Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.250
Filter
Add more filters

Publication year range
1.
Cell ; 177(6): 1553-1565.e16, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31104841

ABSTRACT

Enterovirus B (EV-B), a major proportion of the genus Enterovirus in the family Picornaviridae, is the causative agent of severe human infectious diseases. Although cellular receptors for coxsackievirus B in EV-B have been identified, receptors mediating virus entry, especially the uncoating process of echovirus and other EV-B remain obscure. Here, we found that human neonatal Fc receptor (FcRn) is the uncoating receptor for major EV-B. FcRn binds to the virus particles in the "canyon" through its FCGRT subunit. By obtaining multiple cryo-electron microscopy structures at different stages of virus entry at atomic or near-atomic resolution, we deciphered the underlying mechanisms of enterovirus attachment and uncoating. These structures revealed that different from the attachment receptor CD55, binding of FcRn to the virions induces efficient release of "pocket factor" under acidic conditions and initiates the conformational changes in viral particle, providing a structural basis for understanding the mechanisms of enterovirus entry.


Subject(s)
Enterovirus B, Human/metabolism , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/ultrastructure , Receptors, Fc/metabolism , Receptors, Fc/ultrastructure , Capsid/metabolism , Cryoelectron Microscopy , Enterovirus , Enterovirus B, Human/pathogenicity , Enterovirus Infections/metabolism , Histocompatibility Antigens Class I/physiology , Humans , Models, Molecular , Phylogeny , Receptors, Fc/physiology , Virion , Virus Internalization
2.
Immunity ; 55(5): 800-818, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35545029

ABSTRACT

Pathogenic enteric viruses are a major cause of morbidity and mortality, particularly among children in developing countries. The host response to enteric viruses occurs primarily within the mucosa, where the intestinal immune system must balance protection against pathogens with tissue protection and tolerance to harmless commensal bacteria and food. Here, we summarize current knowledge in natural immunity to enteric viruses, highlighting specialized features of the intestinal immune system. We further discuss how knowledge of intestinal anti-viral mechanisms can be translated into vaccine development with particular focus on immunization in the oral route. Research reveals that the intestine is a complex interface between enteric viruses and the host where environmental factors influence susceptibility and immunity to infection, while viral infections can have lasting implications for host health. A deeper mechanistic understanding of enteric anti-viral immunity with this broader context can ultimately lead to better vaccines for existing and emerging viruses.


Subject(s)
Enterovirus Infections , Vaccines , Viruses , Antigens, Viral , Child , Humans , Immunity, Innate , Intestinal Mucosa , Intestines
3.
Immunity ; 54(10): 2231-2244.e6, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34555337

ABSTRACT

RNA interference (RNAi) is the major antiviral mechanism in plants and invertebrates, but the absence of detectable viral (v)siRNAs in mammalian cells upon viral infection has questioned the functional relevance of this pathway in mammalian immunity. We designed a series of peptides specifically targeting enterovirus A71 (EV-A71)-encoded protein 3A, a viral suppressor of RNAi (VSR). These peptides abrogated the VSR function of EV-A71 in infected cells and resulted in the accumulation of vsiRNAs and reduced viral replication. These vsiRNAs were functional, as evidenced by RISC-loading and silencing of target RNAs. The effects of VSR-targeting peptides (VTPs) on infection with EV-A71 as well as another enterovirus, Coxsackievirus-A16, were ablated upon deletion of Dicer1 or AGO2, core components of the RNAi pathway. In vivo, VTP treatment protected mice against lethal EV-A71 challenge, with detectable vsiRNAs. Our findings provide evidence for the functional relevance of RNAi in mammalian immunity and present a therapeutic strategy for infectious disease.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus Infections/virology , RNA, Viral/antagonists & inhibitors , Animals , Chlorocebus aethiops , Enterovirus A, Human , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Peptides/pharmacology , RNA Interference , RNA, Small Interfering/antagonists & inhibitors , Vero Cells , Virus Replication/drug effects
4.
Cell ; 160(4): 619-630, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25679758

ABSTRACT

A central paradigm within virology is that each viral particle largely behaves as an independent infectious unit. Here, we demonstrate that clusters of enteroviral particles are packaged within phosphatidylserine (PS) lipid-enriched vesicles that are non-lytically released from cells and provide greater infection efficiency than free single viral particles. We show that vesicular PS lipids are co-factors to the relevant enterovirus receptors in mediating subsequent infectivity and transmission, in particular to primary human macrophages. We demonstrate that clustered packaging of viral particles within vesicles enables multiple viral RNA genomes to be collectively transferred into single cells. This study reveals a novel mode of viral transmission, where enteroviral genomes are transmitted from cell-to-cell en bloc in membrane-bound PS vesicles instead of as single independent genomes. This has implications for facilitating genetic cooperativity among viral quasispecies as well as enhancing viral replication.


Subject(s)
Cytoplasmic Vesicles/virology , Enterovirus Infections/transmission , Enterovirus/physiology , Macrophages/virology , Cytoplasmic Vesicles/chemistry , Humans , Macrophages/cytology , Phosphatidylserines , Poliovirus/physiology , RNA, Viral/metabolism , Rhinovirus/physiology , Virus Replication
5.
PLoS Pathog ; 20(4): e1012159, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38662650

ABSTRACT

Human enteroviruses are the most common human pathogen with over 300 distinct genotypes. Previous work with poliovirus has suggested that it is possible to generate antibody responses in humans and animals that can recognize members of multiple enterovirus species. However, cross protective immunity across multiple enteroviruses is not observed epidemiologically in humans. Here we investigated whether immunization of mice or baboons with inactivated poliovirus or enterovirus virus-like-particles (VLPs) vaccines generates antibody responses that can recognize enterovirus D68 or A71. We found that mice only generated antibodies specific for the antigen they were immunized with, and repeated immunization failed to generate cross-reactive antibody responses as measured by both ELISA and neutralization assay. Immunization of baboons with IPV failed to generate neutralizing antibody responses against enterovirus D68 or A71. These results suggest that a multivalent approach to enterovirus vaccination is necessary to protect against enterovirus disease in vulnerable populations.


Subject(s)
Antibodies, Viral , Cross Reactions , Enterovirus Infections , Poliovirus Vaccine, Inactivated , Animals , Mice , Cross Reactions/immunology , Antibodies, Viral/immunology , Enterovirus Infections/immunology , Enterovirus Infections/prevention & control , Enterovirus Infections/virology , Poliovirus Vaccine, Inactivated/immunology , Poliovirus Vaccine, Inactivated/administration & dosage , Vaccines, Virus-Like Particle/immunology , Antibodies, Neutralizing/immunology , Papio/immunology , Humans , Poliovirus/immunology , Female , Antibody Formation/immunology , Enterovirus/immunology , Mice, Inbred BALB C , Enterovirus D, Human/immunology
6.
PLoS Pathog ; 20(3): e1012036, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38457376

ABSTRACT

Viruses actively reprogram the metabolism of the host to ensure the availability of sufficient building blocks for virus replication and spreading. However, relatively little is known about how picornaviruses-a large family of small, non-enveloped positive-strand RNA viruses-modulate cellular metabolism for their own benefit. Here, we studied the modulation of host metabolism by coxsackievirus B3 (CVB3), a member of the enterovirus genus, and encephalomyocarditis virus (EMCV), a member of the cardiovirus genus, using steady-state as well as 13C-glucose tracing metabolomics. We demonstrate that both CVB3 and EMCV increase the levels of pyrimidine and purine metabolites and provide evidence that this increase is mediated through degradation of nucleic acids and nucleotide recycling, rather than upregulation of de novo synthesis. Finally, by integrating our metabolomics data with a previously acquired phosphoproteomics dataset of CVB3-infected cells, we identify alterations in phosphorylation status of key enzymes involved in nucleotide metabolism, providing insight into the regulation of nucleotide metabolism during infection.


Subject(s)
Cardiovirus , Enterovirus Infections , Enterovirus , Picornaviridae , Humans , Enterovirus/physiology , Encephalomyocarditis virus/physiology , Virus Replication , Enterovirus B, Human/physiology , HeLa Cells
7.
PLoS Pathog ; 20(1): e1011967, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38271479

ABSTRACT

Given the serious neurological complications and deaths associated with enterovirus 71 (EV71) infection, there is an urgent need to develop effective antivirals against this viral infection. In this study, we demonstrated that two Cathelicidin-derived peptides, LL-18 and FF-18 were more potent against EV71 infection than the parent peptide LL-37, which is the mature and processed form of Cathelicidin. These peptides could directly bind to the EV71 virus particles, but not to coxsackievirus, indicative of their high specificity. The binding of peptides with the virus surface occupied the viral canyon region in a way that could block virus-receptor interactions and inhibit viral uncoating. In addition, these peptide analogues could also relieve the deleterious effect of EV71 infection in vivo. Therefore, Cathelicidin-derived peptides might be excellent candidates for further development of antivirals to treat EV71 infection.


Subject(s)
Enterovirus A, Human , Enterovirus Infections , Enterovirus , Humans , Cathelicidins/pharmacology , Virus Internalization , Antiviral Agents/metabolism
8.
PLoS Pathog ; 20(2): e1012022, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38359079

ABSTRACT

Enterovirus A71 (EV-A71) infection involves a variety of receptors. Among them, two transmembrane protein receptors have been investigated in detail and shown to be critical for infection: P-selectin glycoprotein ligand-1 (PSGL-1) in lymphocytes (Jurkat cells), and scavenger receptor class B member 2 (SCARB2) in rhabdomyosarcoma (RD) cells. PSGL-1 and SCARB2 have been reported to be expressed on the surface of Jurkat and RD cells, respectively. In the work reported here, we investigated the roles of PSGL-1 and SCARB2 in the process of EV-A71 entry. We first examined the expression of SCARB2 in Jurkat cells, and detected it within the cytoplasm, but not on the cell surface. Further, using PSGL-1 and SCARB2 knockout cells, we found that although both PSGL-1 and SCARB2 are essential for virus infection of Jurkat cells, virus attachment to these cells requires only PSGL-1. These results led us to evaluate the cell surface expression and the roles of SCARB2 in other EV-A71-susceptible cell lines. Surprisingly, in contrast to the results of previous studies, we found that SCARB2 is absent from the surface of RD cells and other susceptible cell lines we examined, and that although SCARB2 is essential for infection of these cells, it is dispensable for virus attachment. These results indicate that a receptor other than SCARB2 is responsible for virus attachment to the cell and probably for internalization of virions, not only in Jurkat cells but also in RD cells and other EV-A71-susceptible cells. SCARB2 is highly concentrated in lysosomes and late endosomes, where it is likely to trigger acid-dependent uncoating of virions, the critical final step of the entry process. Our results suggest that the essential interactions between EV-A71 and SCARB2 occur, not at the cell surface, but within the cell.


Subject(s)
Enterovirus A, Human , Enterovirus Infections , Enterovirus , Humans , Enterovirus/metabolism , Enterovirus A, Human/genetics , Enterovirus A, Human/metabolism , Cell Membrane/metabolism , Cell Line , Receptors, Scavenger/genetics , Receptors, Scavenger/metabolism , Lysosomal Membrane Proteins/genetics
9.
Immunity ; 46(6): 992-1004.e5, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28636969

ABSTRACT

RNA interference (RNAi) functions as a potent antiviral immunity in plants and invertebrates; however, whether RNAi plays antiviral roles in mammals remains unclear. Here, using human enterovirus 71 (HEV71) as a model, we showed HEV71 3A protein as an authentic viral suppressor of RNAi during viral infection. When the 3A-mediated RNAi suppression was impaired, the mutant HEV71 readily triggered the production of abundant HEV71-derived small RNAs with canonical siRNA properties in cells and mice. These virus-derived siRNAs were produced from viral dsRNA replicative intermediates in a Dicer-dependent manner and loaded into AGO, and they were fully active in degrading cognate viral RNAs. Recombinant HEV71 deficient in 3A-mediated RNAi suppression was significantly restricted in human somatic cells and mice, whereas Dicer deficiency rescued HEV71 infection independently of type I interferon response. Thus, RNAi can function as an antiviral immunity, which is induced and suppressed by a human virus, in mammals.


Subject(s)
Enterovirus A, Human/immunology , Enterovirus Infections/immunology , Immunity , RNA Interference , RNA, Viral/immunology , Animals , Argonaute Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Enterovirus A, Human/genetics , HEK293 Cells , Humans , Mammals , Mice , Mice, 129 Strain , Mice, Knockout , Mutation/genetics , Ribonuclease III/metabolism , Viral Proteins/immunology
10.
PLoS Biol ; 21(1): e3001693, 2023 01.
Article in English | MEDLINE | ID: mdl-36689548

ABSTRACT

RNA recombination in positive-strand RNA viruses is a molecular-genetic process, which permits the greatest evolution of the genome and may be essential to stabilizing the genome from the deleterious consequences of accumulated mutations. Enteroviruses represent a useful system to elucidate the details of this process. On the biochemical level, it is known that RNA recombination is catalyzed by the viral RNA-dependent RNA polymerase using a template-switching mechanism. For this mechanism to function in cells, the recombining genomes must be located in the same subcellular compartment. How a viral genome is trafficked to the site of genome replication and recombination, which is membrane associated and isolated from the cytoplasm, is not known. We hypothesized that genome translation was essential for colocalization of genomes for recombination. We show that complete inactivation of internal ribosome entry site (IRES)-mediated translation of a donor enteroviral genome enhanced recombination instead of impairing it. Recombination did not occur by a nonreplicative mechanism. Rather, sufficient translation of the nonstructural region of the genome occurred to support subsequent steps required for recombination. The noncanonical translation initiation factors, eIF2A and eIF2D, were required for IRES-independent translation. Our results support an eIF2A/eIF2D-dependent mechanism under conditions in which the eIF2-dependent mechanism is inactive. Detection of an IRES-independent mechanism for translation of the enterovirus genome provides an explanation for a variety of debated observations, including nonreplicative recombination and persistence of enteroviral RNA lacking an IRES. The existence of an eIF2A/eIF2D-dependent mechanism in enteroviruses predicts the existence of similar mechanisms in other viruses.


Subject(s)
Enterovirus Infections , Enterovirus , Humans , Enterovirus/physiology , Enterovirus Infections/virology , Internal Ribosome Entry Sites , Peptide Initiation Factors/genetics , Protein Biosynthesis , RNA, Viral/genetics , RNA, Viral/metabolism , Host-Pathogen Interactions
11.
J Immunol ; 212(11): 1782-1790, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38629901

ABSTRACT

Enterovirus 71 (EV71) is a significant causative agent of hand, foot, and mouth disease, with potential serious neurologic complications or fatal outcomes. The lack of effective treatments for EV71 infection is attributed to its elusive pathogenicity. Our study reveals that human plasmacytoid dendritic cells (pDCs), the main type I IFN-producing cells, selectively express scavenger receptor class B, member 2 (SCARB2) and P-selectin glycoprotein ligand 1 (PSGL-1), crucial cellular receptors for EV71. Some strains of EV71 can replicate within pDCs and stimulate IFN-α production. The activation of pDCs by EV71 is hindered by Abs to PSGL-1 and soluble PSGL-1, whereas Abs to SCARB2 and soluble SCARB2 have a less pronounced effect. Our data suggest that only strains binding to PSGL-1, more commonly found in severe cases, can replicate in pDCs and induce IFN-α secretion, highlighting the importance of PSGL-1 in these processes. Furthermore, IFN-α secretion by pDCs can be triggered by EV71 or UV-inactivated EV71 virions, indicating that productive infection is not necessary for pDC activation. These findings provide new insights into the interaction between EV71 and pDCs, suggesting that pDC activation could potentially mitigate the severity of EV71-related diseases.


Subject(s)
Dendritic Cells , Enterovirus A, Human , Interferon-alpha , Lysosomal Membrane Proteins , Membrane Glycoproteins , Dendritic Cells/immunology , Dendritic Cells/virology , Humans , Enterovirus A, Human/immunology , Enterovirus A, Human/physiology , Membrane Glycoproteins/metabolism , Lysosomal Membrane Proteins/metabolism , Lysosomal Membrane Proteins/immunology , Interferon-alpha/metabolism , Interferon-alpha/immunology , Receptors, Scavenger/metabolism , Enterovirus Infections/immunology , Enterovirus Infections/virology , Virus Replication
12.
Nucleic Acids Res ; 52(D1): D770-D776, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37930838

ABSTRACT

Rhinovirus (RV), a prominent causative agent of both upper and lower respiratory diseases, ranks among the most prevalent human respiratory viruses. RV infections are associated with various illnesses, including colds, asthma exacerbations, croup and pneumonia, imposing significant and extended societal burdens. Characterized by a high mutation rate and genomic diversity, RV displays a diverse serological landscape, encompassing a total of 174 serotypes identified to date. Understanding RV genetic diversity is crucial for epidemiological surveillance and investigation of respiratory diseases. This study introduces a comprehensive and high-quality RV data resource, designated RVdb (http://rvdb.mgc.ac.cn), covering 26 909 currently identified RV strains, along with RV-related sequences, 3D protein structures and publications. Furthermore, this resource features a suite of web-based utilities optimized for easy browsing and searching, as well as automatic sequence annotation, multiple sequence alignment (MSA), phylogenetic tree construction, RVdb BLAST and a serotyping pipeline. Equipped with a user-friendly interface and integrated online bioinformatics tools, RVdb provides a convenient and powerful platform on which to analyse the genetic characteristics of RVs. Additionally, RVdb also supports the efforts of virologists and epidemiologists to monitor and trace both existing and emerging RV-related infectious conditions in a public health context.


Subject(s)
Asthma , Enterovirus Infections , Picornaviridae Infections , Rhinovirus , Humans , Genomics , Phylogeny , Picornaviridae Infections/genetics , Rhinovirus/genetics
13.
Lancet ; 403(10429): 862-876, 2024 Mar 02.
Article in English | MEDLINE | ID: mdl-38340741

ABSTRACT

Since the discovery of norovirus in 1972 as a cause of what was contemporarily known as acute infectious non-bacterial gastroenteritis, scientific understanding of the viral gastroenteritides has continued to evolve. It is now recognised that a small number of viruses are the predominant cause of acute gastroenteritis worldwide, in both high-income and low-income settings. Although treatment is still largely restricted to the replacement of fluid and electrolytes, improved diagnostics have allowed attribution of illness, enabling both targeted treatment of individual patients and prioritisation of interventions for populations worldwide. Questions remain regarding specific genetic and immunological factors underlying host susceptibility, and the optimal clinical management of patients who are susceptible to severe or prolonged manifestations of disease. Meanwhile, the worldwide implementation of rotavirus vaccines has led to substantial reductions in morbidity and mortality, and spurred interest in vaccine development to diminish the impact of the most prevalent viruses that are implicated in this syndrome.


Subject(s)
Enterovirus Infections , Gastroenteritis , Norovirus , Plastic Surgery Procedures , Humans , Gastroenteritis/therapy , Income
14.
J Virol ; 98(2): e0135823, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38226810

ABSTRACT

Hand, foot, and mouth disease (HFMD) is caused by more than 20 pathogenic enteroviruses belonging to the Picornaviridae family and Enterovirus genus. Since the introduction of the enterovirus-71 (EV71) vaccine in 2016, the number of HFMD cases caused by EV71 has decreased. However, cases of infections caused by other enteroviruses, such as coxsackievirus A6 (CA6) and coxsackievirus A10, have been increasing accordingly. In this study, we used a clinical isolate of CA6 to establish an intragastric infection mouse model using 7-day-old mice to mimic the natural transmission route, by which we investigated the differential gene expression profiles associated with virus infection and pathogenicity. After intragastric infection, mice exhibited hind limb paralysis symptoms and weight loss, similar to those reported for EV71 infection in mice. The skeletal muscle was identified as the main site of virus replication, with a peak viral load reaching 2.31 × 107 copies/mg at 5 dpi and increased infiltration of inflammatory cells. RNA sequencing analysis identified differentially expressed genes (DEGs) after CA6 infection. DEGs in the blood, muscle, brain, spleen, and thymus were predominantly enriched in immune system responses, including pathways such as Toll-like receptor signaling and PI3K-Akt signaling. Our study has unveiled the genes involved in the host immune response during CA6 infection, thereby enhancing our comprehension of the pathological mechanism of HFMD.IMPORTANCEThis study holds great significance for the field of hand, foot, and mouth disease (HFMD). It not only delves into the disease's etiology, transmission pathways, and severe complications but also establishes a novel mouse model that mimics the natural coxsackievirus A6 infection process, providing a pivotal platform to delve deeper into virus replication and pathogenic mechanisms. Additionally, utilizing RNA-seq technology, it unveils the dynamic gene expression changes during infection, offering valuable leads for identifying novel therapeutic drug targets. This research has the potential to enhance our understanding of HFMD, offering fresh perspectives for disease prevention and treatment and positively impacting children's health worldwide.


Subject(s)
Enterovirus Infections , Enterovirus , Hand, Foot and Mouth Disease , Animals , Child , Humans , Mice , Antibodies, Viral , Disease Models, Animal , Enterovirus/pathogenicity , Enterovirus/physiology , Enterovirus A, Human , Enterovirus Infections/pathology , Enterovirus Infections/virology , Gene Expression , Hand, Foot and Mouth Disease/genetics , Phosphatidylinositol 3-Kinases , Virulence
15.
J Virol ; 98(2): e0174923, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38189249

ABSTRACT

Enterovirus 71 (EV71) is one of the major pathogens causing hand, foot, and mouth disease in children under 5 years old, which can result in severe neurological complications and even death. Due to limited treatments for EV71 infection, the identification of novel host factors and elucidation of mechanisms involved will help to counter this viral infection. N-terminal acetyltransferase 6 (NAT6) was identified as an essential host factor for EV71 infection with genome-wide CRISPR/Cas9 screening. NAT6 facilitates EV71 viral replication depending on its acetyltransferase activity but has little effect on viral release. In addition, NAT6 is also required for Echovirus 7 and coxsackievirus B5 infection, suggesting it might be a pan-enterovirus host factor. We further demonstrated that NAT6 is required for Golgi integrity and viral replication organelle (RO) biogenesis. NAT6 knockout significantly inhibited phosphatidylinositol 4-kinase IIIß (PI4KB) expression and PI4P production, both of which are key host factors for enterovirus infection and RO biogenesis. Further mechanism studies confirmed that NAT6 formed a complex with its substrate actin and one of the PI4KB recruiters-acyl-coenzyme A binding domain containing 3 (ACBD3). Through modulating actin dynamics, NAT6 maintained the integrity of the Golgi and the stability of ACBD3, thereby enhancing EV71 infection. Collectively, these results uncovered a novel mechanism of N-acetyltransferase supporting EV71 infection.IMPORTANCEEnterovirus 71 (EV71) is an important pathogen for children under the age of five, and currently, no effective treatment is available. Elucidating the mechanism of novel host factors supporting viral infection will reveal potential antiviral targets and aid antiviral development. Here, we demonstrated that a novel N-acetyltransferase, NAT6, is an essential host factor for EV71 replication. NAT6 could promote viral replication organelle (RO) formation to enhance viral replication. The formation of enterovirus ROs requires numerous host factors, including acyl-coenzyme A binding domain containing 3 (ACBD3) and phosphatidylinositol 4-kinase IIIß (PI4KB). NAT6 could stabilize the PI4KB recruiter, ACBD3, by inhibiting the autophagy degradation pathway. This study provides a fresh insight into the relationship between N-acetyltransferase and viral infection.


Subject(s)
Enterovirus A, Human , Enterovirus Infections , N-Terminal Acetyltransferases , Phosphotransferases (Alcohol Group Acceptor) , Child , Child, Preschool , Humans , 1-Phosphatidylinositol 4-Kinase/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Antiviral Agents , Coenzyme A/metabolism , Coxsackievirus Infections , Enterovirus A, Human/physiology , Enterovirus Infections/metabolism , Enterovirus Infections/virology , Membrane Proteins/metabolism , N-Terminal Acetyltransferases/metabolism , Organelle Biogenesis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Virus Replication/physiology
16.
J Virol ; 98(1): e0155823, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38174926

ABSTRACT

Enterovirus A71 (EV-A71) can induce severe neurological complications and even fatal encephalitis in children, and it has caused several large outbreaks in Taiwan since 1998. We previously generated VP1 codon-deoptimized (VP1-CD) reverse genetics (rg) EV-A71 viruses (rgEV-A71s) that harbor a high-fidelity (HF) 3D polymerase. These VP1-CD-HF rgEV-A71s showed lower replication kinetics in vitro and decreased virulence in an Institute of Cancer Research (ICR) mouse model of EV-A71 infection, while still retaining their antigenicity in comparison to the wild-type virus. In this study, we aimed to further investigate the humoral and cellular immune responses elicited by VP1-CD-HF rgEV-A71s to assess the potential efficacy of these EV-A71 vaccine candidates. Following intraperitoneal (i.p.) injection of VP1-CD-HF rgEV-A71s in mice, we observed a robust induction of EV-A71-specific neutralizing IgG antibodies in the antisera after 21 days. Splenocytes isolated from VP1-CD-HF rgEV-A71s-immunized mice exhibited enhanced proliferative activities and cytokine production (IL-2, IFN-γ, IL-4, IL-6, and TNF-α) upon re-stimulation with VP1-CD-HF rgEV-A71, as compared to control mice treated with adjuvant only. Importantly, administration of antisera from VP1-CD-HF rgEV-A71s-immunized mice protected against lethal EV-A71 challenge in neonatal mice. These findings highlight that our generated VP1-CD-HF rgEV-A71 viruses are capable of inducing both cellular and humoral immune responses, supporting their potential as next-generation EV-A71 vaccines for combating EV-A71 infection.IMPORTANCEEV-A71 can cause severe neurological diseases and cause death in young children. Here, we report the development of synthetic rgEV-A71s with the combination of codon deoptimization and high-fidelity (HF) substitutions that generate genetically stable reverse genetics (rg) viruses as potential attenuated vaccine candidates. Our work provides insight into the development of low-virulence candidate vaccines through a series of viral genetic editing for maintaining antigenicity and genome stability and suggests a strategy for the development of an innovative next-generation vaccine against EV-A71.


Subject(s)
Capsid Proteins , Enterovirus A, Human , Enterovirus Infections , RNA-Dependent RNA Polymerase , Animals , Mice , Antibodies, Viral/immunology , Codon , Enterovirus A, Human/genetics , Enterovirus Infections/immunology , Vaccines, Attenuated , Capsid Proteins/genetics , Immunity, Humoral , Immunity, Cellular , Antibodies, Neutralizing/immunology , Viral Vaccines , Mice, Inbred ICR , Mice, Inbred BALB C , RNA-Dependent RNA Polymerase/genetics
17.
J Virol ; 98(6): e0026824, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38775480

ABSTRACT

Enteroviruses are the causative agents associated with several human and animal diseases, posing a significant threat to human and animal health. As one of the host immune defense strategies, innate immunity plays a crucial role in defending against invading pathogens, where the host utilizes a variety of mechanisms to inhibit or eliminate the pathogen. Here, we report a new strategy for the host to repress enterovirus replication by the 78 kDa glucose-regulated protein (GRP78), also known as heat shock protein family A member 5 (HSPA5). The GRP78 recognizes the EV-encoded RNA-dependent RNA polymerases (RdRPs) 3D protein and interacts with the nuclear factor kappa B kinase complex (CHUK) and subunit beta gene (IKBKB) to facilitate the phosphorylation and nuclear translocation of NF-κB, which induces the production of inflammatory factors and leads to a broad inhibition of enterovirus replication. These findings demonstrate a new role of GRP78 in regulating host innate immunity in response to viral infection and provide new insights into the mechanism underlying enterovirus replication and NF-κB activation.IMPORTANCEGRP78 is known as a molecular chaperone for protein folding and plays a critical role in maintaining protein folding and participating in cell proliferation, cell survival, apoptosis, and metabolism. However, the functions of GRP78 to participate in enterovirus genome replication and innate immune responses are rarely documented. In this study, we explored the functions of the EV-3D-interacting protein GRP78 and found that GRP78 inhibits enterovirus replication by activating NF-κB through binding to EV-F 3D and interacting with the NF-κB signaling molecules CHUK/IKBKB. This is the first report that GRP78 interacts with CHUK/IKBKB to activate the NF-κB signaling pathway, which leads to the expression of the proinflammatory cytokines and inhibition of enterovirus replication. These results demonstrate a unique mechanism of virus replication regulation by GRP78 and provide insights into the prevention and treatment of viral infections.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , I-kappa B Kinase , NF-kappa B , Viral Proteins , Virus Replication , Animals , Humans , Chlorocebus aethiops , Endoplasmic Reticulum Chaperone BiP/metabolism , Enterovirus/growth & development , Enterovirus/immunology , Enterovirus/metabolism , Enterovirus/physiology , Enterovirus Infections/virology , Enterovirus Infections/metabolism , Enterovirus Infections/immunology , Heat-Shock Proteins/metabolism , HEK293 Cells , Host-Pathogen Interactions/immunology , I-kappa B Kinase/metabolism , Immunity, Innate , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , NF-kappa B/metabolism , Phosphorylation , Protein Binding , RNA-Dependent RNA Polymerase/metabolism , Signal Transduction , Vero Cells , Viral Proteins/metabolism
18.
J Virol ; 98(2): e0190923, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38289118

ABSTRACT

Pyroptosis, a pro-inflammatory programmed cell death, has been implicated in the pathogenesis of coronavirus disease 2019 and other viral diseases. Gasdermin family proteins (GSDMs), including GSDMD and GSDME, are key regulators of pyroptotic cell death. However, the mechanisms by which virus infection modulates pyroptosis remain unclear. Here, we employed a mCherry-GSDMD fluorescent reporter assay to screen for viral proteins that impede the localization and function of GSDMD in living cells. Our data indicated that the main protease NSP5 of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) blocked GSDMD-mediated pyroptosis via cleaving residues Q29 and Q193 of GSDMD. While another SARS-CoV-2 protease, NSP3, cleaved GSDME at residue G370 but activated GSDME-mediated pyroptosis. Interestingly, respiratory enterovirus EV-D68-encoded proteases 3C and 2A also exhibit similar differential regulation on the functions of GSDMs by inactivating GSDMD but initiating GSDME-mediated pyroptosis. EV-D68 infection exerted oncolytic effects on human cancer cells by inducing pyroptotic cell death. Our findings provide insights into how respiratory viruses manipulate host cell pyroptosis and suggest potential targets for antiviral therapy as well as cancer treatment.IMPORTANCEPyroptosis plays a crucial role in the pathogenesis of coronavirus disease 2019, and comprehending its function may facilitate the development of novel therapeutic strategies. This study aims to explore how viral-encoded proteases modulate pyroptosis. We investigated the impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and respiratory enterovirus D68 (EV-D68) proteases on host cell pyroptosis. We found that SARS-CoV-2-encoded proteases NSP5 and NSP3 inactivate gasdermin D (GSDMD) but initiate gasdermin E (GSDME)-mediated pyroptosis, respectively. We also discovered that another respiratory virus EV-D68 encodes two distinct proteases 2A and 3C that selectively trigger GSDME-mediated pyroptosis while suppressing the function of GSDMD. Based on these findings, we further noted that EV-D68 infection triggers pyroptosis and produces oncolytic effects in human carcinoma cells. Our study provides new insights into the molecular mechanisms underlying virus-modulated pyroptosis and identifies potential targets for the development of antiviral and cancer therapeutics.


Subject(s)
Endopeptidases , Enterovirus D, Human , Host Microbial Interactions , Oncolytic Viruses , Pyroptosis , SARS-CoV-2 , Humans , Cell Line, Tumor , COVID-19/metabolism , COVID-19/therapy , COVID-19/virology , Endopeptidases/genetics , Endopeptidases/metabolism , Enterovirus D, Human/enzymology , Enterovirus D, Human/genetics , Enterovirus Infections/metabolism , Enterovirus Infections/virology , Gasdermins/antagonists & inhibitors , Gasdermins/genetics , Gasdermins/metabolism , Oncolytic Virotherapy , Oncolytic Viruses/enzymology , Oncolytic Viruses/genetics , SARS-CoV-2/enzymology , SARS-CoV-2/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
19.
J Virol ; 98(6): e0043424, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38690875

ABSTRACT

The globally reemerging respiratory pathogen enterovirus D68 (EV-D68) is implicated in outbreaks of severe respiratory illness and associated with acute flaccid myelitis. However, there remains a lack of effective treatments for EV-D68 infection. In this work, we found that the host Toll-like receptor 7 (TLR7) proteins, which function as powerful innate immune sensors, were selectively elevated in expression in response to EV-D68 infection. Subsequently, we investigated the impact of Vesatolimod (GS-9620), a Toll-like receptor 7 agonist, on EV-D68 replication. Our findings revealed that EV-D68 infection resulted in increased mRNA levels of TLR7. Treatment with Vesatolimod significantly inhibited EV-D68 replication [half maximal effective concentration (EC50) = 0.1427 µM] without inducing significant cytotoxicity at virucidal concentrations. Although Vesatolimod exhibited limited impact on EV-D68 attachment, it suppressed RNA replication and viral protein synthesis after virus entry. Vesatolimod broadly inhibited the replication of circulating isolated strains of EV-D68. Furthermore, our findings demonstrated that treatment with Vesatolimod conferred resistance to both respiratory and neural cells against EV-D68 infection. Overall, these results present a promising strategy for drug development by pharmacologically activating TLR7 to initiate an antiviral state in EV-D68-infected cells selectively.IMPORTANCEConventional strategies for antiviral drug development primarily focus on directly targeting viral proteases or key components, as well as host proteins involved in viral replication. In this study, based on our intriguing discovery that enterovirus D68 (EV-D68) infection specifically upregulates the expression of immune sensor Toll-like receptor 7 (TLR7) protein, which is either absent or expressed at low levels in respiratory cells, we propose a potential antiviral approach utilizing TLR7 agonists to activate EV-D68-infected cells into an anti-viral defense state. Notably, our findings demonstrate that pharmacological activation of TLR7 effectively suppresses EV-D68 replication in respiratory tract cells through a TLR7/MyD88-dependent mechanism. This study not only presents a promising drug candidate and target against EV-D68 dissemination but also highlights the potential to exploit unique alterations in cellular innate immune responses induced by viral infections, selectively inducing a defensive state in infected cells while safeguarding uninfected normal cells from potential adverse effects associated with therapeutic interventions.


Subject(s)
Antiviral Agents , Enterovirus D, Human , Toll-Like Receptor 7 , Virus Replication , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/metabolism , Humans , Virus Replication/drug effects , Enterovirus D, Human/drug effects , Antiviral Agents/pharmacology , Indoles/pharmacology , Enterovirus Infections/virology , Immunity, Innate/drug effects , Cell Line , Virus Internalization/drug effects , Pteridines
20.
PLoS Pathog ; 19(9): e1011465, 2023 09.
Article in English | MEDLINE | ID: mdl-37669302

ABSTRACT

Sex is a significant contributor to the outcome of human infections. Males are frequently more susceptible to viral, bacterial, and fungal infections, often attributed to weaker immune responses. In contrast, a heightened immune response in females enables better pathogen elimination but leaves females more predisposed to autoimmune diseases. Unfortunately, the underlying basis for sex-specific immune responses remains poorly understood. Here, we show a sex difference in the CD8+ T cell response to an enteric virus, Coxsackievirus B3 (CVB3). We found that CVB3 induced expansion of CD8+ T cells in female mice but not in male mice. CVB3 also increased the proportion and number of CD11ahiCD62Llo CD8+ T cells in female mice, indicative of activation. This response was independent of the inoculation route and type I interferon. Using a recombinant CVB3 virus expressing a model CD8+ T cell epitope, we found that the expansion of CD8+ T cells in females is viral-specific and not due to bystander activation. Finally, the depletion of CD8+ T cells, prior to infection, led to enhanced mortality, indicating that CD8+ T cells are protective against CVB3 in female mice. These data demonstrate that CVB3 induces a CD8+ T cell response in female mice and highlight the importance of sex-specific immune responses to viral pathogens.


Subject(s)
Enterovirus Infections , Interferon Type I , Orthopoxvirus , Humans , Animals , Female , Male , Mice , Antigens, Viral , CD8-Positive T-Lymphocytes , Epitopes, T-Lymphocyte
SELECTION OF CITATIONS
SEARCH DETAIL