Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.920
Filtrar
Más filtros

Colección OPSURU
Intervalo de año de publicación
1.
Cell ; 184(13): 3410-3425.e17, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34062120

RESUMEN

To control viral infection, vertebrates rely on both inducible interferon responses and less well-characterized cell-intrinsic responses composed of "at the ready" antiviral effector proteins. Here, we show that E3 ubiquitin ligase TRIM7 is a cell-intrinsic antiviral effector that restricts multiple human enteroviruses by targeting viral 2BC, a membrane remodeling protein, for ubiquitination and proteasome-dependent degradation. Selective pressure exerted by TRIM7 results in emergence of a TRIM7-resistant coxsackievirus with a single point mutation in the viral 2C ATPase/helicase. In cultured cells, the mutation helps the virus evade TRIM7 but impairs optimal viral replication, and this correlates with a hyperactive and structurally plastic 2C ATPase. Unexpectedly, the TRIM7-resistant virus has a replication advantage in mice and causes lethal pancreatitis. These findings reveal a unique mechanism for targeting enterovirus replication and provide molecular insight into the benefits and trade-offs of viral evolution imposed by a host restriction factor.


Asunto(s)
Enterovirus/fisiología , Enterovirus/patogenicidad , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Replicación Viral/fisiología , Adenosina Trifosfatasas/metabolismo , Animales , Línea Celular , Femenino , Humanos , Inflamación/patología , Ratones Endogámicos C57BL , Mutación/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteolisis , ARN Viral/metabolismo , Ubiquitina/metabolismo , Proteínas Virales/genética
2.
Cell ; 178(2): 275-289.e16, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31204099

RESUMEN

Positive-stranded RNA viruses extensively remodel host cell architecture to enable viral replication. Here, we examined the poorly understood formation of specialized membrane compartments that are critical sites for the synthesis of the viral genome. We show that the replication compartments (RCs) of enteroviruses are created through novel membrane contact sites that recruit host lipid droplets (LDs) to the RCs. Viral proteins tether the RCs to the LDs and interact with the host lipolysis machinery to enable transfer of fatty acids from LDs, thereby providing lipids essential for RC biogenesis. Inhibiting the formation of the membrane contact sites between LDs and RCs or inhibition of the lipolysis pathway disrupts RC biogenesis and enterovirus replication. Our data illuminate mechanistic and functional aspects of organelle remodeling in viral infection and establish that pharmacological targeting of contact sites linking viral and host compartments is a potential strategy for antiviral development.


Asunto(s)
Enterovirus/fisiología , Gotas Lipídicas/metabolismo , Replicación Viral , Retículo Endoplásmico/metabolismo , Células HeLa , Humanos , Lipólisis , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo , Internalización del Virus
3.
Cell ; 177(6): 1553-1565.e16, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31104841

RESUMEN

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.


Asunto(s)
Enterovirus Humano B/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase I/ultraestructura , Receptores Fc/metabolismo , Receptores Fc/ultraestructura , Cápside/metabolismo , Microscopía por Crioelectrón , Enterovirus , Enterovirus Humano B/patogenicidad , Infecciones por Enterovirus/metabolismo , Antígenos de Histocompatibilidad Clase I/fisiología , Humanos , Modelos Moleculares , Filogenia , Receptores Fc/fisiología , Virión , Internalización del Virus
4.
Cell ; 160(4): 619-630, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25679758

RESUMEN

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.


Asunto(s)
Vesículas Citoplasmáticas/virología , Infecciones por Enterovirus/transmisión , Enterovirus/fisiología , Macrófagos/virología , Vesículas Citoplasmáticas/química , Humanos , Macrófagos/citología , Fosfatidilserinas , Poliovirus/fisiología , ARN Viral/metabolismo , Rhinovirus/fisiología , Replicación Viral
5.
PLoS Pathog ; 20(3): e1012036, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38457376

RESUMEN

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.


Asunto(s)
Cardiovirus , Infecciones por Enterovirus , Enterovirus , Picornaviridae , Humanos , Enterovirus/fisiología , Virus de la Encefalomiocarditis/fisiología , Replicación Viral , Enterovirus Humano B/fisiología , Células HeLa
6.
PLoS Pathog ; 20(1): e1011967, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38271479

RESUMEN

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.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Humanos , Catelicidinas/farmacología , Internalización del Virus , Antivirales/metabolismo
7.
PLoS Pathog ; 20(2): e1012022, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38359079

RESUMEN

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.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Humanos , Enterovirus/metabolismo , Enterovirus Humano A/genética , Enterovirus Humano A/metabolismo , Membrana Celular/metabolismo , Línea Celular , Receptores Depuradores/genética , Receptores Depuradores/metabolismo , Proteínas de Membrana de los Lisosomas/genética
8.
PLoS Pathog ; 20(8): e1012388, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39102425

RESUMEN

Enteroviruses are a vast genus of positive-sense RNA viruses that cause diseases ranging from common cold to poliomyelitis and viral myocarditis. They encode a membrane-bound AAA+ ATPase, 2C, that has been suggested to serve several roles in virus replication, e.g. as an RNA helicase and capsid assembly factor. Here, we report the reconstitution of full-length, poliovirus 2C's association with membranes. We show that the N-terminal membrane-binding domain of 2C contains a conserved glycine, which is suggested by structure predictions to divide the domain into two amphipathic helix regions, which we name AH1 and AH2. AH2 is the main mediator of 2C oligomerization, and is necessary and sufficient for its membrane binding. AH1 is the main mediator of a novel function of 2C: clustering of membranes. Cryo-electron tomography reveal that several 2C copies mediate this function by localizing to vesicle-vesicle interfaces. 2C-mediated clustering is partially outcompeted by RNA, suggesting a way by which 2C can switch from an early role in coalescing replication organelles and lipid droplets, to a later role where 2C assists RNA replication and particle assembly. 2C is sufficient to recruit RNA to membranes, with a preference for double-stranded RNA (the replicating form of the viral genome). Finally, the in vitro reconstitution revealed that full-length, membrane-bound 2C has ATPase activity and ATP-independent, single-strand ribonuclease activity, but no detectable helicase activity. Together, this study suggests novel roles for 2C in membrane clustering, RNA membrane recruitment and cleavage, and calls into question a role of 2C as an RNA helicase. The reconstitution of functional, 2C-decorated vesicles provides a platform for further biochemical studies into this protein and its roles in enterovirus replication.


Asunto(s)
ARN Viral , Proteínas Virales , Replicación Viral , ARN Viral/metabolismo , ARN Viral/genética , Humanos , Replicación Viral/fisiología , Proteínas Virales/metabolismo , Proteínas Virales/genética , Poliovirus/metabolismo , Poliovirus/fisiología , Membrana Celular/metabolismo , Enterovirus/fisiología , Adenosina Trifosfatasas/metabolismo , Proteínas Portadoras , Proteínas no Estructurales Virales
9.
PLoS Pathog ; 20(9): e1012511, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39298524

RESUMEN

Molecular details of genome packaging are little understood for the majority of viruses. In enteroviruses (EVs), cleavage of the structural protein VP0 into VP4 and VP2 is initiated by the incorporation of RNA into the assembling virion and is essential for infectivity. We have applied a combination of bioinformatic, molecular and structural approaches to generate the first high-resolution structure of an intermediate in the assembly pathway, termed a provirion, which contains RNA and intact VP0. We have demonstrated an essential role of VP0 E096 in VP0 cleavage independent of RNA encapsidation and generated a new model of capsid maturation, supported by bioinformatic analysis. This provides a molecular basis for RNA-dependence, where RNA induces conformational changes required for VP0 maturation, but that RNA packaging itself is not sufficient to induce maturation. These data have implications for understanding production of infectious virions and potential relevance for future vaccine and antiviral drug design.


