Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.035
Filtrar
1.
J Ethnopharmacol ; 336: 118719, 2025 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-39179057

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Morus alba L. are widely used as ethnomedicine and functional food in China, Japan, Korea and other Asian countries. Morus alba L. have a variety of pharmacological activity such as antiviral, antioxidation, anti-cholesterol, anticancer, hypoglycemia, and neuroprotection. Morus alba L. has demonstrated antiviral efficacy against influenza viruses, SARS-CoV-2 and so on, but its potential activity against pseudorabies virus (PRV) remains uncertain. AIM OF THE STUDY: This study endeavors to delve into the anti-pseudorabies virus (PRV) potential of the ethanol extract of Morus alba L. leaves (MLE), while simultaneously elucidating its underlying mechanism of action. MATERIALS AND METHODS: The anti-PRV activities of Morus alba L. extracts at different concentrations were evaluated by qPCR and immunoblotting. The inhibitory effects of MLE on PRV replication in three distinct treatment modes (pretreatment, co-treatment, and post-treatment) were detected by qPCR and indirect immunofluorescence assays. qPCR was used to investigate the effects of MLE on PRV attachment, entrance, and cytokine expression in PRV-infected cells. The chemical components in MLE were analyzed by UPLC-MS/MS. RESULTS: MLE significantly inhibits PRV replication and protein expression in a dose-dependent manner. MLE displays inhibitory effects against PRV at three different modes of treatment. The most significant inhibitory effect of MLE was observed when used in co-treatment mode, resulting in an inhibition rate of 99.42%. MLE inhibits PRV infection in the early stage. MLE inhibits PRV infection by affecting viral attachment and viral entry. Furthermore, MLE exerts its inhibition on PRV replication by mitigating the heightened expression of cytokines (TNF-α and IFN-α) triggered by PRV. Analysis of its chemical composition highlights phenolic acids and flavonoids as the principal constituents of MLE. CONCLUSION: The results illustrate that MLE effectively impedes PRV infection by suppressing viral adsorption and entry, while also curbing the expression of antiviral cytokines. Therefore, MLE may be a potential resource for creating new medications to treat human and animal PRV infections.


Assuntos
Antivirais , Herpesvirus Suídeo 1 , Morus , Extratos Vegetais , Folhas de Planta , Replicação Viral , Herpesvirus Suídeo 1/efeitos dos fármacos , Morus/química , Antivirais/farmacologia , Antivirais/isolamento & purificação , Extratos Vegetais/farmacologia , Animais , Replicação Viral/efeitos dos fármacos , Folhas de Planta/química , Citocinas/metabolismo , Cães , Células Madin Darby de Rim Canino , Internalização do Vírus/efeitos dos fármacos , Ligação Viral/efeitos dos fármacos
2.
Nature ; 633(8030): 704-709, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39232163

RESUMO

Fifty-eight million individuals worldwide are affected by chronic hepatitis C virus (HCV) infection, a primary driver of liver cancer for which no vaccine is available1. The HCV envelope proteins E1 and E2 form a heterodimer (E1/E2), which is the target for neutralizing antibodies2. However, the higher-order organization of these E1/E2 heterodimers, as well as that of any Hepacivirus envelope protein complex, remains unknown. Here we determined the cryo-electron microscopy structure of two E1/E2 heterodimers in a homodimeric arrangement. We reveal how the homodimer is established at the molecular level and provide insights into neutralizing antibody evasion and membrane fusion by HCV, as orchestrated by E2 motifs such as hypervariable region 1 and antigenic site 412, as well as the organization of the transmembrane helices, including two internal to E1. This study addresses long-standing questions on the higher-order oligomeric arrangement of Hepacivirus envelope proteins and provides a critical framework in the design of novel HCV vaccine antigens.


Assuntos
Anticorpos Neutralizantes , Microscopia Crioeletrônica , Hepacivirus , Multimerização Proteica , Proteínas do Envelope Viral , Humanos , Motivos de Aminoácidos , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/química , Hepacivirus/química , Hepacivirus/ultraestrutura , Evasão da Resposta Imune , Fusão de Membrana , Modelos Moleculares , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/ultraestrutura , Internalização do Vírus
3.
Emerg Microbes Infect ; 13(1): 2400559, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39222358

RESUMO

Porcine deltacoronavirus (PDCoV) is an emerging pathogen that can cause severe diarrhoea and high mortality in suckling piglets. Moreover, evidence of PDCoV infection in humans has raised concerns regarding potential public health risks. To identify potential therapeutic targets for PDCoV, we performed a genome-wide CRISPR/Cas9 library screening to find key host factors important to PDCoV infection. Several host genes in this screen were enriched, including ANPEP, which encodes the PDCoV receptor aminopeptidase N (APN). Furthermore, we discovered C16orf62, also known as the VPS35 endosomal protein sorting factor like (VPS35L), as an important host factor required for PDCoV infection. C16orf62 is an important component of the multiprotein retriever complex involved in protein recycling in the endosomal compartment and its gene knockout led to a remarkable decrease in the binding and internalization of PDCoV into host cells. While we did not find evidence for direct interaction between C16orf62 and the viral s (spike) protein, C16orf62 gene knockout was shown to downregulate APN expression at the cell surface. This study marks the first instance of a genome-wide CRISPR/Cas9-based screen tailored for PDCoV, revealing C16orf62 as a host factor required for PDCoV replication. These insights may provide promising avenues for the development of antiviral drugs against PDCoV infection.


Assuntos
Sistemas CRISPR-Cas , Infecções por Coronavirus , Deltacoronavirus , Doenças dos Suínos , Animais , Suínos , Doenças dos Suínos/virologia , Deltacoronavirus/genética , Infecções por Coronavirus/virologia , Infecções por Coronavirus/veterinária , Humanos , Antígenos CD13/metabolismo , Antígenos CD13/genética , Técnicas de Inativação de Genes , Interações Hospedeiro-Patógeno , Internalização do Vírus
4.
Proc Natl Acad Sci U S A ; 121(37): e2404250121, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39231203

RESUMO

Human cytomegalovirus (HCMV) glycoprotein B (gB) is a class III membrane fusion protein required for viral entry. HCMV vaccine candidates containing gB have demonstrated moderate clinical efficacy, but no HCMV vaccine has been approved. Here, we used structure-based design to identify and characterize amino acid substitutions that stabilize gB in its metastable prefusion conformation. One variant containing two engineered interprotomer disulfide bonds and two cavity-filling substitutions (gB-C7), displayed increased expression and thermostability. A 2.8 Å resolution cryoelectron microscopy structure shows that gB-C7 adopts a prefusion-like conformation, revealing additional structural elements at the membrane-distal apex. Unlike previous observations for several class I viral fusion proteins, mice immunized with postfusion or prefusion-stabilized forms of soluble gB protein displayed similar neutralizing antibody titers, here specifically against an HCMV laboratory strain on fibroblasts. Collectively, these results identify initial strategies to stabilize class III viral fusion proteins and provide tools to probe gB-directed antibody responses.


Assuntos
Citomegalovirus , Proteínas do Envelope Viral , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Citomegalovirus/imunologia , Humanos , Animais , Camundongos , Microscopia Crioeletrônica , Conformação Proteica , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Internalização do Vírus , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Estabilidade Proteica , Vacinas contra Citomegalovirus/imunologia , Substituição de Aminoácidos , Modelos Moleculares
5.
PLoS Pathog ; 20(9): e1012493, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39236072

RESUMO

Grasping the roles of epitopes in viral glycoproteins is essential for unraveling the structure and function of these proteins. Up to now, all identified epitopes have been found to either neutralize, have no effect on, or enhance viral entry into cells. Here, we used nanobodies (single-domain antibodies) as probes to investigate a unique epitope on the SARS-CoV-2 spike protein, located outside the protein's receptor-binding domain. Nanobody binding to this epitope enhances the cell entry of prototypic SARS-CoV-2, while neutralizing the cell entry of SARS-CoV-2 Omicron variant. Moreover, nanobody binding to this epitope promotes both receptor binding activity and post-attachment activity of prototypic spike, explaining the enhanced viral entry. The opposite occurs with Omicron spike, explaining the neutralized viral entry. This study reveals a unique epitope that can both enhance and neutralize viral entry across distinct viral variants, suggesting that epitopes may vary their roles depending on the viral context. Consequently, antibody therapies should be assessed across different viral variants to confirm their efficacy and safety.


Assuntos
Anticorpos Neutralizantes , COVID-19 , Epitopos , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Internalização do Vírus , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , Humanos , Epitopos/imunologia , COVID-19/imunologia , COVID-19/virologia , Anticorpos Neutralizantes/imunologia , Anticorpos de Domínio Único/imunologia , Anticorpos Antivirais/imunologia , Animais
6.
Proc Natl Acad Sci U S A ; 121(40): e2403260121, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39298475

RESUMO

Cellular processes are controlled by the thermodynamics of the underlying biomolecular interactions. Frequently, structural investigations use one monomeric binding partner, while ensemble measurements of binding affinities generally yield one affinity representative of a 1:1 interaction, despite the majority of the proteome consisting of oligomeric proteins. For example, viral entry and inhibition in SARS-CoV-2 involve a trimeric spike surface protein, a dimeric angiotensin-converting enzyme 2 (ACE2) cell-surface receptor and dimeric antibodies. Here, we reveal that cooperativity correlates with infectivity and inhibition as opposed to 1:1 binding strength. We show that ACE2 oligomerizes spike more strongly for more infectious variants, while exhibiting weaker 1:1 affinity. Furthermore, we find that antibodies use induced oligomerization both as a primary inhibition mechanism and to enhance the effects of receptor-site blocking. Our results suggest that naive affinity measurements are poor predictors of potency, and introduce an antibody-based inhibition mechanism for oligomeric targets. More generally, they point toward a much broader role of induced oligomerization in controlling biomolecular interactions.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Ligação Proteica , Multimerização Proteica , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , SARS-CoV-2/metabolismo , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/química , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/química , Humanos , COVID-19/virologia , COVID-19/metabolismo , COVID-19/imunologia , Internalização do Vírus/efeitos dos fármacos , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/metabolismo , Termodinâmica
7.
Int J Mol Sci ; 25(17)2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39273582

RESUMO

Angiotensin-converting enzyme 2 (ACE2) is considered a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor of high importance, but due to its non-ubiquitous expression, studies of other proteins that may participate in virus internalisation have been undertaken. To date, many alternative receptors have been discovered. Their functioning may provide an explanation for some of the events observed in severe COVID-19 that cannot be directly explained by the model in which ACE2 constitutes the central point of infection. Diabetes mellitus type 2 (T2D) can induce severe COVID-19 development. Although many mechanisms associated with ACE2 can lead to increased SARS-CoV-2 virulence in diabetes, proteins such as basigin (CD147), glucose-regulated protein 78 kDa (GRP78), cluster of differentiation 4 (CD4), transferrin receptor (TfR), integrins α5ß1/αvß3, or ACE2 co-receptors neuropilin 2 (NRP2), vimentin, and even syalilated gangliosides may also be responsible for worsening the COVID-19 course. On the other hand, some others may play protective roles. Understanding how diabetes-associated mechanisms can induce severe COVID-19 via modification of virus receptor functioning needs further extensive studies.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Diabetes Mellitus Tipo 2 , Chaperona BiP do Retículo Endoplasmático , SARS-CoV-2 , COVID-19/metabolismo , COVID-19/virologia , COVID-19/complicações , Humanos , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Enzima de Conversão de Angiotensina 2/metabolismo , Chaperona BiP do Retículo Endoplasmático/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/virologia , Internalização do Vírus , Receptores Virais/metabolismo
8.
Nat Commun ; 15(1): 8221, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39300135

RESUMO

The main vectors of Zika virus (ZIKV) and dengue virus (DENV) are Aedes aegypti and Ae. albopictus, with Ae. aegypti being more competent. However, the underlying mechanisms remain unclear. Here, we find Ae. albopictus shows comparable vector competence to ZIKV/DENV with Ae. aegypti by blood-feeding after antibiotic treatment or intrathoracic injection. This suggests that midgut microbiota can influence vector competence. Enterobacter hormaechei_B17 (Eh_B17) is isolated from field-collected Ae. albopictus and conferred resistance to ZIKV/DENV infection in Ae. aegypti after gut-transplantation. Sphingosine, a metabolite secreted by Eh_B17, effectively suppresses ZIKV infection in both Ae. aegypti and cell cultures by blocking viral entry during the fusion step, with an IC50 of approximately 10 µM. A field survey reveals that Eh_B17 preferentially colonizes Ae. albopictus compared to Ae. aegypti. And field Ae. albopictus positive for Eh_B17 are more resistant to ZIKV infection. These findings underscore the potential of gut symbiotic bacteria, such as Eh_B17, to modulate the arbovirus vector competence of Aedes mosquitoes. As a natural antiviral agent, Eh_B17 holds promise as a potential candidate for blocking ZIKV/DENV transmission.


Assuntos
Aedes , Vírus da Dengue , Enterobacter , Microbioma Gastrointestinal , Mosquitos Vetores , Esfingosina , Simbiose , Zika virus , Aedes/virologia , Aedes/microbiologia , Aedes/efeitos dos fármacos , Animais , Mosquitos Vetores/microbiologia , Mosquitos Vetores/virologia , Mosquitos Vetores/efeitos dos fármacos , Zika virus/fisiologia , Zika virus/efeitos dos fármacos , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/fisiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacologia , Enterobacter/efeitos dos fármacos , Enterobacter/fisiologia , Infecção por Zika virus/transmissão , Infecção por Zika virus/virologia , Dengue/transmissão , Dengue/virologia , Dengue/prevenção & controle , Feminino , Internalização do Vírus/efeitos dos fármacos , Humanos
9.
J Med Virol ; 96(9): e29913, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39257039

RESUMO

This study aimed to investigate the impact of different types of nasal inflammation on the regulation of entry-associated genes of respiratory viruses, including severe acute respiratory syndrome coronavirus 2 (SARS CoV-2), Middle East respiratory syndrome coronavirus (MERS-CoV), human coronavirus 229E (HCoV-229E), and influenza virus, in the nasal epithelium. Subjects were classified into three groups: control, eosinophilic chronic rhinosinusitis (ECRS), and noneosinophilic CRS (NECRS) groups. Angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine subtype 2 (TMPRSS2), alanyl aminopeptidase (ANPEP), dipeptidyl peptidase 4 (DPP4), and beta-galactoside alpha-2,6-sialyltransferase 1 (ST6GAL1), and beta-galactoside alpha-2,3-sialyltransferase 4 (ST3GAL4) were selected as key entry-associated genes for SARS-CoV-2, HCoV-229E, MERS-CoV, and influenza, respectively, and were evaluated. Brushing samples obtained from each group and human nasal epithelial cells cultured using an air-liquid interface system were treated for 7 days with typical inflammatory cytokines and analyzed using real-time polymerase chain reaction. Western blot analysis and confocal microscopy were performed. The entry-associated genes showed distinct regulation patterns in response to each interleukin-4 (IL-4), interleukin-13 (IL-13), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ). Specifically, ACE2 significantly decreased in type 2 cytokines (IL-4 and IL-13), while TMPRSS2 significantly decreased in type 1 cytokines (TNF-α and IFN-γ). ANPEP significantly decreased in both types of cytokines. Remarkably, DPP4 significantly increased in type 2 cytokines and decreased in type 1 cytokines. Moreover, ST6GAL1 and ST3GAL4 significantly increased in type 2 cytokines and decreased in type 1 cytokines, particularly IFN-γ. These findings were supported by western blot analysis and confocal imaging results, especially for ACE2 and DPP4. The findings regarding differential regulation suggest that patients with ECRS, primarily mediated by type 2 inflammation, may have lower susceptibility to SARS-CoV-2 and HCoV-229E infections but higher susceptibility to MERS-CoV and influenza infections.


Assuntos
Citocinas , Mucosa Nasal , Internalização do Vírus , Humanos , Citocinas/genética , Citocinas/metabolismo , Mucosa Nasal/virologia , Adulto , Masculino , Feminino , Pessoa de Meia-Idade , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Sinusite/virologia , Sinusite/genética , Sinusite/imunologia , SARS-CoV-2/imunologia , Rinite/virologia , Rinite/genética , Rinite/imunologia , Regulação da Expressão Gênica , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , COVID-19/imunologia , COVID-19/virologia , Coronavirus Humano 229E/genética , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/metabolismo , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia
10.
Int J Mol Sci ; 25(17)2024 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-39273370

RESUMO

Zika virus (ZIKV; family, Flaviviridae), which causes congenital Zika syndrome, Guillain-Barré Syndrome, and other severe diseases, is transmitted mainly by mosquitoes; however, the virus can be transmitted through other routes. Among the three structural and seven nonstructural proteins, the surface envelope (E) protein of ZIKV plays a critical role in viral entry and pathogenesis, making it a key target for the development of effective entry inhibitors. This review article describes the life cycle, genome, and encoded proteins of ZIKV, illustrates the structure and function of the ZIKV E protein, summarizes E protein-targeting entry inhibitors (with a focus on those based on natural products and small molecules), and highlights challenges that may potentially hinder the development of effective inhibitors of ZIKV infection. Overall, the article will provide useful guidance for further development of safe and potent ZIKV entry inhibitors targeting the viral E protein.


Assuntos
Antivirais , Proteínas do Envelope Viral , Internalização do Vírus , Infecção por Zika virus , Zika virus , Zika virus/efeitos dos fármacos , Zika virus/fisiologia , Internalização do Vírus/efeitos dos fármacos , Humanos , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/antagonistas & inibidores , Infecção por Zika virus/virologia , Infecção por Zika virus/tratamento farmacológico , Antivirais/farmacologia , Animais
11.
Nat Commun ; 15(1): 7668, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39227374

RESUMO

The fusion of viruses with cellular membranes is a critical step in the life cycle of enveloped viruses. This process is facilitated by viral fusion proteins, many of which are conformationally pH-sensitive. The specifics of how changes in pH initiate this fusion have remained largely elusive. This study presents the cryo-electron microscopy (cryo-EM) structures of a prototype class III fusion protein, GP64, in its prefusion and early intermediate states, revealing the structural intermediates accompanying the membrane fusion process. The structures identify the involvement of a pH-sensitive switch, comprising H23, H245, and H304, in sensing the low pH that triggers the initial step of membrane fusion. The pH sensing role of this switch is corroborated by assays of cell-cell syncytium formation and dual dye-labeling. The findings demonstrate that coordination between multiple histidine residues acts as a pH sensor and activator. The involvement of a multi-histidine switch in viral fusion is applicable to fusogens of human-infecting thogotoviruses and other viruses, which could lead to strategies for developing anti-viral therapies and vaccines.


Assuntos
Microscopia Crioeletrônica , Histidina , Concentração de Íons de Hidrogênio , Histidina/química , Histidina/metabolismo , Humanos , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Fusão de Membrana , Animais , Modelos Moleculares , Internalização do Vírus , Conformação Proteica
12.
Results Probl Cell Differ ; 73: 375-417, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39242387

RESUMO

Multicellular organisms require cell-to-cell communication to maintain homeostasis and thrive. For cells to communicate, a network of filamentous, actin-rich tunneling nanotubes (TNTs) plays a pivotal role in facilitating efficient cell-to-cell communication by connecting the cytoplasm of adjacent or distant cells. Substantial documentation indicates that diverse cell types employ TNTs in a sophisticated and intricately organized fashion for both long and short-distance communication. Paradoxically, several pathogens, including viruses, exploit the structural integrity of TNTs to facilitate viral entry and rapid cell-to-cell spread. These pathogens utilize a "surfing" mechanism or intracellular transport along TNTs to bypass high-traffic cellular regions and evade immune surveillance and neutralization. Although TNTs are present across various cell types in healthy tissue, their magnitude is increased in the presence of viruses. This heightened induction significantly amplifies the role of TNTs in exacerbating disease manifestations, severity, and subsequent complications. Despite significant advancements in TNT research within the realm of infectious diseases, further studies are imperative to gain a precise understanding of TNTs' roles in diverse pathological conditions. Such investigations are essential for the development of novel therapeutic strategies aimed at leveraging TNT-associated mechanisms for clinical applications. In this chapter, we emphasize the significance of TNTs in the life cycle of viruses, showcasing the potential for a targeted approach to impede virus-host cell interactions during the initial stages of viral infections. This approach holds promise for intervention and prevention strategies.


Assuntos
Comunicação Celular , Humanos , Animais , Nanotubos/química , Internalização do Vírus , Estruturas da Membrana Celular
13.
Nat Commun ; 15(1): 7711, 2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-39231976

RESUMO

Baculovirus is an obligate parasitic virus of the phylum Arthropoda. Baculovirus including Autographa californica multiple nucleopolyhedrovirus (AcMNPV) has been widely used in the laboratory and industrial preparation of proteins or protein complexes. Due to its large packaging capacity and non-replicative and non-integrative natures in mammals, baculovirus has been proposed as a gene therapy vector for transgene delivery. However, the mechanism of baculovirus transduction in mammalian cells has not been fully illustrated. Here, we employed a cell surface protein-focused CRISPR screen to identify host dependency factors for baculovirus transduction in mammalian cells. The screening experiment uncovered a series of baculovirus host factors in human cells, including exostosin-like glycosyltransferase 3 (EXTL3) and NPC intracellular cholesterol transporter 1 (NPC1). Further investigation illustrated that EXTL3 affected baculovirus attachment and entry by participating in heparan sulfate biosynthesis. In addition, NPC1 promoted baculovirus transduction by mediating membrane fusion and endosomal escape. Moreover, in vivo, baculovirus transduction in Npc1-/+ mice showed that disruption of Npc1 gene significantly reduced baculovirus transduction in mouse liver. In summary, our study revealed the functions of EXTL3 and NPC1 in baculovirus attachment, entry, and endosomal escape in mammalian cells, which is useful for understanding baculovirus transduction in human cells.


Assuntos
N-Acetilglucosaminiltransferases , Proteína C1 de Niemann-Pick , Nucleopoliedrovírus , Animais , Nucleopoliedrovírus/genética , Nucleopoliedrovírus/fisiologia , Humanos , Camundongos , N-Acetilglucosaminiltransferases/metabolismo , N-Acetilglucosaminiltransferases/genética , Células HEK293 , Endossomos/metabolismo , Heparitina Sulfato/metabolismo , Internalização do Vírus , Transdução Genética , Células Sf9 , Fígado/metabolismo , Fígado/virologia , Sistemas CRISPR-Cas
14.
BMC Infect Dis ; 24(1): 934, 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39251948

RESUMO

BACKGROUND: Coinfection with two phylogenetically distinct Human Immunodeficiency Virus-1 (HIV-1) variants might provide an opportunity for rapid viral expansion and the emergence of fit variants that drive disease progression. However, autologous neutralising immune responses are known to drive Envelope (Env) diversity which can either enhance replicative capacity, have no effect, or reduce viral fitness. This study investigated whether in vivo outgrowth of coinfecting variants was linked to pseudovirus and infectious molecular clones' infectivity to determine whether diversification resulted in more fit virus with the potential to increase disease progression. RESULTS: For most participants, emergent recombinants displaced the co-transmitted variants and comprised the major population at 52 weeks postinfection with significantly higher entry efficiency than other co-circulating viruses. Our findings suggest that recombination within gp41 might have enhanced Env fusogenicity which contributed to the increase in pseudovirus entry efficiency. Finally, there was a significant correlation between pseudovirus entry efficiency and CD4 + T cell count, suggesting that the enhanced replicative capacity of recombinant variants could result in more virulent viruses. CONCLUSION: Coinfection provides variants with the opportunity to undergo rapid recombination that results in more infectious virus. This highlights the importance of monitoring the replicative fitness of emergent viruses.


Assuntos
Coinfecção , Infecções por HIV , HIV-1 , Filogenia , Humanos , Infecções por HIV/virologia , Infecções por HIV/complicações , HIV-1/genética , HIV-1/fisiologia , Coinfecção/virologia , Evolução Molecular , Produtos do Gene env do Vírus da Imunodeficiência Humana/genética , Proteína gp41 do Envelope de HIV/genética , Masculino , Feminino , Recombinação Genética , Internalização do Vírus , Adulto , Contagem de Linfócito CD4 , Replicação Viral
15.
Int J Mol Sci ; 25(15)2024 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-39125815

RESUMO

Neurological symptoms associated with COVID-19, acute and long term, suggest SARS-CoV-2 affects both the peripheral and central nervous systems (PNS/CNS). Although studies have shown olfactory and hematogenous invasion into the CNS, coinciding with neuroinflammation, little attention has been paid to susceptibility of the PNS to infection or to its contribution to CNS invasion. Here we show that sensory and autonomic neurons in the PNS are susceptible to productive infection with SARS-CoV-2 and outline physiological and molecular mechanisms mediating neuroinvasion. Our infection of K18-hACE2 mice, wild-type mice, and golden Syrian hamsters, as well as primary peripheral sensory and autonomic neuronal cultures, show viral RNA, proteins, and infectious virus in PNS neurons, satellite glial cells, and functionally connected CNS tissues. Additionally, we demonstrate, in vitro, that neuropilin-1 facilitates SARS-CoV-2 neuronal entry. SARS-CoV-2 rapidly invades the PNS prior to viremia, establishes a productive infection in peripheral neurons, and results in sensory symptoms often reported by COVID-19 patients.


Assuntos
COVID-19 , Neuropilina-1 , SARS-CoV-2 , Animais , SARS-CoV-2/fisiologia , SARS-CoV-2/patogenicidade , COVID-19/virologia , COVID-19/patologia , COVID-19/metabolismo , Camundongos , Neuropilina-1/metabolismo , Neuropilina-1/genética , Viremia/virologia , Sistema Nervoso Central/virologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/metabolismo , Células Receptoras Sensoriais/virologia , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/patologia , Mesocricetus , Humanos , Enzima de Conversão de Angiotensina 2/metabolismo , Camundongos Endogâmicos C57BL , Internalização do Vírus , Masculino
16.
ACS Nano ; 18(34): 23090-23103, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39143650

RESUMO

Actin- and microtubule (MT)-based transport systems are essential for intracellular transport. During influenza A virus (IAV) infection, MTs provide long tracks for virus trafficking toward the nucleus. However, the role of the actin cytoskeleton in IAV entry and especially the transit process is still ambiguous. Here, by using quantum dot-based single-virus tracking, it was revealed that the actin cytoskeleton was crucial for the virus entry via clathrin-mediated endocytosis (CME). After entry via CME, the virus reached MTs through three different pathways: the virus (1) was driven by myosin VI to move along actin filaments to reach MTs (AF); (2) was propelled by actin tails assembled by an Arp2/3-dependent mechanism to reach MTs (AT); and (3) directly reached MTs without experiencing actin-related movement (NA). Therefore, the NA pathway was the main one and the fastest for the virus to reach MTs. The AT pathway was activated only when plenty of viruses entered the cell. The viruses transported by the AF and AT pathways shared similar moving velocities, durations, and displacements. This study comprehensively visualized the role of the actin cytoskeleton in IAV entry and transport, revealing different pathways for IAV to reach MTs after entry. The results are of great significance for globally understanding IAV infection and the cellular endocytic transport pathway.


Assuntos
Endocitose , Vírus da Influenza A , Microtúbulos , Pontos Quânticos , Pontos Quânticos/química , Microtúbulos/metabolismo , Microtúbulos/virologia , Humanos , Vírus da Influenza A/fisiologia , Internalização do Vírus , Animais , Cães , Células Madin Darby de Rim Canino , Citoesqueleto de Actina/metabolismo
17.
Viruses ; 16(8)2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39205315

RESUMO

The efficiency of virus internalization into target cells is a major determinant of infectivity. SARS-CoV-2 internalization occurs via S-protein-mediated cell binding followed either by direct fusion with the plasma membrane or endocytosis and subsequent fusion with the endosomal membrane. Despite the crucial role of virus internalization, the precise kinetics of the processes involved remains elusive. We developed a pipeline, which combines live-cell microscopy and advanced image analysis, for measuring the rates of multiple internalization-associated molecular events of single SARS-CoV-2-virus-like particles (VLPs), including endosome ingression and pH change. Our live-cell imaging experiments demonstrate that only a few minutes after binding to the plasma membrane, VLPs ingress into RAP5-negative endosomes via dynamin-dependent scission. Less than two minutes later, VLP speed increases in parallel with a pH drop below 5, yet these two events are not interrelated. By co-imaging fluorescently labeled nucleocapsid proteins, we show that nucleocapsid release occurs with similar kinetics to VLP acidification. Neither Omicron mutations nor abrogation of the S protein polybasic cleavage site affected the rate of VLP internalization, indicating that they do not confer any significant advantages or disadvantages during this process. Finally, we observe that VLP internalization occurs two to three times faster in VeroE6 than in A549 cells, which may contribute to the greater susceptibility of the former cell line to SARS-CoV-2 infection. Taken together, our precise measurements of the kinetics of VLP internalization-associated processes shed light on their contribution to the effectiveness of SARS-CoV-2 propagation in cells.


Assuntos
COVID-19 , Endossomos , SARS-CoV-2 , Internalização do Vírus , SARS-CoV-2/fisiologia , SARS-CoV-2/metabolismo , Humanos , Cinética , COVID-19/virologia , COVID-19/metabolismo , Endossomos/metabolismo , Endossomos/virologia , Endocitose , Animais , Concentração de Íons de Hidrogênio , Chlorocebus aethiops , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero , Membrana Celular/metabolismo , Membrana Celular/virologia , Vírion/metabolismo
18.
Int J Mol Sci ; 25(16)2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39201788

RESUMO

Nipah virus (NiV) is known to be a highly pathogenic zoonotic virus, which is included in the World Health Organization Research & Development Blueprint list of priority diseases with up to 70% mortality rate. Due to its high pathogenicity and outbreak potency, a therapeutic countermeasure against NiV is urgently needed. As NiV needs to be handled within a Biological Safety Level (BSL) 4 facility, we had developed a safe drug screening platform utilizing a baculovirus expression vector system (BEVS) based on a NiV-induced syncytium formation that could be handled within a BSL-1 facility. To reconstruct the NiV-induced syncytium formation in BEVS, two baculoviruses were generated to express recombinant proteins that are responsible for inducing the syncytium formation, including one baculovirus exhibiting co-expressed NiV fusion protein (NiV-F) and NiV attachment glycoprotein (NiV-G) and another exhibiting human EphrinB2 protein. Interestingly, syncytium formation was observed in infected insect cells when the medium was modified to have a lower pH level and supplemented with cholesterol. Fusion inhibitory properties of several compounds, such as phytochemicals and a polysulfonated naphthylamine compound, were evaluated using this platform. Among these compounds, suramin showed the highest fusion inhibitory activity against NiV-induced syncytium in the baculovirus expression system. Moreover, our in silico results provide a molecular-level glimpse of suramin's interaction with NiV-G's central hole and EphrinB2's G-H loop, which could be the possible reason for its fusion inhibitory activity.


Assuntos
Baculoviridae , Avaliação Pré-Clínica de Medicamentos , Células Gigantes , Vírus Nipah , Vírus Nipah/genética , Vírus Nipah/efeitos dos fármacos , Baculoviridae/genética , Animais , Humanos , Células Gigantes/efeitos dos fármacos , Células Gigantes/metabolismo , Células Gigantes/virologia , Avaliação Pré-Clínica de Medicamentos/métodos , Vetores Genéticos/genética , Antivirais/farmacologia , Suramina/farmacologia , Efrina-B2/metabolismo , Efrina-B2/genética , Infecções por Henipavirus/virologia , Células Sf9 , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus/efeitos dos fármacos
19.
J Virol ; 98(9): e0090124, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39194240

RESUMO

Nervous necrosis virus (NNV) is a highly neurotropic virus that poses a persistent threat to the survival of multiple fish species. However, its inimitable neuropathogenesis remains largely elusive. To rummage potential partners germane to the nervous system, we investigated the interaction between red-spotted grouper NNV (RGNNV) and grouper brain by immunoprecipitation coupled with mass spectrometry and discerned Nectin1 as a novel host factor subtly involved in viral early invasion events. Nectin1 was abundant in neural tissues and implicated in the inception of tunnel nanotubes triggered by RGNNV. Its overexpression not only dramatically potentiated the replication dynamics of RGNNV in susceptible cells, but also empowered non-sensitive cells to expeditiously capture free virions within 2 min. This potency was impervious to low temperatures but was dose-dependently suppressed by soluble protein or specific antibody of Nectin1 ectodomain, indicating Nectin1 as an attachment receptor for RGNNV. Mechanistically, efficient hijacking of virions by Nectin1 strictly depended on intricate linkages to different modules of viral capsid protein, especially the direct binding between the IgC1 loop and P-domain. More strikingly, despite abortive proliferation in Nectin1-reconstructed CHSE-214 cells, a non-sensitive cell, RGNNV could gain access to the intracellular compartment by capitalizing on Nectin1, thereby inducing canonical cytoplasmic vacuolation. Altogether, our findings delineate a candidate entrance for RGNNV infiltration into the nervous system, which may shed unprecedented insights into the exploration and elucidation of RGNNV pathogenesis.IMPORTANCENervous necrosis virus (NNV) is one of the most virulent pathogens in the aquaculture industry, which inflicts catastrophic damage to ecology, environment, and economy annually around the world. Nevertheless, its idiosyncratic invasion and latency mechanisms pose enormous hardships to epidemic prevention and control. In this study, deploying grouper brain as a natural screening library, a single-transmembrane glycoprotein, Nectin1, was first identified as an emergent functional receptor for red-spotted grouper NNV (RGNNV) that widely allocated in nervous tissues and directly interacted with viral capsid protein through distinct Ig-like loops to bridge virus-host crosstalk, apprehend free virions, and concomitantly propel viral entry. Our findings illuminate the critical role of Nectin1 in RGNNV attachment and entry and provide a potential target for future clinical intervention strategies in the therapeutic race against RGNNV.


Assuntos
Doenças dos Peixes , Nectinas , Nodaviridae , Infecções por Vírus de RNA , Internalização do Vírus , Animais , Nectinas/metabolismo , Nodaviridae/fisiologia , Doenças dos Peixes/virologia , Doenças dos Peixes/metabolismo , Infecções por Vírus de RNA/virologia , Infecções por Vírus de RNA/metabolismo , Infecções por Vírus de RNA/veterinária , Replicação Viral , Ligação Viral , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Encéfalo/virologia , Encéfalo/metabolismo , Vírion/metabolismo , Linhagem Celular
20.
J Virol ; 98(9): e0079524, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39207135

RESUMO

While investigating methods to target gene delivery vectors to specific cell types, we examined the potential of using a nanobody against the SARS-CoV-2 Spike protein receptor-binding domain to direct lentivirus infection of Spike-expressing cells. Using four different approaches, we found that lentiviruses with surface-exposed nanobody domains selectively infect Spike-expressing cells. Targeting is dependent on the fusion function of the Spike protein, and conforms to a model in which nanobody binding to the Spike protein triggers the Spike fusion machinery. The nanobody-Spike interaction also is capable of directing cell-cell fusion and the selective infection of nanobody-expressing cells by Spike-pseudotyped lentivirus vectors. Significantly, cells infected with SARS-CoV-2 are efficiently and selectively infected by lentivirus vectors pseudotyped with a chimeric nanobody protein. Our results suggest that cells infected by any virus that forms syncytia may be targeted for gene delivery by using an appropriate nanobody or virus receptor mimic. Vectors modified in this fashion may prove useful in the delivery of immunomodulators to infected foci to mitigate the effects of viral infections.IMPORTANCEWe have discovered that lentiviruses decorated on their surfaces with a nanobody against the SARS-CoV-2 Spike protein selectively infect Spike-expressing cells. Infection is dependent on the specificity of the nanobody and the fusion function of the Spike protein and conforms to a reverse fusion model, in which nanobody binding to Spike triggers the Spike fusion machinery. The nanobody-Spike interaction also can drive cell-cell fusion and infection of nanobody-expressing cells with viruses carrying the Spike protein. Importantly, cells infected with SARS-CoV-2 are selectively infected with nanobody-decorated lentiviruses. These results suggest that cells infected by any virus that expresses an active receptor-binding fusion protein may be targeted by vectors for delivery of cargoes to mitigate infections.


Assuntos
Vetores Genéticos , Lentivirus , SARS-CoV-2 , Anticorpos de Domínio Único , Glicoproteína da Espícula de Coronavírus , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Anticorpos de Domínio Único/imunologia , Lentivirus/genética , Humanos , SARS-CoV-2/imunologia , SARS-CoV-2/fisiologia , Vetores Genéticos/genética , Células HEK293 , COVID-19/virologia , COVID-19/imunologia , Ligação Proteica , Internalização do Vírus , Fusão Celular , Técnicas de Transferência de Genes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA