Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.110
Filtrar
1.
J Virol ; 98(5): e0195923, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38634598

RESUMEN

The role of Culex mosquitoes in the transmission of Japanese encephalitis virus (JEV) is crucial, yet the mechanisms of JEV infection in these vectors remain unclear. Previous research has indicated that various host factors participate in JEV infection. Herein, we present evidence that mosquito sialic acids enhance JEV infection both in vivo and in vitro. By treating mosquitoes and C6/36 cells with neuraminidase or lectin, the function of sialic acids is effectively blocked, resulting in significant inhibition of JEV infection. Furthermore, knockdown of the sialic acid biosynthesis genes in Culex mosquitoes also leads to a reduction in JEV infection. Moreover, our research revealed that sialic acids play a role in the attachment of JEV to mosquito cells, but not in its internalization. To further explore the mechanisms underlying the promotion of JEV attachment by sialic acids, we conducted immunoprecipitation experiments to confirm the direct binding of sialic acids to the last α-helix in JEV envelope protein domain III. Overall, our study contributes to a molecular comprehension of the interaction between mosquitoes and JEV and offers potential strategies for preventing the dissemination of flavivirus in natural environments.IMPORTANCEIn this study, we aimed to investigate the impact of glycoconjugate sialic acids on mosquito infection with Japanese encephalitis virus (JEV). Our findings demonstrate that sialic acids play a crucial role in enhancing JEV infection by facilitating the attachment of the virus to the cell membrane. Furthermore, our investigation revealed that sialic acids directly bind to the final α-helix in the JEV envelope protein domain III, thereby accelerating virus adsorption. Collectively, our results highlight the significance of mosquito sialic acids in JEV infection within vectors, contributing to a better understanding of the interaction between mosquitoes and JEV.


Asunto(s)
Culex , Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa , Mosquitos Vectores , Ácidos Siálicos , Acoplamiento Viral , Virus de la Encefalitis Japonesa (Especie)/fisiología , Virus de la Encefalitis Japonesa (Especie)/metabolismo , Animales , Culex/virología , Culex/metabolismo , Encefalitis Japonesa/virología , Encefalitis Japonesa/metabolismo , Mosquitos Vectores/virología , Ácidos Siálicos/metabolismo , Línea Celular , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/genética , Internalización del Virus , Ratones , Neuraminidasa/metabolismo , Neuraminidasa/genética
2.
J Virol ; 98(5): e0004724, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38651898

RESUMEN

RNA viruses lack proofreading in their RNA polymerases and therefore exist as genetically diverse populations. By exposing these diverse viral populations to selective pressures, viruses with mutations that confer fitness advantages can be enriched. To examine factors important for viral tropism and host restriction, we passaged murine norovirus (MNV) in a human cell line, HeLa cells, to select mutant viruses with increased fitness in non-murine cells. A major determinant of host range is expression of the MNV receptor CD300lf on mouse cells, but additional host factors may limit MNV replication in human cells. We found that viruses passaged six times in HeLa cells had enhanced replication compared with the parental virus. The passaged viruses had several mutations throughout the viral genome, which were primarily located in the viral non-structural coding regions. Although viral attachment was not altered for the passaged viruses, their replication was higher than the parental virus when the entry was bypassed, suggesting that the mutant viruses overcame a post-entry block in human cells. Three mutations in the viral NS1 protein were sufficient for enhanced post-entry replication in human cells. We found that the human cell-adapted MNV variants had reduced fitness in murine BV2 cells and infected mice, with reduced viral titers. These results suggest a fitness tradeoff, where increased fitness in a non-native host cell reduces fitness in a natural host environment. Overall, this work suggests that MNV tropism is determined by the presence of not only the viral receptor but also post-entry factors. IMPORTANCE: Viruses infect specific species and cell types, which is dictated by the expression of host factors required for viral entry as well as downstream replication steps. Murine norovirus (MNV) infects mouse cells, but not human cells. However, human cells expressing the murine CD300lf receptor support MNV replication, suggesting that receptor expression is a major determinant of MNV tropism. To determine whether other factors influence MNV tropism, we selected for variants with enhanced replication in human cells. We identified mutations that enhance MNV replication in human cells and demonstrated that these mutations enhance infection at a post-entry replication step. Therefore, MNV infection of human cells is restricted at both entry and post-entry stages. These results shed new light on factors that influence viral tropism and host range.


Asunto(s)
Especificidad del Huésped , Mutación , Norovirus , Tropismo Viral , Internalización del Virus , Replicación Viral , Norovirus/genética , Norovirus/fisiología , Humanos , Animales , Ratones , Células HeLa , Infecciones por Caliciviridae/virología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Genoma Viral , Receptores Virales/metabolismo , Receptores Virales/genética , Acoplamiento Viral
3.
Viruses ; 16(4)2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38675866

RESUMEN

Gu-Sui-Bu, the dried rhizome of Davallia mariesii, is a traditional Chinese herbal remedy with a significant history of treating osteoporosis and inflammatory conditions. However, its potential as an anti-influenza agent and its underlying mechanisms of action remain unexplored. To obtain a more potent extract from D. mariesii and gain insights into its mechanism of action against influenza A virus (IAV), we utilized a partitioning process involving organic solvents and water, resulting in the isolation of butanolic subfractions of the D. mariesii extract (DMBE). DMBE exhibited a broad anti-viral spectrum, effectively inhibiting IAV, with an EC50 of 24.32 ± 6.19 µg/mL and a selectivity index of 6.05. We subsequently conducted a series of in vitro assays to evaluate the antiviral effects of DMBE and to uncover its mechanisms of action. DMBE was found to inhibit IAV during the early stages of infection by hindering the attachment of the virus onto and its penetration into host cells. Importantly, DMBE was observed to hinder IAV-mediated cell-cell fusion. It also inhibited neuraminidase activity, plaque size, and the expression levels of phospho-AKT. In summary, this study provides evidence for the effectiveness of D. mariesii as a complementary and alternative herbal remedy against IAV. Specifically, our data highlight DMBE's capabilities in inhibiting viral entry and the release of virions.


Asunto(s)
Antivirales , Virus de la Influenza A , Extractos Vegetales , Antivirales/farmacología , Antivirales/química , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/fisiología , Humanos , Extractos Vegetales/farmacología , Extractos Vegetales/química , Animales , Células de Riñón Canino Madin Darby , Perros , Internalización del Virus/efectos de los fármacos , Sapindaceae/química , Replicación Viral/efectos de los fármacos , Acoplamiento Viral/efectos de los fármacos , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/química , Neuraminidasa/metabolismo , Células A549 , Línea Celular
4.
Viruses ; 16(4)2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38675987

RESUMEN

Consistent with the biochemistry of coronaviruses as well established over decades, SARS-CoV-2 makes its initial attachment to host cells through the binding of its spike protein (SP) to sialylated glycans (containing the monosaccharide sialic acid) on the cell surface. The virus can then slide over and enter via ACE2. SARS-CoV-2 SP attaches particularly tightly to the trillions of red blood cells (RBCs), platelets and endothelial cells in the human body, each cell very densely coated with sialic acid surface molecules but having no ACE2 or minimal ACE2. These interlaced attachments trigger the blood cell aggregation, microvascular occlusion and vascular damage that underlie the hypoxia, blood clotting and related morbidities of severe COVID-19. Notably, the two human betacoronaviruses that express a sialic acid-cleaving enzyme are benign, while the other three-SARS, SARS-CoV-2 and MERS-are virulent. RBC aggregation experimentally induced in several animal species using an injected polysaccharide caused most of the same morbidities of severe COVID-19. This glycan biochemistry is key to disentangling controversies that have arisen over the efficacy of certain generic COVID-19 treatment agents and the safety of SP-based COVID-19 vaccines. More broadly, disregard for the active physiological role of RBCs yields unreliable or erroneous reporting of pharmacokinetic parameters as routinely obtained for most drugs and other bioactive agents using detection in plasma, with whole-blood levels being up to 30-fold higher. Appreciation of the active role of RBCs can elucidate the microvascular underpinnings of other health conditions, including cardiovascular disease, and therapeutic opportunities to address them.


Asunto(s)
COVID-19 , Polisacáridos , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Humanos , COVID-19/metabolismo , COVID-19/virología , SARS-CoV-2/metabolismo , Polisacáridos/metabolismo , Animales , Glicoproteína de la Espiga del Coronavirus/metabolismo , Betacoronavirus/metabolismo , Infecciones por Coronavirus/metabolismo , Eritrocitos/metabolismo , Eritrocitos/virología , Pandemias , Microvasos/metabolismo , Microvasos/virología , Acoplamiento Viral , Tratamiento Farmacológico de COVID-19 , Células Endoteliales/metabolismo , Células Endoteliales/virología , Enzima Convertidora de Angiotensina 2/metabolismo , Agregación Eritrocitaria
5.
Nat Microbiol ; 9(4): 905-921, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38528146

RESUMEN

Some viruses are rarely transmitted orally or sexually despite their presence in saliva, breast milk, or semen. We previously identified that extracellular vesicles (EVs) in semen and saliva inhibit Zika virus infection. However, the antiviral spectrum and underlying mechanism remained unclear. Here we applied lipidomics and flow cytometry to show that these EVs expose phosphatidylserine (PS). By blocking PS receptors, targeted by Zika virus in the process of apoptotic mimicry, they interfere with viral attachment and entry. Consequently, physiological concentrations of EVs applied in vitro efficiently inhibited infection by apoptotic mimicry dengue, West Nile, Chikungunya, Ebola and vesicular stomatitis viruses, but not severe acute respiratory syndrome coronavirus 2, human immunodeficiency virus 1, hepatitis C virus and herpesviruses that use other entry receptors. Our results identify the role of PS-rich EVs in body fluids in innate defence against infection via viral apoptotic mimicries, explaining why these viruses are primarily transmitted via PS-EV-deficient blood or blood-ingesting arthropods rather than direct human-to-human contact.


Asunto(s)
Líquidos Corporales , Vesículas Extracelulares , Virus , Infección por el Virus Zika , Virus Zika , Femenino , Humanos , Fosfatidilserinas , Acoplamiento Viral
6.
Microb Pathog ; 190: 106628, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38508422

RESUMEN

Rotavirus infections in suckling and weaning piglets cause severe dehydration and death, resulting in significant economic losses in the pig breeding industry. With the continuous emergence of porcine rotavirus (PoRV) variants and poor vaccine cross-protection among various genotypes, there is an urgent need to develop alternative strategies such as seeking effective antiviral products from nature, microbial metabolites and virus-host protein interaction. Sialidases play a crucial role in various physiopathological processes and offer a promising target for developing antivirus drugs. However, the effect of bacterial-derived sialidases on the infection of PoRVs remains largely unknown. Herein, we investigated the impact of bacterial-derived sialidases (sialidase Cp and Vc) on PoRV strain OSU(Group A) infection, using differentiated epithelial monkey kidney cells (MA104) as a model. Our results indicated that the pretreatment of MA104 with exogenous sialidases effectively suppressed PoRV OSU in a concentration-dependent manner. Notably, even at a concentration of 0.01 µU/mL, sialidases significantly inhibited the virus (MOI = 0.01). Meanwhile, we found that sialidase Vc pretreatment sharply reduced the binding rate of PoRV OSU. Last, we demonstrated that PoRV OSU might recognize α-2,3-linked sialic acid as the primary attachment factor in MA104. Our findings provide new insights into the underlying mechanism of PoRV OSU infections, shedding lights on the development of alternative antivirus approaches based on bacteria-virus interaction.


Asunto(s)
Neuraminidasa , Infecciones por Rotavirus , Rotavirus , Replicación Viral , Animales , Neuraminidasa/metabolismo , Neuraminidasa/genética , Rotavirus/efectos de los fármacos , Rotavirus/fisiología , Porcinos , Replicación Viral/efectos de los fármacos , Línea Celular , Células Epiteliales/virología , Células Epiteliales/microbiología , Acoplamiento Viral/efectos de los fármacos , Ácido N-Acetilneuramínico/metabolismo , Ácido N-Acetilneuramínico/farmacología , Antivirales/farmacología , Haplorrinos , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/microbiología
7.
Sci Immunol ; 9(93): eadj9534, 2024 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-38517951

RESUMEN

Antigenic drift, the gradual accumulation of amino acid substitutions in the influenza virus hemagglutinin (HA) receptor protein, enables viral immune evasion. Antibodies (Abs) specific for the drift-resistant HA stem region are a promising universal influenza vaccine target. Although anti-stem Abs are not believed to block viral attachment, here we show that complement component 1q (C1q), a 460-kilodalton protein with six Ab Fc-binding domains, confers attachment inhibition to anti-stem Abs and enhances their fusion and neuraminidase inhibition. As a result, virus neutralization activity in vitro is boosted up to 30-fold, and in vivo protection from influenza PR8 infection in mice is enhanced. These effects reflect increased steric hindrance and not increased Ab avidity. C1q greatly expands the anti-stem Ab viral escape repertoire to include residues throughout the HA, some of which cause antigenic alterations in the globular region or modulate HA receptor avidity. We also show that C1q enhances the neutralization activity of non-receptor binding domain anti-SARS-CoV-2 spike Abs, an effect dependent on spike density on the virion surface. These findings demonstrate that C1q can greatly expand Ab function and thereby contribute to viral evolution and immune escape.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Ratones , Animales , Humanos , Hemaglutininas , Complemento C1q , Acoplamiento Viral , Glicoproteínas Hemaglutininas del Virus de la Influenza , Anticuerpos Antivirales
8.
Cell Mol Life Sci ; 81(1): 71, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38300320

RESUMEN

Hexosylceramides (HexCer) are implicated in the infection process of various pathogens. However, the molecular and cellular functions of HexCer in infectious cycles are poorly understood. Investigating the enveloped virus Uukuniemi (UUKV), a bunyavirus of the Phenuiviridae family, we performed a lipidomic analysis with mass spectrometry and determined the lipidome of both infected cells and derived virions. We found that UUKV alters the processing of HexCer to glycosphingolipids (GSL) in infected cells. The infection resulted in the overexpression of glucosylceramide (GlcCer) synthase (UGCG) and the specific accumulation of GlcCer and its subsequent incorporation into viral progeny. UUKV and several pathogenic bunyaviruses relied on GlcCer in the viral envelope for binding to various host cell types. Overall, our results indicate that GlcCer is a structural determinant of virions crucial for bunyavirus infectivity. This study also highlights the importance of glycolipids on virions in facilitating interactions with host cell receptors and infectious entry of enveloped viruses.


Asunto(s)
Orthobunyavirus , Glucosilceramidas , Acoplamiento Viral , Lipidómica , Espectrometría de Masas
9.
Nat Struct Mol Biol ; 31(3): 447-454, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38233573

RESUMEN

Hepatitis B virus (HBV), a leading cause of developing hepatocellular carcinoma affecting more than 290 million people worldwide, is an enveloped DNA virus specifically infecting hepatocytes. Myristoylated preS1 domain of the HBV large surface protein binds to the host receptor sodium-taurocholate cotransporting polypeptide (NTCP), a hepatocellular bile acid transporter, to initiate viral entry. Here, we report the cryogenic-electron microscopy structure of the myristoylated preS1 (residues 2-48) peptide bound to human NTCP. The unexpectedly folded N-terminal half of the peptide embeds deeply into the outward-facing tunnel of NTCP, whereas the C-terminal half formed extensive contacts on the extracellular surface. Our findings reveal an unprecedented induced-fit mechanism for establishing high-affinity virus-host attachment and provide a blueprint for the rational design of anti-HBV drugs targeting virus entry.


Asunto(s)
Virus de la Hepatitis B , Simportadores , Humanos , Virus de la Hepatitis B/genética , Hepatocitos/metabolismo , Unión Proteica , Acoplamiento Viral , Péptidos/metabolismo , Simportadores/metabolismo , Internalización del Virus
10.
Virol Sin ; 39(2): 277-289, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38246238

RESUMEN

Influenza A virus (IAV) binds sialic acid receptors on the cell surface to enter the host cells, which is the key step in initiating infection, transmission and pathogenesis. Understanding the factors that contribute to the highly efficient entry of IAV into human cells will help elucidate the mechanism of viral entry and pathogenicity, and provide new targets for intervention. In the present study, we reported a novel membrane protein, C1QTNF5, which binds to the hemagglutinin protein of IAV and promotes IAV infection in vitro and in vivo. We found that the HA1 region of IAV hemagglutinin is critical for the interaction with C1QTNF5 protein, and C1QTNF5 interacts with hemagglutinin mainly through its N-terminus (1-103 aa). In addition, we further demonstrated that overexpression of C1QTNF5 promotes IAV entry, while blocking the interaction between C1QTNF5 and IAV hemagglutinin greatly inhibits viral entry. However, C1QTNF5 does not function as a receptor to mediate IAV infection in sialic acid-deficient CHO-Lec2 cells, but promotes IAV to attach to these cells, suggesting that C1QTNF5 is an important attachment factor for IAV. This work reveals C1QTNF5 as a novel IAV attachment factor and provides a new perspective for antiviral strategies.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza , Virus de la Influenza A , Acoplamiento Viral , Internalización del Virus , Humanos , Animales , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Virus de la Influenza A/genética , Virus de la Influenza A/fisiología , Células CHO , Cricetulus , Receptores Virales/metabolismo , Receptores Virales/genética , Células HEK293 , Ratones , Unión Proteica , Infecciones por Orthomyxoviridae/virología , Gripe Humana/virología , Gripe Humana/genética , Gripe Humana/metabolismo , Células A549
11.
Vet Microbiol ; 287: 109913, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38006719

RESUMEN

Japanese encephalitis virus (JEV) is a flavivirus that is spread through mosquito bites and is the leading cause of viral encephalitis in Asia. JEV can infect a variety of cell types; however, crucial receptor molecules remain unclear. The purpose of this study was to determine whether porcine CD4 protein is a receptor protein that impacts JEV entry into PK15 cells and subsequent viral replication. We confirmed the interaction between the JEV E protein and the CD4 protein through Co-IP, virus binding and internalization, antibody blocking, and overexpression and created a PK-15 cell line with CD4 gene knockdown by CRISPR/Cas9. The results show that CD4 interacts with JEV E and that CD4 knockdown cells altered virus adsorption and internalization, drastically reducing virus attachment. The level of viral transcription in CD4 antibody-blocked cells, vs. control cells, was decreased by 49.1%. Based on these results, we believe that CD4 is a receptor protein for JEVs. Furthermore, most viral receptors appear to be associated with lipid rafts, and colocalization studies demonstrate the presence of CD4 protein on lipid rafts. RT‒qPCR and WB results show that virus replication was suppressed in PK-15-CD4KD cells. The difference in viral titer between KD and WT PK-15 cells peaked at 24 h, and the viral titer in WT PK-15 cells was 5.6 × 106, whereas in PK-15-CD4KD cells, it was only 1.8 × 106, a 64% drop, demonstrating that CD4 deficiency has an effect on the process of viral replication. These findings suggest that JEV enters porcine kidney cells via lipid raft-colocalized CD4, and the proliferation process is positively correlated with CD4.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa , Receptores Virales , Enfermedades de los Porcinos , Animales , Asia , Línea Celular , Virus de la Encefalitis Japonesa (Especie)/fisiología , Encefalitis Japonesa/metabolismo , Encefalitis Japonesa/veterinaria , Encefalitis Japonesa/virología , Receptores Virales/metabolismo , Porcinos , Enfermedades de los Porcinos/virología , Acoplamiento Viral , Replicación Viral
12.
Front Immunol ; 14: 1259237, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37920471

RESUMEN

Introduction: Glucose Regulated Proteins/Binding protein (GRP78/Bip), a representative molecular chaperone, effectively influences and actively participates in the replication processes of many viruses. Little is known, however, about the functional involvement of GRP78 in the replication of Newcastle disease virus (NDV) and the underlying mechanisms. Methods: The method of this study are to establish protein interactomes between host cell proteins and the NDV Hemagglutinin-neuraminidase (HN) protein, and to systematically investigate the regulatory role of the GRP78-HN protein interaction during the NDV replication cycle. Results: Our study revealed that GRP78 is upregulated during NDV infection, and its direct interaction with HN is mediated by the N-terminal 326 amino acid region. Knockdown of GRP78 by small interfering RNAs (siRNAs) significantly suppressed NDV infection and replication. Conversely, overexpression of GRP78 resulted in a significant increase in NDV replication, demonstrating its role as a positive regulator in the NDV replication cycle. We further showed that the direct interaction between GRP78 and HN protein enhanced the attachment of NDV to cells, and masking of GRP78 expressed on the cell surface with specific polyclonal antibodies (pAbs) inhibited NDV attachment and replication. Discussion: These findings highlight the essential role of GRP78 in the adsorption stage during the NDV infection cycle, and, importantly, identify the critical domain required for GRP78-HN interaction, providing novel insights into the molecular mechanisms involved in NDV replication and infection.


Asunto(s)
Chaperón BiP del Retículo Endoplásmico , Virus de la Enfermedad de Newcastle , Animales , Neuraminidasa/metabolismo , Hemaglutininas , Acoplamiento Viral , Proteína HN/genética , Proteína HN/metabolismo , Proteína HN/farmacología , Proteínas Virales/farmacología
13.
Infect Genet Evol ; 116: 105516, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37924857

RESUMEN

Nipah virus (NiV) is a lethal bat-borne zoonotic virus that causes mild to acute respiratory distress and neurological manifestations in humans with a high mortality rate. NiV transmission to humans occurs via consumption of bat-contaminated fruit and date palm sap (DPS), or through direct contact with infected individuals and livestock. Since NiV outbreaks were first reported in pigs from Malaysia and Singapore, non-neutralizing antibodies against NiV attachment Glycoprotein (G) have also been detected in a few domestic mammals. NiV infection is initiated after NiV G binds to the host cell receptors Ephrin-B2 and Ephrin-B3. In this study, we assessed the degree of NiV host tropism in domestic and peridomestic mammals commonly found in Bangladesh that may be crucial in the transmission of NiV by serving as intermediate hosts. We carried out a protein-protein docking analysis of NiV G complexes (n = 52) with Ephrin-B2 and B3 of 13 domestic and peridomestic species using bioinformatics tools. Protein models were generated by homology modelling and the structures were validated for model quality. The different protein-protein complexes in this study were stable, and their binding affinity (ΔG) scores ranged between -8.0 to -19.1 kcal/mol. NiV Bangladesh (NiV-B) strain displayed stronger binding to Ephrin receptors, especially with Ephrin-B3 than the NiV Malaysia (NiV-M) strain, correlating with the observed higher pathogenicity of NiV-B strains. From the docking result, we found that Ephrin receptors of domestic rat (R. norvegicus) had a higher binding affinity for NiV G, suggesting greater susceptibility to NiV infections compared to other study species. Investigations for NiV exposure to domestic/peridomestic animals will help us knowing more the possible role of rats and other animals as intermediate hosts of NiV and would improve future NiV outbreak control and prevention in humans and domestic animals.


Asunto(s)
Quirópteros , Infecciones por Henipavirus , Virus Nipah , Animales , Ratas , Efrina-B2/genética , Efrina-B2/química , Efrina-B2/metabolismo , Efrina-B3/química , Efrina-B3/metabolismo , Glicoproteínas/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de la Familia Eph/metabolismo , Porcinos , Acoplamiento Viral
14.
ACS Nano ; 17(23): 23317-23330, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37982733

RESUMEN

Antivirals are indispensable tools that can be targeted at viral domains directly or at cellular domains indirectly to obstruct viral infections and reduce pathogenicity. Despite their transformative use in healthcare, antivirals have been clinically approved to treat only 10 of the more than 200 known pathogenic human viruses. Additionally, many virus functions are intimately coupled with host cellular processes, which presents challenges in antiviral development due to the limited number of clear targets per virus, necessitating extensive insight into these molecular processes. Compounding this challenge, many viral pathogens have evolved to evade effective antivirals. We hypothesize that a viral attachment blocking chimera (VirABloC) composed of a viral binder and a bulky scaffold that sterically blocks interactions between a viral particle and a host cell may be suitable for the development of antivirals that are agnostic to the extravirion epitope that is being bound. We test this hypothesis by modifying a nanobody that specifically recognizes a nonessential epitope presented on the extravirion surface of pseudorabies virus strain 486 with a 3-dimensional wireframe DNA origami structure ∼100 nm in diameter. The nanobody switches from having no inhibitory properties to 4.2 ± 0.9 nM IC50 when conjugated with the DNA origami scaffold. Mechanistic studies support that inhibition is mediated by the noncovalent attachment of the DNA origami scaffold to the virus particle, which obstructs the attachment of the viruses onto host cells. These results support the potential of VirABloC as a generalizable approach to developing antivirals.


Asunto(s)
Herpesvirus Suido 1 , Virus , Animales , Humanos , Herpesvirus Suido 1/genética , Acoplamiento Viral , ADN , Epítopos , Antivirales
15.
J Virol ; 97(10): e0083223, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37796128

RESUMEN

IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of several B cell malignancies and Kaposi's sarcoma. We analyzed the function of K8.1, the major antigenic component of the KSHV virion in the infection of different cells. To do this, we deleted K8.1 from the viral genome. It was found that K8.1 is critical for the infection of certain epithelial cells, e.g., a skin model cell line but not for infection of many other cells. K8.1 was found to mediate attachment of the virus to cells where it plays a role in infection. In contrast, we did not find K8.1 or a related protein from a closely related monkey virus to activate fusion of the viral and cellular membranes, at least not under the conditions tested. These findings suggest that K8.1 functions in a highly cell-specific manner during KSHV entry, playing a crucial role in the attachment of KSHV to, e.g., skin epithelial cells.


Asunto(s)
Glicoproteínas , Herpesvirus Humano 8 , Queratinocitos , Proteínas Virales , Acoplamiento Viral , Internalización del Virus , Humanos , Glicoproteínas/deficiencia , Glicoproteínas/genética , Glicoproteínas/metabolismo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiología , Queratinocitos/metabolismo , Queratinocitos/virología , Sarcoma de Kaposi/virología , Proteínas Virales/genética , Proteínas Virales/metabolismo , Fusión de Membrana , Piel/citología
16.
Front Cell Infect Microbiol ; 13: 1180344, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37577376

RESUMEN

Nipah virus (NiV) and Hendra virus (HeV) are newly emerging dangerous zoonotic pathogens of the Henipavirus genus of the Paramyxoviridae family. NiV and HeV (HNVs) which are transmitted by bats cause acute respiratory disease and fatal encephalitis in humans. To date, as there is a lack of antiviral drugs or effective antiviral therapies, the development of vaccines against those two viruses is of primary importance, and the immunogen design is crucial to the success of vaccines. In this study, the full-length protein (G), the ectodomain (Ge) and the head domain (Gs) of NiV attachment glycoprotein were delivered by the replication-defective type 5 adenovirus vector (Ad5) respectively, and the recombinant Ad5-NiV vaccine candidates (Ad5-NiVG, Ad5-NiVGe and Ad5-NiVGs) were constructed and their immunogenicity were evaluated in mice. The results showed that all the vaccine candidates stimulated specific humoral and cellular immune responses efficiently and rapidly against both NiV and HeV, and the Ad5-NiVGe elicited the strongest immune responses after a single-dose immunization. Furthermore, the potent conserved T-cell epitope DTLYFPAVGFL shared by NiV and HeV was identified in the study, which may provide valid information on the mechanism of HNVs-specific cellular immunity. In summary, this study demonstrates that the Ad5-NiVGe could be a potent vaccine candidate against HNVs by inducing robust humoral and cellular immune responses.


Asunto(s)
Virus Hendra , Virus Nipah , Humanos , Animales , Ratones , Virus Hendra/fisiología , Virus Nipah/genética , Virus Nipah/metabolismo , Acoplamiento Viral , Glicoproteínas/genética , Glicoproteínas/metabolismo , Vacunas Sintéticas , Inmunidad Celular , Adenoviridae/genética
17.
Viruses ; 15(7)2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37515193

RESUMEN

Influenza A viruses (IAVs) initiate infection via binding of the viral hemagglutinin (HA) to sialylated glycans on host cells. HA's receptor specificity towards individual glycans is well studied and clearly critical for virus infection, but the contribution of the highly heterogeneous and complex glycocalyx to virus-cell adhesion remains elusive. Here, we use two complementary methods, glycan arrays and single-virus force spectroscopy (SVFS), to compare influenza virus receptor specificity with virus binding to live cells. Unexpectedly, we found that HA's receptor binding preference does not necessarily reflect virus-cell specificity. We propose SVFS as a tool to elucidate the cell binding preference of IAVs, thereby including the complex environment of sialylated receptors within the plasma membrane of living cells.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Humanos , Virus de la Influenza A/metabolismo , Receptores Virales/metabolismo , Acoplamiento Viral , Polisacáridos/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/química
18.
Int J Food Microbiol ; 403: 110304, 2023 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-37429117

RESUMEN

Quorum sensing (QS) plays an important role in phage-host interactions. Shewanella baltica can't produce the N-acyl-homoserine lactones (AHLs) signal molecules but can eavesdrop on exogenous AHLs through its LuxR receptor. However, no clear evidence exists regarding the involvement of AHLs-mediated QS systems in S. baltica in regulating phage infection. Here, we report that AHLs modulated the phage resistance of S. baltica OS155. Specifically, we characterized a S. baltica phage vB_Sb_QDWS and preliminarily identified that lipopolysaccharide (LPS) is an important receptor for phage vB_Sb_QDWS. AHLs could protect S. baltica against phage infection by decreasing LPS-mediated phage adsorption. The expression of genes galU and tkt, which are essential for LPS synthesis, down-regulated significantly in response to AHLs autoinducers. Our finding confirms the important roles of QS in virus-host interactions and would be helpful to develop novel phage strategies for food spoilage control.


Asunto(s)
Acil-Butirolactonas , Proteínas Bacterianas , Bacteriófagos , Shewanella , Transactivadores , Percepción de Quorum , Shewanella/metabolismo , Shewanella/virología , Transducción de Señal , Acil-Butirolactonas/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Bacteriófagos/fisiología , Acoplamiento Viral , Receptores Virales/metabolismo , Proteínas Bacterianas/metabolismo , Lipopolisacáridos/metabolismo , Expresión Génica
19.
Annu Rev Virol ; 10(1): 283-304, 2023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37285578

RESUMEN

N- and O-glycans are both important constituents of viral envelope glycoproteins. O-linked glycosylation can be initiated by any of 20 different human polypeptide O-acetylgalactosaminyl transferases, resulting in an important functional O-glycan heterogeneity. O-glycans are organized as solitary glycans or in clusters of multiple glycans forming mucin-like domains. They are functional both in the viral life cycle and in viral colonization of their host. Negatively charged O-glycans are crucial for the interactions between glycosaminoglycan-binding viruses and their host. A novel mechanism, based on controlled electrostatic repulsion, explains how such viruses solve the conflict between optimized viral attachment to target cells and efficient egress of progeny virus. Conserved solitary O-glycans appear important for viral uptake in target cells by contributing to viral envelope fusion. Dual roles of viral O-glycans in the host B cell immune response, either epitope blocking or epitope promoting, may be exploitable for vaccine development. Finally, specific virus-induced O-glycans may be involved in viremic spread.


Asunto(s)
Polisacáridos , Proteínas del Envoltorio Viral , Humanos , Proteínas del Envoltorio Viral/metabolismo , Polisacáridos/metabolismo , Glicosilación , Acoplamiento Viral , Epítopos/metabolismo
20.
Proc Natl Acad Sci U S A ; 120(24): e2219404120, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37276413

RESUMEN

Nogo-66 receptor 1 (NgR1) binds a variety of structurally dissimilar ligands in the adult central nervous system to inhibit axon extension. Disruption of ligand binding to NgR1 and subsequent signaling can improve neuron outgrowth, making NgR1 an important therapeutic target for diverse neurological conditions such as spinal crush injuries and Alzheimer's disease. Human NgR1 serves as a receptor for mammalian orthoreovirus (reovirus), but the mechanism of virus-receptor engagement is unknown. To elucidate how NgR1 mediates cell binding and entry of reovirus, we defined the affinity of interaction between virus and receptor, determined the structure of the virus-receptor complex, and identified residues in the receptor required for virus binding and infection. These studies revealed that central NgR1 surfaces form a bridge between two copies of viral capsid protein σ3, establishing that σ3 serves as a receptor ligand for reovirus. This unusual binding interface produces high-avidity interactions between virus and receptor to prime early entry steps. These studies refine models of reovirus cell-attachment and highlight the evolution of viruses to engage multiple receptors using distinct capsid components.


Asunto(s)
Orthoreovirus , Reoviridae , Animales , Humanos , Receptor Nogo 1/metabolismo , Acoplamiento Viral , Proteínas Virales/metabolismo , Ligandos , Reoviridae/metabolismo , Orthoreovirus/metabolismo , Receptores Virales/metabolismo , Mamíferos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...