Asunto(s)
Proteínas de la Cápside , Ensamble de Virus , Ensamble de Virus/fisiología , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Humanos , ARN Viral/genética , ARN Viral/metabolismo , Virión/metabolismo , Enterovirus/fisiología , Cápside/metabolismo , Infecciones por Enterovirus/virología , Infecciones por Enterovirus/metabolismo
10.
PLoS Pathog ; 20(4): e1012159, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38662650

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales , Reacciones Cruzadas , Infecciones por Enterovirus , Vacuna Antipolio de Virus Inactivados , Animales , Ratones , Reacciones Cruzadas/inmunología , Anticuerpos Antivirales/inmunología , Infecciones por Enterovirus/inmunología , Infecciones por Enterovirus/prevención & control , Infecciones por Enterovirus/virología , Vacuna Antipolio de Virus Inactivados/inmunología , Vacuna Antipolio de Virus Inactivados/administración & dosificación , Vacunas de Partículas Similares a Virus/inmunología , Anticuerpos Neutralizantes/inmunología , Papio/inmunología , Humanos , Poliovirus/inmunología , Femenino , Formación de Anticuerpos/inmunología , Enterovirus/inmunología , Ratones Endogámicos BALB C , Enterovirus Humano D/inmunología
11.
PLoS Biol ; 21(1): e3001693, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36689548

RESUMEN

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.


Asunto(s)
Infecciones por Enterovirus , Enterovirus , Humanos , Enterovirus/fisiología , Infecciones por Enterovirus/virología , Sitios Internos de Entrada al Ribosoma , Factores de Iniciación de Péptidos/genética , Biosíntesis de Proteínas , ARN Viral/genética , ARN Viral/metabolismo , Interacciones Huésped-Patógeno
12.
Nucleic Acids Res ; 52(18): 11218-11233, 2024 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-39036953

RESUMEN

Enteroviruses contain conserved RNA structures at the extreme 5' end of their genomes that recruit essential proteins 3CD and PCBP2 to promote genome replication. However, the high-resolution structures and mechanisms of these replication-linked RNAs (REPLRs) are limited. Here, we determined the crystal structures of the coxsackievirus B3 and rhinoviruses B14 and C15 REPLRs at 1.54, 2.2 and 2.54 Šresolution, revealing a highly conserved H-type four-way junction fold with co-axially stacked sA-sD and sB-sC helices that are stabilized by a long-range A•C•U base-triple. Such conserved features observed in the crystal structures also allowed us to predict the models of several other enteroviral REPLRs using homology modeling, which generated models almost identical to the experimentally determined structures. Moreover, our structure-guided binding studies with recombinantly purified full-length human PCBP2 showed that two previously proposed binding sites, the sB-loop and 3' spacer, reside proximally and bind a single PCBP2. Additionally, the DNA oligos complementary to the 3' spacer, the high-affinity PCBP2 binding site, abrogated its interactions with enteroviral REPLRs, suggesting the critical roles of this single-stranded region in recruiting PCBP2 for enteroviral genome replication and illuminating the promising prospects of developing therapeutics against enteroviral infections targeting this replication platform.


Asunto(s)
Genoma Viral , Modelos Moleculares , Conformación de Ácido Nucleico , ARN Viral , Proteínas de Unión al ARN , Replicación Viral , ARN Viral/química , ARN Viral/genética , ARN Viral/metabolismo , Replicación Viral/genética , Humanos , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Sitios de Unión , Cristalografía por Rayos X , Enterovirus/genética , Enterovirus Humano B/genética
13.
J Virol ; 98(2): e0135823, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38226810

RESUMEN

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.


Asunto(s)
Infecciones por Enterovirus , Enterovirus , Enfermedad de Boca, Mano y Pie , Animales , Niño , Humanos , Ratones , Anticuerpos Antivirales , Modelos Animales de Enfermedad , Enterovirus/patogenicidad , Enterovirus/fisiología , Enterovirus Humano A , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Expresión Génica , Enfermedad de Boca, Mano y Pie/genética , Fosfatidilinositol 3-Quinasas , Virulencia
14.
J Virol ; 98(6): e0026824, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38775480

RESUMEN

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.


Asunto(s)
Chaperón BiP del Retículo Endoplásmico , Quinasa I-kappa B , FN-kappa B , Proteínas Virales , Replicación Viral , Animales , Humanos , Chlorocebus aethiops , Chaperón BiP del Retículo Endoplásmico/metabolismo , Enterovirus/crecimiento & desarrollo , Enterovirus/inmunología , Enterovirus/metabolismo , Enterovirus/fisiología , Infecciones por Enterovirus/virología , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/inmunología , Proteínas de Choque Térmico/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Quinasa I-kappa B/metabolismo , Inmunidad Innata , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , FN-kappa B/metabolismo , Fosforilación , Unión Proteica , ARN Polimerasa Dependiente del ARN/metabolismo , Transducción de Señal , Células Vero , Proteínas Virales/metabolismo
15.
J Virol ; 98(7): e0081324, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38904364

RESUMEN

Enteroviruses are single-stranded, positive-sense RNA viruses causing endoplasmic reticulum (ER) stress to induce or modulate downstream signaling pathways known as the unfolded protein responses (UPR). However, viral and host factors involved in the UPR related to viral pathogenesis remain unclear. In the present study, we aimed to identify the major regulator of enterovirus-induced UPR and elucidate the underlying molecular mechanisms. We showed that host Golgi-specific brefeldin A-resistant guanine nucleotide exchange factor 1 (GBF1), which supports enteroviruses replication, was a major regulator of the UPR caused by infection with enteroviruses. In addition, we found that severe UPR was induced by the expression of 3A proteins encoded in human pathogenic enteroviruses, such as enterovirus A71, coxsackievirus B3, poliovirus, and enterovirus D68. The N-terminal-conserved residues of 3A protein interact with the GBF1 and induce UPR through inhibition of ADP-ribosylation factor 1 (ARF1) activation via GBF1 sequestration. Remodeling and expansion of ER and accumulation of ER-resident proteins were observed in cells infected with enteroviruses. Finally, 3A induced apoptosis in cells infected with enteroviruses via activation of the protein kinase RNA-like endoplasmic reticulum kinase (PERK)/C/EBP homologous protein (CHOP) pathway of UPR. Pharmaceutical inhibition of PERK suppressed the cell death caused by infection with enteroviruses, suggesting the UPR pathway is a therapeutic target for treating diseases caused by infection with enteroviruses.IMPORTANCEInfection caused by several plus-stranded RNA viruses leads to dysregulated ER homeostasis in the host cells. The mechanisms underlying the disruption and impairment of ER homeostasis and its significance in pathogenesis upon enteroviral infection remain unclear. Our findings suggested that the 3A protein encoded in human pathogenic enteroviruses disrupts ER homeostasis by interacting with GBF1, a major regulator of UPR. Enterovirus-mediated infections drive ER into pathogenic conditions, where ER-resident proteins are accumulated. Furthermore, in such scenarios, the PERK/CHOP signaling pathway induced by an unresolved imbalance of ER homeostasis essentially drives apoptosis. Therefore, elucidating the mechanisms underlying the virus-induced disruption of ER homeostasis might be a potential target to mitigate the pathogenesis of enteroviruses.


Asunto(s)
Estrés del Retículo Endoplásmico , Retículo Endoplásmico , Factores de Intercambio de Guanina Nucleótido , Homeostasis , Respuesta de Proteína Desplegada , Humanos , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Infecciones por Enterovirus/virología , Infecciones por Enterovirus/metabolismo , Apoptosis , Enterovirus/fisiología , Enterovirus/metabolismo , Células HeLa , Replicación Viral , Factor 1 de Ribosilacion-ADP/metabolismo , Factor 1 de Ribosilacion-ADP/genética , Células HEK293 , Interacciones Huésped-Patógeno , Transducción de Señal , eIF-2 Quinasa/metabolismo
16.
PLoS Pathog ; 19(10): e1011662, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37788227

RESUMEN

Coxsackievirus A10 (CVA10) has recently emerged as one of the major causative agents of hand, foot, and mouth disease. CVA10 may also cause a variety of complications. No approved vaccine or drug is currently available for CVA10. The residues of CVA10 critical for viral attachment, infectivity and in vivo pathogenicity have not been identified by experiment. Here, we report the identification of CVA10 residues important for binding to cellular receptor KREMEN1. We identified VP2 N142 as a key receptor-binding residue by screening of CVA10 mutants resistant to neutralization by soluble KREMEN1 protein. The receptor-binding residue N142 is exposed on the canyon rim but highly conserved in all naturally occurring CVA10 strains, which provides a counterexample to the canyon hypothesis. Residue N142 when mutated drastically reduced receptor-binding activity, resulting in decreased viral attachment and infection in cell culture. More importantly, residue N142 when mutated reduced viral replication in limb muscle and spinal cord of infected mice, leading to lower mortality and less severe clinical symptoms. Additionally, residue N142 when mutated could decrease viral binding affinity to anti-CVA10 polyclonal antibodies and a neutralizing monoclonal antibody and render CVA10 resistant to neutralization by the anti-CVA10 antibodies. Overall, our study highlights the essential role of VP2 residue N142 of CVA10 in the interactions with KREMEN1 receptor and neutralizing antibodies and viral virulence in mice, facilitating the understanding of the molecular mechanisms of CVA10 infection and immunity. Our study also provides important information for rational development of antibody-based treatment and vaccines against CVA10 infection.


Asunto(s)
Anticuerpos Neutralizantes , Enterovirus , Animales , Ratones , Enterovirus/genética , Virulencia , Anticuerpos Antivirales
17.
PLoS Pathog ; 19(4): e1011317, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37071661

RESUMEN

Metabolism is key to cellular processes that underlie the ability of a virus to productively infect. Polyamines are small metabolites vital for many host cell processes including proliferation, transcription, and translation. Polyamine depletion also inhibits virus infection via diverse mechanisms, including inhibiting polymerase activity and viral translation. We showed that Coxsackievirus B3 (CVB3) attachment requires polyamines; however, the mechanism was unknown. Here, we report polyamines' involvement in translation, through a process called hypusination, promotes expression of cholesterol synthesis genes by supporting SREBP2 synthesis, the master transcriptional regulator of cholesterol synthesis genes. Measuring bulk transcription, we find polyamines support expression of cholesterol synthesis genes, regulated by SREBP2. Thus, polyamine depletion inhibits CVB3 by depleting cellular cholesterol. Exogenous cholesterol rescues CVB3 attachment, and mutant CVB3 resistant to polyamine depletion exhibits resistance to cholesterol perturbation. This study provides a novel link between polyamine and cholesterol homeostasis, a mechanism through which polyamines impact CVB3 infection.


Asunto(s)
Infecciones por Coxsackievirus , Infecciones por Enterovirus , Enterovirus , Humanos , Enterovirus/metabolismo , Poliaminas/metabolismo , Replicación Viral , Enterovirus Humano B
18.
PLoS Pathog ; 19(6): e1011420, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37262073

RESUMEN

Enterovirus A71 (EV-A71) infection is a major cause of severe hand, foot and mouth disease (HFMD) in young children. The characteristics of EV-A71 neutralizing antibodies in HFMD patients are not well understood. In this study, we identified and cloned EV-A71-neutralizing antibodies by single cell RNA and B cell receptor sequencing of peripheral blood mononuclear cells. From 145 plasmablasts, we identified two IgG1 monoclonal antibodies (mAbs) and six IgM mAbs that neutralized EV-A71. Four of the IgM mAbs harbor germline variable sequences and neutralize EV-A71 potently. Two genetically similar IgM antibodies from two patients have recurrent heavy chain variable domain gene usage and similar complementarity-determining region 3 sequences. We mapped the residues of EV-A71 critical for neutralization through selection of virus variants resistant to antibody neutralization in the presence of neutralizing mAbs. The residues critical for neutralization are conserved among EV-A71 genotypes. Epitopes for the two genetically similar antibodies overlap with the SCARB2 binding site of EV-A71. We used escape variants to measure the epitope-specific antibody response in acute phase serum samples from EV-A71 infected HFMD patients. We found that these epitopes are immunogenic and contributed to the neutralizing antibody response against the virus. Our findings advance understanding of antibody response to EV-A71 infection in young children and have translational potential: the IgM mAbs could potentially be used for prevention or treatment of EV-A71 infections.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Enfermedad de Boca, Mano y Pie , Niño , Humanos , Preescolar , Enterovirus/genética , Enterovirus Humano A/genética , Leucocitos Mononucleares , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Epítopos , Inmunoglobulina M , Anticuerpos Monoclonales , Antígenos Virales/genética
19.
PLoS Pathog ; 19(9): e1011673, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37721955

RESUMEN

The cellular protein GBF1, an activator of Arf GTPases (ArfGEF: Arf guanine nucleotide exchange factor), is recruited to the replication organelles of enteroviruses through interaction with the viral protein 3A, and its ArfGEF activity is required for viral replication, however how GBF1-dependent Arf activation supports the infection remains enigmatic. Here, we investigated the development of resistance of poliovirus, a prototype enterovirus, to increasing concentrations of brefeldin A (BFA), an inhibitor of GBF1. High level of resistance required a gradual accumulation of multiple mutations in the viral protein 2C. The 2C mutations conferred BFA resistance even in the context of a 3A mutant previously shown to be defective in the recruitment of GBF1 to replication organelles, and in cells depleted of GBF1, suggesting a GBF1-independent replication mechanism. Still, activated Arfs accumulated on the replication organelles of this mutant even in the presence of BFA, its replication was inhibited by a pan-ArfGEF inhibitor LM11, and the BFA-resistant phenotype was compromised in Arf1-knockout cells. Importantly, the mutations strongly increased the interaction of 2C with the activated form of Arf1. Analysis of other enteroviruses revealed a particularly strong interaction of 2C of human rhinovirus 1A with activated Arf1. Accordingly, the replication of this virus was significantly less sensitive to BFA than that of poliovirus. Thus, our data demonstrate that enterovirus 2Cs may behave like Arf1 effector proteins and that GBF1 but not Arf activation can be dispensable for enterovirus replication. These findings have important implications for the development of host-targeted anti-viral therapeutics.


Asunto(s)
Infecciones por Enterovirus , Enterovirus , Proteínas de Unión al GTP Monoméricas , Poliovirus , Humanos , Enterovirus/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Factor 1 de Ribosilacion-ADP/genética , Factor 1 de Ribosilacion-ADP/metabolismo , Células HeLa , Poliovirus/genética , Proteínas Virales/metabolismo , Antígenos Virales/metabolismo , Brefeldino A/farmacología , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo
20.
PLoS Pathog ; 19(2): e1011126, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36735752

RESUMEN

Foot-and-mouth disease, a class of animal diseases, is caused by foot-and-mouth disease virus (FMDV). The metabolic changes during FMDV infection remain unclear. Here, PK-15 cells, serum, and tonsils infected with FMDV were analyzed by metabolomics. A total of 284 metabolites in cells were significantly changed after FMDV infection, and most of them belong to amino acids and nucleotides. Further studies showed that FMDV infection significantly enhanced aspartate in vitro and in vivo. The amino acid transporter solute carrier family 38 member 8 (SLC38A8) was responsible for FMDV-upregulated aspartate. Enterovirus 71 (EV71) and Seneca Valley virus (SVV) infection also enhanced aspartate by SLC38A8. Aspartate aminotransferase activity was also elevated in FMDV-, EV71-, and SVV-infected cells, which may lead to reversible transition between the TCA cycle and amino acids synthesis. Aspartate and SLC38A8 were essential for FMDV, EV71, and SVV replication in cells. In addition, aspartate and SLC38A8 also promoted FMDV and EV71 replication in mice. Detailed analysis indicated that FMDV infection promoted the transfer of mTOR to lysosome to enhance interaction between mTOR and Rheb, and activated PI3K/AKT/TSC2/Rheb/mTOR/p70S6K1 pathway to promote viral replication. The mTORC1 signaling pathway was responsible for FMDV-induced SLC38A8 protein expression. For the first time, our data identified metabolic changes during FMDV infection. These data identified a novel mechanism used by FMDV to upregulate aspartate to promote viral replication and will provide new perspectives for developing new preventive strategies.


Asunto(s)
Enterovirus , Virus de la Fiebre Aftosa , Fiebre Aftosa , Animales , Ratones , Sistemas de Transporte de Aminoácidos Neutros , Ácido Aspártico/metabolismo , Virus de la Fiebre Aftosa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Replicación Viral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA