV), an enveloped RNA virus of the Paramyxoviridae family and the causative agent of mumps, affects the salivary glands and other glandular tissues as well as the central nervous system. The virus enters the cell by inducing the fusion of its envelope with the plasma membrane of the target cell. Membrane fusion is mediated by MuV envelope proteins: the hemagglutinin-neuraminidase and fusion (F) protein. Cleavage of the MuV F protein (MuV-F) into two subunits by the cellular protease furin is a prerequisite for fusion and virus infectivity. Here, we show that 293T (a derivative of HEK293) cells do not produce syncytia upon expression of MuV envelope proteins or MuV infection. This failure is caused by the inefficient MuV-F cleavage despite the presence of functional furin in 293T cells. An expression cloning strategy revealed that overexpression of lysosome-associated membrane proteins (LAMPs) confers on 293T cells the ability to produce syncytia upon expression of MuV envelope proteins. The LAMP family comprises the ubiquitously expressed LAMP1 and LAMP2, the interferon-stimulated gene product LAMP3, and the cell type-specific proteins. The expression level of the LAMP3 gene, but not of LAMP1 and LAMP2 genes, differed markedly between 293T and HEK293 cells. Overexpression of LAMP1, LAMP2, or LAMP3 allowed 293T cells to process MuV-F efficiently. Furthermore, these LAMPs were found to interact with both MuV-F and furin. Our results indicate that LAMPs support the furin-mediated cleavage of MuV-F and that, among them, LAMP3 may be critical for the process, at least in certain cells.IMPORTANCE The cellular protease furin mediates proteolytic cleavage of many host and pathogen proteins and plays an important role in viral envelope glycoprotein maturation. MuV, an enveloped RNA virus of the Paramyxoviridae family and an important human pathogen, enters the cell through the fusion of its envelope with the plasma membrane of the target cell. Membrane fusion is mediated by the viral attachment protein and the F protein. Cleavage of MuV-F into two subunits by furin is a prerequisite for fusion and virus infectivity. Here, we show that LAMPs support the furin-mediated cleavage of MuV-F. Expression levels of LAMPs affect the processing of MuV-F and MuV-mediated membrane fusion. Among LAMPs, the interferon-stimulated gene product LAMP3 is most critical in certain cells. Our study provides potential targets for anti-MuV therapeutics.


Asunto(s)
Furina/genética , Interacciones Huésped-Patógeno/genética , Proteínas de Membrana de los Lisosomas/genética , Lisosomas/virología , Virus de la Parotiditis/genética , Proteínas de Neoplasias/genética , Proteínas Virales de Fusión/genética , Células A549 , Animales , Membrana Celular/metabolismo , Membrana Celular/virología , Chlorocebus aethiops , Furina/metabolismo , Regulación de la Expresión Génica , Células Gigantes/química , Células Gigantes/metabolismo , Células HEK293 , Proteína HN/genética , Proteína HN/metabolismo , Células HeLa , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Virus de la Parotiditis/metabolismo , Proteínas de Neoplasias/metabolismo , Unión Proteica , Proteolisis , Transducción de Señal , Células Vero , Proteínas Virales de Fusión/metabolismo , Internalización del Virus
8.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31619562

RESUMEN

Mumps virus (MuV), an enveloped negative-strand RNA virus belonging to the family Paramyxoviridae, enters the host cell through membrane fusion mediated by two viral envelope proteins, an attachment protein hemagglutinin-neuraminidase (MuV-HN) and a fusion (F) protein. However, how the binding of MuV-HN to glycan receptors triggers membrane fusion is not well understood. The crystal structure of the MuV-HN head domain forms a tetramer (dimer of dimers) like other paramyxovirus attachment proteins. In the structure, a sulfate ion (SO42-) was found at the interface between two dimers, which may be replaced by a hydrogen phosphate ion (HPO42-) under physiological conditions. The anion is captured by the side chain of a positively charged arginine residue at position 139 of one monomer each from both dimers. Substitution of alanine or lysine for arginine at this position compromised the fusion support activity of MuV-HN without affecting its cell surface expression, glycan-receptor binding, and interaction with the F protein. Furthermore, the substitution appeared to affect the tetramer formation of the head domain as revealed by blue native-PAGE analysis. These results, together with our previous similar findings with the measles virus attachment protein head domain, suggest that the dimer-dimer interaction within the tetramer may play an important role in triggering membrane fusion during paramyxovirus entry.IMPORTANCE Despite the use of effective live vaccines, mumps outbreaks still occur worldwide. Mumps virus (MuV) infection typically causes flu-like symptoms and parotid gland swelling but sometimes leads to orchitis, oophoritis, and neurological complications, such as meningitis, encephalitis, and deafness. MuV enters the host cell through membrane fusion mediated by two viral proteins, a receptor-binding attachment protein, and a fusion protein, but its detailed mechanism is not fully understood. In this study, we show that the tetramer (dimer of dimers) formation of the MuV attachment protein head domain is supported by an anion located at the interface between two dimers and that the dimer-dimer interaction plays an important role in triggering the activation of the fusion protein and causing membrane fusion. These results not only further our understanding of MuV entry but provide useful information about a possible target for antiviral drugs.


Asunto(s)
Fusión de Membrana , Virus de la Parotiditis/metabolismo , Multimerización de Proteína , Proteínas Virales de Fusión/metabolismo , Acoplamiento Viral , Internalización del Virus , Sustitución de Aminoácidos , Células HEK293 , Humanos , Virus de la Parotiditis/genética , Mutación Missense , Fosfatos/metabolismo , Dominios Proteicos , Sulfatos/metabolismo , Proteínas Virales de Fusión/genética
9.
Proc Natl Acad Sci U S A ; 115(10): 2496-2501, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29463726

RESUMEN

Measles virus (MeV), a major cause of childhood morbidity and mortality, is highly immunotropic and one of the most contagious pathogens. MeV may establish, albeit rarely, persistent infection in the central nervous system, causing fatal and intractable neurodegenerative diseases such as subacute sclerosing panencephalitis and measles inclusion body encephalitis. Recent studies have suggested that particular substitutions in the MeV fusion (F) protein are involved in the pathogenesis by destabilizing the F protein and endowing it with hyperfusogenicity. Here we show the crystal structures of the prefusion MeV-F alone and in complex with the small compound AS-48 or a fusion inhibitor peptide. Notably, these independently developed inhibitors bind the same hydrophobic pocket located at the region connecting the head and stalk of MeV-F, where a number of substitutions in MeV isolates from neurodegenerative diseases are also localized. Since these inhibitors could suppress membrane fusion mediated by most of the hyperfusogenic MeV-F mutants, the development of more effective inhibitors based on the structures may be warranted to treat MeV-induced neurodegenerative diseases.


Asunto(s)
Antivirales , Péptidos , Proteínas Virales de Fusión , Animales , Antivirales/química , Antivirales/metabolismo , Sitios de Unión , Células CHO , Chlorocebus aethiops , Cricetulus , Modelos Moleculares , Péptidos/química , Péptidos/metabolismo , Células Vero , Proteínas Virales de Fusión/antagonistas & inhibidores , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo
10.
Curr Top Microbiol Immunol ; 392: 219-29, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26162566

RESUMEN

RNA viruses usually consist of mutant spectra because of high error rates of viral RNA polymerases. Growth competition occurs among different viral variants, and the fittest clones predominate under given conditions. Individual variants, however, may not be entirely independent of each other, and internal interactions within mutant spectra can occur. Examples of cooperative and interfering interactions that exert enhancing and suppressing effects on replication of the wild-type virus, respectively, have been described, but their underlying mechanisms have not been well defined. It was recently found that the cooperation between wild-type and variant measles virus genomes produces a new phenotype through the heterooligomer formation of a viral protein. This observation provides a molecular mechanism underlying cooperative interactions within mutant spectra. Careful attention to individual sequences, in addition to consensus sequences, may disclose further examples of internal interactions within mutant spectra.


Asunto(s)
Virus del Sarampión/fisiología , Mutación , Animales , Evolución Molecular , Humanos , Sarampión/virología , Virus del Sarampión/clasificación , Virus del Sarampión/genética , Modelos Genéticos , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
11.
J Virol ; 89(5): 2710-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25520515

RESUMEN

UNLABELLED: Subacute sclerosing panencephalitis (SSPE) is caused by persistent measles virus (MV) infection in the central nervous system (CNS). Since human neurons, its main target cells, do not express known MV receptors (signaling lymphocyte activation molecule [SLAM] and nectin 4), it remains to be understood how MV infects and spreads in them. We have recently reported that fusion-enhancing substitutions in the extracellular domain of the MV fusion (F) protein (T461I and S103I/N462S/N465S), which are found in multiple SSPE virus isolates, promote MV spread in human neuroblastoma cell lines and brains of suckling hamsters. In this study, we show that hyperfusogenic viruses with these substitutions also spread efficiently in human primary neuron cultures without inducing syncytia. These substitutions were found to destabilize the prefusion conformation of the F protein trimer, thereby enhancing fusion activity. However, these hyperfusogenic viruses exhibited stronger cytopathology and produced lower titers at later time points in SLAM- or nectin 4-expressing cells compared to the wild-type MV. Although these viruses spread efficiently in the brains of SLAM knock-in mice, they did not in the spleens. Taken together, the results suggest that enhanced fusion activity is beneficial for MV to spread in neuronal cells where no cytopathology occurs, but detrimental to other types of cells due to strong cytopathology. Acquisition of enhanced fusion activity through substitutions in the extracellular domain of the F protein may be crucial for MV's extensive spread in the CNS and development of SSPE. IMPORTANCE: Subacute sclerosing panencephalitis (SSPE) is a fatal disease caused by persistent measles virus (MV) infection in the central nervous system (CNS). Its cause is not well understood, and no effective therapy is currently available. Recently, we have reported that enhanced fusion activity of MV through the mutations in its fusion protein is a major determinant of efficient virus spread in human neuronal cells and brains of suckling hamsters. In this study, we show that those mutations render the conformation of the fusion protein less stable, thereby making it hyperfusogenic. Our results also show that enhanced fusion activity is beneficial for MV to spread in the CNS but detrimental to other types of cells in peripheral tissues, which are strongly damaged by the virus. Our findings provide important insight into the mechanism for the development of SSPE after MV infection.


Asunto(s)
Virus del Sarampión/fisiología , Neuronas/virología , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Animales , Células Cultivadas , Efecto Citopatogénico Viral , Humanos , Virus del Sarampión/genética , Ratones Transgénicos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Virales de Fusión/genética
12.
Rinsho Byori ; 64(9): 1057-1064, 2016 09.
Artículo en Japonés | MEDLINE | ID: mdl-30609459

RESUMEN

Human metapneumovirus (HMPV) was first isolated in 2001 from young children with symptoms of acute respiratory infections. Studies revealed that HMPV has circulated worldwide for more than 50 years in hu- man populations. HMPV is classified into two major, distinct groups, A and B, which show antigenic differ- ences, but clinical symptoms of infection with these groups are indistinguishable. The symptoms are often severe and similar to those of respiratory syncytial virus infections. HMPV is detected in more than 10% of children under five years old suffering from acute respiratory infections. Elderly people are also affected with HMPV and may develop severe respiratory diseases. Recently, point-of-care testing based on immu- nochromatography became available in Japan under the coverage of medical insurance, further revealing clini- cal pictures of HMPV infections and improving clinical treatment and control measures. Sensitive nucleo- tide amplification techniques are also available for the detection of HMPV. However, the virus titers and amounts of viral antigens decline significantly after 5 days of illness. Therefore, laboratory testing to detect HMPV antigens or genomes should be conducted using clinical specimens before 4 days of illness. Assays to detect immunoglobulin specific to HMPV (ELISA and neutralizing assay) have also been established, alt- hough they have not yet been approved as extracorporeal diagnostic medicines. [Review].


Asunto(s)
Metapneumovirus , Infecciones por Paramyxoviridae , Humanos , Metapneumovirus/aislamiento & purificación , Infecciones por Paramyxoviridae/epidemiología , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/transmisión , Infecciones por Paramyxoviridae/virología , Neumonía , Índice de Severidad de la Enfermedad , Vacunación
13.
J Biol Chem ; 288(12): 8085-8091, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23362271

RESUMEN

Measles virus (MV), an enveloped RNA virus belonging to the Paramyxoviridae family, enters the cell through membrane fusion mediated by two viral envelope proteins, an attachment protein hemagglutinin (H) and a fusion (F) protein. The crystal structure of the receptor-binding head domain of MV-H bound to its cellular receptor revealed that the MV-H head domain forms a tetrameric assembly (dimer of dimers), which occurs in two forms (forms I and II). In this study, we show that mutations in the putative dimer-dimer interface of the head domain in either form inhibit the ability of MV-H to support membrane fusion, without greatly affecting its cell surface expression, receptor binding, and interaction with the F protein. Notably, some anti-MV-H neutralizing monoclonal antibodies are directed to the region around the dimer-dimer interface in form I rather than receptor-binding sites. These observations suggest that the dimer-dimer interactions of the MV-H head domain, especially that in form I, contribute to triggering membrane fusion, and that conformational shift of head domain tetramers plays a role in the process. Furthermore, our results indicate that although the stalk and transmembrane regions may be mainly responsible for the tetramer formation of MV-H, the head domain alone can form tetramers, albeit at a low efficiency.


Asunto(s)
Hemaglutininas Virales/genética , Virus del Sarampión/genética , Mutación Missense , Internalización del Virus , Animales , Antígenos CD/metabolismo , Células CHO , Cricetinae , Células HEK293 , Hemaglutininas Virales/química , Hemaglutininas Virales/metabolismo , Humanos , Virus del Sarampión/fisiología , Proteína Cofactora de Membrana/metabolismo , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Receptores de Superficie Celular/metabolismo , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Proteínas Virales de Fusión/metabolismo
14.
J Virol ; 87(5): 2648-59, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23255801

RESUMEN

Subacute sclerosing panencephalitis (SSPE) is a fatal degenerative disease caused by persistent measles virus (MV) infection in the central nervous system (CNS). From the genetic study of MV isolates obtained from SSPE patients, it is thought that defects of the matrix (M) protein play a crucial role in MV pathogenicity in the CNS. In this study, we report several notable mutations in the extracellular domain of the MV fusion (F) protein, including those found in multiple SSPE strains. The F proteins with these mutations induced syncytium formation in cells lacking SLAM and nectin 4 (receptors used by wild-type MV), including human neuronal cell lines, when expressed together with the attachment protein hemagglutinin. Moreover, recombinant viruses with these mutations exhibited neurovirulence in suckling hamsters, unlike the parental wild-type MV, and the mortality correlated with their fusion activity. In contrast, the recombinant MV lacking the M protein did not induce syncytia in cells lacking SLAM and nectin 4, although it formed larger syncytia in cells with either of the receptors. Since human neuronal cells are mainly SLAM and nectin 4 negative, fusion-enhancing mutations in the extracellular domain of the F protein may greatly contribute to MV spread via cell-to-cell fusion in the CNS, regardless of defects of the M protein.


Asunto(s)
Antígenos CD/metabolismo , Encéfalo/virología , Moléculas de Adhesión Celular/metabolismo , Virus del Sarampión/fisiología , Neuronas/virología , Receptores de Superficie Celular/metabolismo , Proteínas Virales de Fusión/metabolismo , Sustitución de Aminoácidos , Animales , Antígenos CD/genética , Moléculas de Adhesión Celular/genética , Fusión Celular , Línea Celular , Chlorocebus aethiops , Cricetinae , Células Gigantes/virología , Humanos , Virus del Sarampión/genética , Proteínas Mutantes/metabolismo , Mutación , Neuronas/metabolismo , Receptores de Superficie Celular/genética , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Panencefalitis Esclerosante Subaguda/mortalidad , Panencefalitis Esclerosante Subaguda/virología , Células Vero , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética
15.
Sci Adv ; 9(4): eadf3731, 2023 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-36706187

RESUMEN

Measles virus (MeV), which is usually non-neurotropic, sometimes persists in the brain and causes subacute sclerosing panencephalitis (SSPE) several years after acute infection, serving as a model for persistent viral infections. The persisting MeVs have hyperfusogenic mutant fusion (F) proteins that likely enable cell-cell fusion at synapses and "en bloc transmission" between neurons. We here show that during persistence, F protein fusogenicity is generally enhanced by cumulative mutations, yet mutations paradoxically reducing the fusogenicity may be selected alongside the wild-type (non-neurotropic) MeV genome. A mutant F protein having SSPE-derived substitutions exhibits lower fusogenicity than the hyperfusogenic F protein containing some of those substitutions, but by the wild-type F protein coexpression, the fusogenicity of the former F protein is enhanced, while that of the latter is nearly abolished. These findings advance the understanding of the long-term process of MeV neuropathogenicity and provide critical insight into the genotype-phenotype relationships of en bloc transmitted viruses.


Asunto(s)
Panencefalitis Esclerosante Subaguda , Humanos , Panencefalitis Esclerosante Subaguda/genética , Panencefalitis Esclerosante Subaguda/metabolismo , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Virus del Sarampión/genética , Encéfalo/metabolismo , Mutación
16.
J Biol Chem ; 285(27): 20882-90, 2010 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-20435897

RESUMEN

Measles virus (MV), an enveloped negative-strand RNA virus, remains a major cause of morbidity and mortality in developing countries. MV predominantly infects immune cells by using signaling lymphocyte activation molecule (SLAM; also called CD150) as a receptor, but it also infects polarized epithelial cells, forming tight junctions in a SLAM-independent manner. Although the ability of MV to infect polarized epithelial cells is thought to be important for its transmission, the epithelial cell receptor for MV has not been identified. A transcriptional repressor, Snail, induces epithelial-mesenchymal transition (EMT), in which epithelial cells lose epithelial cell phenotypes, such as adherens and tight junctions. In this study, EMT was induced by expressing Snail in a lung adenocarcinoma cell line, II-18, which is highly susceptible to wild-type MV. Snail-expressing II-18 cells lost adherens and tight junctions. Microarray analysis confirmed the induction of EMT in II-18 cells and suggested a novel function of Snail in protein degradation and distribution. Importantly, wild-type MV no longer entered EMT-induced II-18 cells, suggesting that the epithelial cell receptor is down-regulated by the induction of EMT. Other polarized cell lines, NCI-H358 and HT-29, also lost susceptibility to wild-type MV when EMT was induced. However, the complete formation of tight junctions rather reduced MV entry into HT-29 cells. Taken together, these data suggest that the unidentified epithelial cell receptor for MV is involved in the formation of epithelial intercellular junctions.


Asunto(s)
Células Epiteliales/citología , Virus del Sarampión/patogenicidad , Sarampión/prevención & control , Mesodermo/citología , Animales , Línea Celular , Susceptibilidad a Enfermedades , Células Epiteliales/fisiología , Células Epiteliales/virología , Citometría de Flujo , Predisposición Genética a la Enfermedad , Vectores Genéticos , Genoma Viral , Humanos , Activación de Linfocitos , Macaca mulatta/virología , Sarampión/inmunología , Sarampión/transmisión , Sarampión/veterinaria , Virus del Sarampión/genética , Proteína Cofactora de Membrana/fisiología , Mesodermo/fisiología , Mesodermo/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , Plásmidos , Receptores Virales/fisiología , Uniones Estrechas/fisiología , Esparcimiento de Virus
17.
J Virol ; 83(20): 10374-83, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19656884

RESUMEN

The genome of measles virus (MV) is encapsidated by the nucleocapsid (N) protein and associates with RNA-dependent RNA polymerase to form the ribonucleoprotein complex. The matrix (M) protein is believed to play an important role in MV assembly by linking the ribonucleoprotein complex with envelope glycoproteins. Analyses using a yeast two-hybrid system and coimmunoprecipitation in mammalian cells revealed that the M protein interacts with the N protein and that two leucine residues at the carboxyl terminus of the N protein (L523 and L524) are critical for the interaction. In MV minigenome reporter gene assays, the M protein inhibited viral RNA synthesis only when it was able to interact with the N protein. The N protein colocalized with the M protein at the plasma membrane when the proteins were coexpressed in plasmid-transfected or MV-infected cells. In contrast, the N protein formed small dots in the perinuclear area when it was expressed without the M protein, or it was incapable of interacting with the M protein. Furthermore, a recombinant MV possessing a mutant N protein incapable of interacting with the M protein grew much less efficiently than the parental virus. Since the M protein has an intrinsic ability to associate with the plasma membrane, it may retain the ribonucleoprotein complex at the plasma membrane by binding to the N protein, thereby stopping viral RNA synthesis and promoting viral particle production. Consequently, our results indicate that the M protein regulates MV RNA synthesis and assembly via its interaction with the N protein.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus del Sarampión/metabolismo , Proteínas de la Nucleocápside/metabolismo , ARN Viral/metabolismo , Proteínas de la Matriz Viral/metabolismo , Ensamble de Virus , Animales , Línea Celular , Chlorocebus aethiops , Células HeLa , Humanos , Virus del Sarampión/genética , Proteínas de la Nucleocápside/genética , ARN Viral/genética , Técnicas del Sistema de Dos Híbridos , Células Vero , Proteínas de la Matriz Viral/genética
18.
J Virol ; 82(9): 4630-7, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18287234

RESUMEN

Measles is one of the most contagious human infectious diseases and remains a major cause of childhood morbidity and mortality worldwide. The signaling lymphocyte activation molecule (SLAM), also called CD150, is a cellular receptor for measles virus (MV), presumably accounting for its tropism for immune cells and its immunosuppressive properties. On the other hand, pathological studies have shown that MV also infects epithelial cells at a later stage of infection, although its mechanism has so far been unknown. In this study, we show that wild-type MV can infect and produce syncytia in human polarized epithelial cell lines independently of SLAM and CD46 (a receptor for the vaccine strains of MV). Progeny viral particles are released exclusively from the apical surface of these polarized epithelial cell lines. We have also identified amino acid residues on the MV attachment protein that are likely to interact with a putative receptor on epithelial cells. All of these residues have aromatic side chains and may form a receptor-binding pocket located in a different position from the putative SLAM- and CD46-binding sites on the MV attachment protein. Thus, our results indicate that MV has an intrinsic ability to infect both polarized epithelial and immune cells by using distinctive receptor-binding sites on the attachment protein corresponding to each of their respective receptors. The ability of MV to infect polarized epithelial cells and its exclusive release from the apical surface may facilitate its efficient transmission via aerosol droplets, resulting in its highly contagious nature.


Asunto(s)
Células Epiteliales/virología , Hemaglutininas/metabolismo , Linfocitos/virología , Virus del Sarampión/patogenicidad , Animales , Antígenos CD , Sitios de Unión , Línea Celular , Polaridad Celular , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/virología , Proteína Cofactora de Membrana , Receptores de Superficie Celular , Receptores Virales/metabolismo , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Acoplamiento Viral
19.
J Virol ; 82(17): 8296-306, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18562542

RESUMEN

Measles is an acute febrile infectious disease with high morbidity and mortality. The genome of measles virus (MV), the causative agent, encodes two accessory products, V and C proteins, that play important roles in MV virulence. The V but not the C protein of the IC-B strain (a well-characterized virulent strain of MV) has been shown to block the Jak/Stat signaling pathway and counteract the cellular interferon (IFN) response. We have recently shown that a recombinant IC-B strain that lacks C protein expression replicates poorly in certain cell lines, and its growth defect is related to translational inhibition and strong IFN induction. Here, we show that the V protein of the MV IC-B strain also blocks the IFN induction pathway mediated by the melanoma differentiation-associated gene 5 product, thus actively interfering with the host IFN response at two different steps. On the other hand, the C protein per se possesses no activity to block the IFN induction pathway. Our data indicate that the C protein acts as a regulator of viral RNA synthesis, thereby acting indirectly to suppress IFN induction. Since recombinant MVs with C protein defective in modulating viral RNA synthesis or lacking C protein expression strongly stimulate IFN production, in spite of V protein production, both the C and V proteins must be required for MV to fully circumvent the host IFN response.


Asunto(s)
Interferones/metabolismo , Virus del Sarampión/fisiología , Fosfoproteínas/fisiología , Proteínas Virales/fisiología , Animales , Chlorocebus aethiops , Técnica del Anticuerpo Fluorescente Directa , Regulación Viral de la Expresión Génica , Genes Reporteros , Luciferasas de Renilla/metabolismo , Virus del Sarampión/genética , Plásmidos , Biosíntesis de Proteínas/fisiología , Células Vero , Proteínas Virales/química
20.
J Virol ; 82(23): 11979-84, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18799577

RESUMEN

Live attenuated vaccines against measles have been developed through adaptation of clinical isolates of measles virus (MV) in various cultured cells. Analyses using recombinant MVs with chimeric genomes between wild-type and Edmonston vaccine strains indicated that viruses possessing the polymerase protein genes of the Edmonston strain exhibited attenuated viral gene expression and growth in cultured cells as well as in mice expressing an MV receptor, signaling lymphocyte activation molecule, regardless of whether the virus genome had the wild-type or vaccine-type promoter sequence. These data demonstrate that the polymerase protein genes of the Edmonston strain contribute to its attenuated phenotype.


Asunto(s)
Antígenos CD/fisiología , Vacuna Antisarampión/inmunología , Virus del Sarampión/genética , ARN Polimerasa Dependiente del ARN/genética , Receptores de Superficie Celular/fisiología , Vacunas Sintéticas/inmunología , Animales , Antígenos CD/genética , Chlorocebus aethiops , Proteínas Fluorescentes Verdes/genética , Ratones , ARN Viral/biosíntesis , ARN Polimerasa Dependiente del ARN/inmunología , Receptores de Superficie Celular/genética , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Vacunas Atenuadas/inmunología , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA