Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 22(1): 25-31, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33154590

RESUMEN

Clinical manifestations of COVID-19 caused by the new coronavirus SARS-CoV-2 are associated with age1,2. Adults develop respiratory symptoms, which can progress to acute respiratory distress syndrome (ARDS) in the most severe form, while children are largely spared from respiratory illness but can develop a life-threatening multisystem inflammatory syndrome (MIS-C)3-5. Here, we show distinct antibody responses in children and adults after SARS-CoV-2 infection. Adult COVID-19 cohorts had anti-spike (S) IgG, IgM and IgA antibodies, as well as anti-nucleocapsid (N) IgG antibody, while children with and without MIS-C had reduced breadth of anti-SARS-CoV-2-specific antibodies, predominantly generating IgG antibodies specific for the S protein but not the N protein. Moreover, children with and without MIS-C had reduced neutralizing activity as compared to both adult COVID-19 cohorts, indicating a reduced protective serological response. These results suggest a distinct infection course and immune response in children independent of whether they develop MIS-C, with implications for developing age-targeted strategies for testing and protecting the population.


Asunto(s)
Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , COVID-19/inmunología , Proteínas de la Nucleocápside/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Adolescente , Adulto , Anciano , COVID-19/virología , Niño , Preescolar , Femenino , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Masculino , Persona de Mediana Edad , SARS-CoV-2/fisiología , Adulto Joven
2.
J Virol ; 98(3): e0187423, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38329336

RESUMEN

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.


Asunto(s)
Adenosina Monofosfato , Alanina , Virus del Sarampión , Sarampión , Panencefalitis Esclerosante Subaguda , Proteínas Virales , Preescolar , Humanos , Adenosina Monofosfato/administración & dosificación , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/uso terapéutico , Alanina/administración & dosificación , Alanina/análogos & derivados , Alanina/uso terapéutico , Autopsia , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/virología , Progresión de la Enfermedad , Resultado Fatal , Genoma Viral/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Sarampión/complicaciones , Sarampión/tratamiento farmacológico , Sarampión/virología , Virus del Sarampión/efectos de los fármacos , Virus del Sarampión/genética , Virus del Sarampión/metabolismo , Proteínas Mutantes/análisis , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Calidad de Vida , ARN Viral/análisis , ARN Viral/genética , Panencefalitis Esclerosante Subaguda/tratamiento farmacológico , Panencefalitis Esclerosante Subaguda/etiología , Panencefalitis Esclerosante Subaguda/virología , Proteínas Virales/análisis , Proteínas Virales/genética , Proteínas Virales/metabolismo
3.
PLoS Biol ; 18(9): e3000849, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32898168

RESUMEN

Despite limited genomic diversity, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has shown a wide range of clinical manifestations in different patient populations. The mechanisms behind these host differences are still unclear. Here, we examined host response gene expression across infection status, viral load, age, and sex among shotgun RNA sequencing profiles of nasopharyngeal (NP) swabs from 430 individuals with PCR-confirmed SARS-CoV-2 and 54 negative controls. SARS-CoV-2 induced a strong antiviral response with up-regulation of antiviral factors such as OAS1-3 and IFIT1-3 and T helper type 1 (Th1) chemokines CXCL9/10/11, as well as a reduction in transcription of ribosomal proteins. SARS-CoV-2 culture in human airway epithelial (HAE) cultures replicated the in vivo antiviral host response 7 days post infection, with no induction of interferon-stimulated genes after 3 days. Patient-matched longitudinal specimens (mean elapsed time = 6.3 days) demonstrated reduction in interferon-induced transcription, recovery of transcription of ribosomal proteins, and initiation of wound healing and humoral immune responses. Expression of interferon-responsive genes, including ACE2, increased as a function of viral load, while transcripts for B cell-specific proteins and neutrophil chemokines were elevated in patients with lower viral load. Older individuals had reduced expression of the Th1 chemokines CXCL9/10/11 and their cognate receptor CXCR3, as well as CD8A and granzyme B, suggesting deficiencies in trafficking and/or function of cytotoxic T cells and natural killer (NK) cells. Relative to females, males had reduced B cell-specific and NK cell-specific transcripts and an increase in inhibitors of nuclear factor kappa-B (NF-κB) signaling, possibly inappropriately throttling antiviral responses. Collectively, our data demonstrate that host responses to SARS-CoV-2 are dependent on viral load and infection time course, with observed differences due to age and sex that may contribute to disease severity.


Asunto(s)
Antivirales/inmunología , Betacoronavirus/fisiología , Infecciones por Coronavirus/inmunología , Neumonía Viral/inmunología , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , COVID-19 , Niño , Preescolar , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/virología , Femenino , Regulación de la Expresión Génica , Humanos , Inmunidad/genética , Cinética , Masculino , Persona de Mediana Edad , Nasofaringe/inmunología , Nasofaringe/virología , Pandemias , Neumonía Viral/epidemiología , Neumonía Viral/virología , Proteínas Ribosómicas/genética , SARS-CoV-2 , Factores Sexuales , Transducción de Señal/genética , Carga Viral , Cicatrización de Heridas/genética , Adulto Joven
4.
PLoS Pathog ; 16(9): e1008883, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32956394

RESUMEN

Infection by human parainfluenza viruses (HPIVs) causes widespread lower respiratory diseases, including croup, bronchiolitis, and pneumonia, and there are no vaccines or effective treatments for these viruses. HPIV3 is a member of the Respirovirus species of the Paramyxoviridae family. These viruses are pleomorphic, enveloped viruses with genomes composed of single-stranded negative-sense RNA. During viral entry, the first step of infection, the viral fusion complex, comprised of the receptor-binding glycoprotein hemagglutinin-neuraminidase (HN) and the fusion glycoprotein (F), mediates fusion upon receptor binding. The HPIV3 transmembrane protein HN, like the receptor-binding proteins of other related viruses that enter host cells using membrane fusion, binds to a receptor molecule on the host cell plasma membrane, which triggers the F glycoprotein to undergo major conformational rearrangements, promoting viral entry. Subsequent fusion of the viral and host membranes allows delivery of the viral genetic material into the host cell. The intermediate states in viral entry are transient and thermodynamically unstable, making it impossible to understand these transitions using standard methods, yet understanding these transition states is important for expanding our knowledge of the viral entry process. In this study, we use cryo-electron tomography (cryo-ET) to dissect the stepwise process by which the receptor-binding protein triggers F-mediated fusion, when forming a complex with receptor-bearing membranes. Using an on-grid antibody capture method that facilitates examination of fresh, biologically active strains of virus directly from supernatant fluids and a series of biological tools that permit the capture of intermediate states in the fusion process, we visualize the series of events that occur when a pristine, authentic viral particle interacts with target receptors and proceeds from the viral entry steps of receptor engagement to membrane fusion.


Asunto(s)
Membrana Celular/metabolismo , Proteína HN/metabolismo , Virus de la Parainfluenza 3 Humana/metabolismo , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Animales , Membrana Celular/ultraestructura , Chlorocebus aethiops , Humanos , Virus de la Parainfluenza 3 Humana/ultraestructura , Células Vero
5.
J Am Chem Soc ; 143(15): 5958-5966, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33825470

RESUMEN

The lower respiratory tract infections affecting children worldwide are in large part caused by the parainfluenza viruses (HPIVs), particularly HPIV3, along with human metapneumovirus and respiratory syncytial virus, enveloped negative-strand RNA viruses. There are no vaccines for these important human pathogens, and existing treatments have limited or no efficacy. Infection by HPIV is initiated by viral glycoprotein-mediated fusion between viral and host cell membranes. A viral fusion protein (F), once activated in proximity to a target cell, undergoes a series of conformational changes that first extend the trimer subunits to allow insertion of the hydrophobic domains into the target cell membrane and then refold the trimer into a stable postfusion state, driving the merger of the viral and host cell membranes. Lipopeptides derived from the C-terminal heptad repeat (HRC) domain of HPIV3 F inhibit infection by interfering with the structural transitions of the trimeric F assembly. Clinical application of this strategy, however, requires improving the in vivo stability of antiviral peptides. We show that the HRC peptide backbone can be modified via partial replacement of α-amino acid residues with ß-amino acid residues to generate α/ß-peptides that retain antiviral activity but are poor protease substrates. Relative to a conventional α-lipopeptide, our best α/ß-lipopeptide exhibits improved persistence in vivo and improved anti-HPIV3 antiviral activity in animals.


Asunto(s)
Lipopéptidos/farmacología , Virus de la Parainfluenza 3 Humana/efectos de los fármacos , Infecciones del Sistema Respiratorio/patología , Secuencia de Aminoácidos , Aminoácidos/química , Aminoácidos/metabolismo , Animales , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , Línea Celular , Colesterol/química , Diseño de Fármacos , Humanos , Lipopéptidos/química , Lipopéptidos/metabolismo , Virus de la Parainfluenza 3 Humana/aislamiento & purificación , Multimerización de Proteína , Ratas , Infecciones del Sistema Respiratorio/virología , Distribución Tisular , Temperatura de Transición , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Internalización del Virus/efectos de los fármacos
6.
J Am Chem Soc ; 142(5): 2140-2144, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31951396

RESUMEN

Human parainfluenza virus 3 (HPIV3) and respiratory syncytial virus (RSV) are leading causes of lower respiratory tract infections. There are currently no vaccines or antiviral therapeutics to treat HPIV3 or RSV infections. We recently reported a peptide (VIQKI), derived from the C-terminal heptad repeat (HRC) domain of the HPIV3 fusion (F) glycoprotein that inhibits infection by both HPIV3 and RSV. The dual inhibitory activity of VIQKI is due to its unique ability to bind to the N-terminal heptad repeat (HRN) domains of both HPIV3 and RSV F, thereby preventing the native HRN-HRC interactions required for viral entry. Here we describe the structure-guided design of dual inhibitors of HPIV3 and RSV fusion with improved efficacy. We show that VIQKI derivatives possessing one (I456F) or two (I454F/I456F) phenylalanine substitutions near the N-terminus exhibit more stable assemblies with the RSV-HRN domain and enhanced antiviral efficacy against both HPIV3 and RSV infection. Cocrystal structures of the new Phe-substituted inhibitors coassembled with HPIV3 or RSV-HRN domains reveal that the I456F substitution makes intimate hydrophobic contact with the core trimers of both HPIV3 and RSV F.


Asunto(s)
Antivirales/farmacología , Oligopéptidos/farmacología , Virus de la Parainfluenza 3 Humana/efectos de los fármacos , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Secuencia de Aminoácidos , Antivirales/química , Cristalografía por Rayos X , Interacciones Hidrofóbicas e Hidrofílicas , Estructura Molecular , Oligopéptidos/química , Virus de la Parainfluenza 3 Humana/fisiología , Conformación Proteica , Virus Sincitial Respiratorio Humano/fisiología
7.
J Virol ; 93(4)2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30487282

RESUMEN

During a measles virus (MeV) epidemic in 2009 in South Africa, measles inclusion body encephalitis (MIBE) was identified in several HIV-infected patients. Years later, children are presenting with subacute sclerosing panencephalitis (SSPE). To investigate the features of established MeV neuronal infections, viral sequences were analyzed from brain tissue samples of a single SSPE case and compared with MIBE sequences previously obtained from patients infected during the same epidemic. Both the SSPE and the MIBE viruses had amino acid substitutions in the ectodomain of the F protein that confer enhanced fusion properties. Functional analysis of the fusion complexes confirmed that both MIBE and SSPE F protein mutations promoted fusion with less dependence on interaction by the viral receptor-binding protein with known MeV receptors. While the SSPE F required the presence of a homotypic attachment protein, MeV H, in order to fuse, MIBE F did not. Both F proteins had decreased thermal stability compared to that of the corresponding wild-type F protein. Finally, recombinant viruses expressing MIBE or SSPE fusion complexes spread in the absence of known MeV receptors, with MIBE F-bearing viruses causing large syncytia in these cells. Our results suggest that alterations to the MeV fusion complex that promote fusion and cell-to-cell spread in the absence of known MeV receptors is a key property for infection of the brain.IMPORTANCE Measles virus can invade the central nervous system (CNS) and cause severe neurological complications, such as MIBE and SSPE. However, mechanisms by which MeV enters the CNS and triggers the disease remain unclear. We analyzed viruses from brain tissue of individuals with MIBE or SSPE, infected during the same epidemic, after the onset of neurological disease. Our findings indicate that the emergence of hyperfusogenic MeV F proteins is associated with infection of the brain. We also demonstrate that hyperfusogenic F proteins permit MeV to enter cells and spread without the need to engage nectin-4 or CD150, known receptors for MeV that are not present on neural cells.


Asunto(s)
Virus del Sarampión/genética , Panencefalitis Esclerosante Subaguda/genética , Proteínas Virales de Fusión/genética , Sustitución de Aminoácidos , Animales , Encéfalo/virología , Moléculas de Adhesión Celular/metabolismo , Chlorocebus aethiops , Epidemias , Femenino , Genotipo , Células Gigantes/virología , Células HEK293 , Humanos , Masculino , Sarampión/epidemiología , Sarampión/metabolismo , Sarampión/virología , Mutación , Neuronas/virología , Sudáfrica , Panencefalitis Esclerosante Subaguda/virología , Células Vero , Proteínas Virales de Fusión/metabolismo
8.
J Virol ; 93(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30728259

RESUMEN

A clinical isolate of measles virus (MeV) bearing a single amino acid alteration in the viral fusion protein (F; L454W) was previously identified in two patients with lethal sequelae of MeV central nervous system (CNS) infection. The mutation dysregulated the viral fusion machinery so that the mutated F protein mediated cell fusion in the absence of known MeV cellular receptors. While this virus could feasibly have arisen via intrahost evolution of the wild-type (wt) virus, it was recently shown that the same mutation emerged under the selective pressure of small-molecule antiviral treatment. Under these conditions, a potentially neuropathogenic variant emerged outside the CNS. While CNS adaptation of MeV was thought to generate viruses that are less fit for interhost spread, we show that two animal models can be readily infected with CNS-adapted MeV via the respiratory route. Despite bearing a fusion protein that is less stable at 37°C than the wt MeV F, this virus infects and replicates in cotton rat lung tissue more efficiently than the wt virus and is lethal in a suckling mouse model of MeV encephalitis even with a lower inoculum. Thus, either during lethal MeV CNS infection or during antiviral treatment in vitro, neuropathogenic MeV can emerge, can infect new hosts via the respiratory route, and is more pathogenic (at least in these animal models) than wt MeV.IMPORTANCE Measles virus (MeV) infection can be severe in immunocompromised individuals and lead to complications, including measles inclusion body encephalitis (MIBE). In some cases, MeV persistence and subacute sclerosing panencephalitis (SSPE) occur even in the face of an intact immune response. While they are relatively rare complications of MeV infection, MIBE and SSPE are lethal. This work addresses the hypothesis that despite a dysregulated viral fusion complex, central nervous system (CNS)-adapted measles virus can spread outside the CNS within an infected host.


Asunto(s)
Sistema Nervioso Central/virología , Encefalitis Viral , Cuerpos de Inclusión Viral , Pulmón/virología , Virus del Sarampión/fisiología , Sarampión , Mutación Missense , Proteínas Virales de Fusión , Replicación Viral , Sustitución de Aminoácidos , Animales , Sistema Nervioso Central/metabolismo , Chlorocebus aethiops , Modelos Animales de Enfermedad , Encefalitis Viral/genética , Encefalitis Viral/metabolismo , Encefalitis Viral/transmisión , Humanos , Cuerpos de Inclusión Viral/genética , Cuerpos de Inclusión Viral/metabolismo , Pulmón/metabolismo , Sarampión/metabolismo , Sarampión/transmisión , Ratones , Ratones Transgénicos , Sigmodontinae , Células Vero , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo
9.
J Am Chem Soc ; 141(32): 12648-12656, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31268705

RESUMEN

Human parainfluenza virus 3 (HPIV3) and respiratory syncytial virus (RSV) cause lower respiratory infection in infants and young children. There are no vaccines for these pathogens, and existing treatments have limited or questionable efficacy. Infection by HPIV3 or RSV requires fusion of the viral and cell membranes, a process mediated by a trimeric fusion glycoprotein (F) displayed on the viral envelope. Once triggered, the pre-fusion form of F undergoes a series of conformational changes that first extend the molecule to allow for insertion of the hydrophobic fusion peptide into the target cell membrane and then refold the trimeric assembly into an energetically stable post-fusion state, a process that drives the merger of the viral and host cell membranes. Peptides derived from defined regions of HPIV3 F inhibit infection by HPIV3 by interfering with the structural transitions of the trimeric F assembly. Here we describe lipopeptides derived from the C-terminal heptad repeat (HRC) domain of HPIV3 F that potently inhibit infection by both HPIV3 and RSV. The lead peptide inhibits RSV infection as effectively as does a peptide corresponding to the RSV HRC domain itself. We show that the inhibitors bind to the N-terminal heptad repeat (HRN) domains of both HPIV3 and RSV F with high affinity. Co-crystal structures of inhibitors bound to the HRN domains of HPIV3 or RSV F reveal remarkably different modes of binding in the N-terminal segment of the inhibitor.


Asunto(s)
Lipopéptidos/farmacología , Virus de la Parainfluenza 3 Humana/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Virus Sincitiales Respiratorios/efectos de los fármacos , Inhibidores de Proteínas Virales de Fusión/farmacología , Proteínas Virales de Fusión/farmacología , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Lipopéptidos/metabolismo , Pruebas de Sensibilidad Microbiana , Virus de la Parainfluenza 3 Humana/química , Fragmentos de Péptidos/metabolismo , Unión Proteica , Mucosa Respiratoria/virología , Virus Sincitiales Respiratorios/química , Inhibidores de Proteínas Virales de Fusión/metabolismo , Proteínas Virales de Fusión/metabolismo , Internalización del Virus/efectos de los fármacos
10.
J Infect Dis ; 218(2): 218-227, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29566184

RESUMEN

Background: The emerging zoonotic paramyxovirus Nipah virus (NiV) causes severe respiratory and neurological disease in humans, with high fatality rates. Nipah virus can be transmitted via person-to-person contact, posing a high risk for epidemic outbreaks. However, a broadly applicable approach for human NiV outbreaks in field settings is lacking. Methods: We engineered new antiviral lipopeptides and analyzed in vitro fusion inhibition to identify an optimal candidate for prophylaxis of NiV infection in the lower respiratory tract, and we assessed antiviral efficiency in 2 different animal models. Results: We show that lethal NiV infection can be prevented with lipopeptides delivered via the respiratory route in both hamsters and nonhuman primates. By targeting retention of peptides for NiV prophylaxis in the respiratory tract, we avoid its systemic delivery in individuals who need only prevention, and thus we increase the safety of treatment and enhance utility of the intervention. Conclusions: The experiments provide a proof of concept for the use of antifusion lipopeptides for prophylaxis of lethal NiV. These results advance the goal of rational development of potent lipopeptide inhibitors with desirable pharmacokinetic and biodistribution properties and a safe effective delivery method to target NiV and other pathogenic viruses.


Asunto(s)
Quimioprevención/métodos , Infecciones por Henipavirus/prevención & control , Lipopéptidos/administración & dosificación , Virus Nipah/fisiología , Enfermedades de los Primates/prevención & control , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Inhibidores de Proteínas Virales de Fusión/administración & dosificación , Animales , Bronconeumonía/prevención & control , Bronconeumonía/veterinaria , Chlorocebus aethiops , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Mesocricetus
11.
Molecules ; 22(11)2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-29088094

RESUMEN

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.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Hidroxicolesteroles/química , Virus del Sarampión/patogenicidad , Proteínas Virales de Fusión/química , Antivirales/metabolismo , Fusión Celular , Dispersión Dinámica de Luz , Eritrocitos/efectos de los fármacos , Humanos , Resonancia por Plasmón de Superficie , Liposomas Unilamelares/metabolismo , Proteínas Virales de Fusión/genética
12.
Molecules ; 22(7)2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28714870

RESUMEN

A set of lipopeptides was recently reported for their broad-spectrum antiviral activity against viruses belonging to the Paramyxoviridae family, including human parainfluenza virus type 3 and Nipah virus. Among them, the peptide with a 24-unit PEG linker connecting it to a cholesterol moiety (VG-PEG24-Chol) was found to be the best membrane fusion inhibitory peptide. Here, we evaluated the interaction of the same set of peptides with biomembrane model systems and isolated human peripheral blood mononuclear cells (PBMC). VG-PEG24-Chol showed the highest insertion rate and it was among the peptides that induced a larger change on the surface pressure of cholesterol rich membranes. This peptide also displayed a high affinity towards PBMC membranes. These data provide new information about the dynamics of peptide-membrane interactions of a specific group of antiviral peptides, known for their potential as multipotent paramyxovirus antivirals.


Asunto(s)
Antivirales/química , Membrana Celular/química , Lipopéptidos/química , Polietilenglicoles/química , Antivirales/metabolismo , Antivirales/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Colesterol/química , Humanos , Leucocitos Mononucleares/química , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Lipopéptidos/metabolismo , Lipopéptidos/farmacología , Liposomas/química , Paramyxovirinae/química , Relación Estructura-Actividad
13.
J Virol ; 88(22): 13495-502, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25210187

RESUMEN

UNLABELLED: Human parainfluenza viruses (HPIVs) cause widespread respiratory infections, with no vaccines or effective treatments. We show that the molecular determinants for HPIV3 growth in vitro are fundamentally different from those required in vivo and that these differences impact inhibitor susceptibility. HPIV infects its target cells by coordinated action of the hemagglutinin-neuraminidase receptor-binding protein (HN) and the fusion envelope glycoprotein (F), which together comprise the molecular fusion machinery; upon receptor engagement by HN, the prefusion F undergoes a structural transition, extending and inserting into the target cell membrane and then refolding into a postfusion structure that fuses the viral and cell membranes. Peptides derived from key regions of F can potently inhibit HPIV infection at the entry stage, by interfering with the structural transition of F. We show that clinically circulating viruses have fusion machinery that is more stable and less readily activated than viruses adapted to growth in culture. Fusion machinery that is advantageous for growth in human airway epithelia and in vivo confers susceptibility to peptide fusion inhibitors in the host lung tissue or animal, but the same fusion inhibitors have no effect on viruses whose fusion glycoproteins are suited for growth in vitro. We propose that for potential clinical efficacy, antivirals should be evaluated using clinical isolates in natural host tissue rather than lab strains of virus in cultured cells. The unique susceptibility of clinical strains in human tissues reflects viral inhibition in vivo. IMPORTANCE: Acute respiratory infection is the leading cause of mortality in young children under 5 years of age, causing nearly 20% of childhood deaths worldwide each year. The paramyxoviruses, including human parainfluenza viruses (HPIVs), cause a large share of these illnesses. There are no vaccines or drugs for the HPIVs. Inhibiting entry of viruses into the human cell is a promising drug strategy that blocks the first step in infection. To develop antivirals that inhibit entry, it is critical to understand the first steps of infection. We found that clinical viruses isolated from patients have very different entry properties from those of the viruses generally studied in laboratories. The viral entry mechanism is less active and more sensitive to fusion inhibitory molecules. We propose that to interfere with viral infection, we test clinically circulating viruses in natural tissues, to develop antivirals against respiratory disease caused by HPIVs.


Asunto(s)
Virus de la Parainfluenza 3 Humana/fisiología , Internalización del Virus , Animales , Células Cultivadas , Femenino , Proteína HN/metabolismo , Humanos , Sigmodontinae , Proteínas Virales de Fusión/metabolismo
14.
J Antimicrob Chemother ; 69(5): 1286-97, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24464268

RESUMEN

OBJECTIVES: The aim of the present work was to evaluate the interaction of two new HIV fusion inhibitors {HIVP3 [C34-polyethylene glycol (PEG)4-cholesterol] and HIVP4 [(C34-PEG4)2-cholesterol]} with membrane model systems and human blood cells in order to clarify where and how the fusion inhibitors locate, allowing us to understand their mechanism of action at the molecular level, and which strategies may be followed to increase efficacy. METHODS: Lipid vesicles with defined compositions were used for peptide partition and localization studies, based on the intrinsic fluorescence of HIVP3 and HIVP4. Lipid monolayers were employed in surface pressure studies. Finally, human erythrocytes and peripheral blood mononuclear cells (PBMCs) isolated from blood samples were used in dipole potential assays. RESULTS: Membrane partition, dipole potential and surface pressure assays indicate that the new fusion inhibitors interact preferentially with cholesterol-rich liquid-ordered membranes, mimicking biological membrane microdomains known as lipid rafts. HIVP3 and HIVP4 are able to interact with human erythrocytes and PBMCs to a similar degree as a previously described simpler drug with monomeric C34 and lacking the PEG spacer, C34-cholesterol. However, the pocket-binding domain (PBD) of both HIVP3 and HIVP4 is more exposed to the aqueous environment than in C34-cholesterol. CONCLUSIONS: The present data allow us to conclude that more efficient blocking of HIV entry results from the synergism between the membranotropic behaviour and the enhanced exposure of the PBD.


Asunto(s)
Proteína gp41 de Envoltorio del VIH/farmacología , Inhibidores de Fusión de VIH/farmacología , Fragmentos de Péptidos/farmacología , Membrana Celular/metabolismo , Eritrocitos/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Unión Proteica
15.
J Virol ; 87(24): 13619-27, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24109225

RESUMEN

Paramyxoviruses, including the human pathogen measles virus (MV) and the avian Newcastle disease virus (NDV), enter host cells through fusion of the viral envelope with the target cell membrane. This fusion is driven by the concerted action of two viral envelope glycoproteins: the receptor binding protein and the fusion protein (F). The MV receptor binding protein (hemagglutinin [H]) attaches to proteinaceous receptors on host cells, while the receptor binding protein of NDV (hemagglutinin-neuraminidase [HN]) interacts with sialic acid-containing receptors. The receptor-bound HN/H triggers F to undergo conformational changes that render it competent to mediate fusion of the viral and cellular membranes. The mechanism of fusion activation has been proposed to be different for sialic acid-binding viruses and proteinaceous receptor-binding viruses. We report that a chimeric protein containing the NDV HN receptor binding region and the MV H stalk domain can activate MV F to fuse, suggesting that the signal to the stalk of a protein-binding receptor binding molecule can be transmitted from a sialic acid binding domain. By engineering the NDV HN globular domain to interact with a proteinaceous receptor, the fusion activation signal was preserved. Our findings are consistent with a unified mechanism of fusion activation, at least for the Paramyxovirinae subfamily, in which the receptor binding domains of the receptor binding proteins are interchangeable and the stalk determines the specificity of F activation.


Asunto(s)
Proteína HN/metabolismo , Hemaglutininas/metabolismo , Virus del Sarampión/metabolismo , Sarampión/virología , Ácido N-Acetilneuramínico/metabolismo , Enfermedad de Newcastle/virología , Virus de la Enfermedad de Newcastle/metabolismo , Animales , Aves , Línea Celular , Proteína HN/química , Proteína HN/genética , Hemaglutininas/química , Hemaglutininas/genética , Humanos , Sarampión/genética , Sarampión/metabolismo , Virus del Sarampión/química , Virus del Sarampión/genética , Enfermedad de Newcastle/genética , Enfermedad de Newcastle/metabolismo , Virus de la Enfermedad de Newcastle/química , Virus de la Enfermedad de Newcastle/genética , Unión Proteica , Estructura Terciaria de Proteína , Receptores Virales/genética , Receptores Virales/metabolismo , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo , Internalización del Virus
16.
J Virol ; 87(24): 13785-94, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24109233

RESUMEN

Measles virus (MV) infection causes an acute childhood disease that can include infection of the central nervous system and can rarely progress to severe neurological disease for which there is no specific treatment. We generated potent antiviral peptide inhibitors of MV entry and spreading and MV-induced cell fusion. Dimers of MV-specific peptides derived from the C-terminal heptad repeat region of the MV fusion protein, conjugated to cholesterol, efficiently protect SLAM transgenic mice from fatal MV infection. Fusion inhibitors hold promise for the prophylaxis of MV infection in unvaccinated and immunocompromised people, as well as potential for the treatment of grave neurological complications of measles.


Asunto(s)
Antivirales/farmacología , Encéfalo/virología , Virus del Sarampión/efectos de los fármacos , Sarampión/prevención & control , Proteínas Virales de Fusión/antagonistas & inhibidores , Animales , Encéfalo/efectos de los fármacos , Línea Celular , Humanos , Sarampión/tratamiento farmacológico , Sarampión/mortalidad , Sarampión/virología , Virus del Sarampión/genética , Virus del Sarampión/fisiología , Ratones , Ratones Transgénicos , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Internalización del Virus/efectos de los fármacos
17.
J Virol ; 87(20): 10980-96, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23903846

RESUMEN

Paramyxoviruses, including the emerging lethal human Nipah virus (NiV) and the avian Newcastle disease virus (NDV), enter host cells through fusion of the viral and target cell membranes. For paramyxoviruses, membrane fusion is the result of the concerted action of two viral envelope glycoproteins: a receptor binding protein and a fusion protein (F). The NiV receptor binding protein (G) attaches to ephrin B2 or B3 on host cells, whereas the corresponding hemagglutinin-neuraminidase (HN) attachment protein of NDV interacts with sialic acid moieties on target cells through two regions of its globular domain. Receptor-bound G or HN via its stalk domain triggers F to undergo the conformational changes that render it competent to mediate fusion of the viral and cellular membranes. We show that chimeric proteins containing the NDV HN receptor binding regions and the NiV G stalk domain require a specific sequence at the connection between the head and the stalk to activate NiV F for fusion. Our findings are consistent with a general mechanism of paramyxovirus fusion activation in which the stalk domain of the receptor binding protein is responsible for F activation and a specific connecting region between the receptor binding globular head and the fusion-activating stalk domain is required for transmitting the fusion signal.


Asunto(s)
Virus Nipah/fisiología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Línea Celular , Análisis Mutacional de ADN , Humanos , Virus de la Enfermedad de Newcastle/genética , Virus Nipah/genética , Mapeo de Interacción de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas del Envoltorio Viral/genética
18.
Science ; 384(6703): eadm8693, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38935733

RESUMEN

Measles virus (MeV) presents a public health threat that is escalating as vaccine coverage in the general population declines and as populations of immunocompromised individuals, who cannot be vaccinated, increase. There are no approved therapeutics for MeV. Neutralizing antibodies targeting viral fusion are one potential therapeutic approach but have not yet been structurally characterized or advanced to clinical use. We present cryo-electron microscopy (cryo-EM) structures of prefusion F alone [2.1-angstrom (Å) resolution], F complexed with a fusion-inhibitory peptide (2.3-Å resolution), F complexed with the neutralizing and protective monoclonal antibody (mAb) 77 (2.6-Å resolution), and an additional structure of postfusion F (2.7-Å resolution). In vitro assays and examination of additional EM classes show that mAb 77 binds prefusion F, arrests F in an intermediate state, and prevents transition to the postfusion conformation. These structures shed light on antibody-mediated neutralization that involves arrest of fusion proteins in an intermediate state.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Microscopía por Crioelectrón , Virus del Sarampión , Proteínas Virales de Fusión , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/química , Virus del Sarampión/inmunología , Virus del Sarampión/química , Proteínas Virales de Fusión/inmunología , Proteínas Virales de Fusión/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/química , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/química , Humanos , Conformación Proteica
19.
J Biol Chem ; 287(1): 778-793, 2012 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-22110138

RESUMEN

Parainfluenza viruses enter host cells by fusing the viral and target cell membranes via concerted action of their two envelope glycoproteins: the hemagglutinin-neuraminidase (HN) and the fusion protein (F). Receptor-bound HN triggers F to undergo conformational changes that render it fusion-competent. To address the role of receptor engagement and to elucidate how HN and F interact during the fusion process, we used bimolecular fluorescence complementation to follow the dynamics of human parainfluenza virus type 3 (HPIV3) HN/F pairs in living cells. We show that HN and F associate before receptor engagement. HN drives the formation of HN-F clusters at the site of fusion, and alterations in HN-F interaction determine the fusogenicity of the glycoprotein pair. An interactive site, at the HN dimer interface modulates HN fusion activation property, which is critical for infection of the natural host. This first evidence for the sequence of initial events that lead to viral entry may indicate a new paradigm for understanding Paramyxovirus infection.


Asunto(s)
Hemaglutininas Virales/metabolismo , Neuraminidasa/metabolismo , Virus de la Parainfluenza 3 Humana/fisiología , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Células HEK293 , Hemaglutininas Virales/química , Hemaglutininas Virales/genética , Humanos , Modelos Moleculares , Imagen Molecular , Mutación , Neuraminidasa/química , Neuraminidasa/genética , Virus de la Parainfluenza 3 Humana/metabolismo , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Receptores de Superficie Celular/metabolismo , Espectrometría de Fluorescencia , Especificidad por Sustrato
20.
J Virol ; 86(10): 5730-41, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22438532

RESUMEN

The hemagglutinin-neuraminidase (HN) protein of paramyxoviruses carries out three distinct activities contributing to the ability of HN to promote viral fusion and entry: receptor binding, receptor cleavage (neuraminidase), and activation of the fusion protein. The relationship between receptor binding and fusion triggering functions of HN are not fully understood. For Newcastle disease virus (NDV), one bifunctional site (site I) on HN's globular head can mediate both receptor binding and neuraminidase activities, and a second site (site II) in the globular head is also capable of mediating receptor binding. The receptor analog, zanamivir, blocks receptor binding and cleavage activities of NDV HN's site I while activating receptor binding by site II. Comparison of chimeric proteins in which the globular head of NDV HN is connected to the stalk region of either human parainfluenza virus type 3 (HPIV3) or Nipah virus receptor binding proteins indicates that receptor binding to NDV HN site II not only can activate its own fusion (F) protein but can also activate the heterotypic fusion proteins. We suggest a general model for paramyxovirus fusion activation in which receptor engagement at site II plays an active role in F activation.


Asunto(s)
Proteína HN/metabolismo , Virus de la Enfermedad de Newcastle/fisiología , Infecciones por Paramyxoviridae/metabolismo , Paramyxovirinae/metabolismo , Receptores Virales/metabolismo , Proteínas Virales/metabolismo , Internalización del Virus , Sitios de Unión , Proteínas Portadoras , Línea Celular , Proteína HN/química , Proteína HN/genética , Humanos , Virus de la Enfermedad de Newcastle/química , Virus de la Enfermedad de Newcastle/enzimología , Virus de la Enfermedad de Newcastle/genética , Infecciones por Paramyxoviridae/virología , Paramyxovirinae/genética , Estructura Terciaria de Proteína , Proteínas Virales de Fusión/metabolismo , Proteínas Virales/química , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA