Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.308
Filtrar
1.
J Virol ; 98(5): e0035024, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38591900

RESUMO

Feline calicivirus (FCV) is one of the few members of the Caliciviridae family that grows well in cell lines and, therefore, serves as a surrogate to study the biology of other viruses in the family. Conley et al. (14) demonstrated that upon the receptor engagement to the capsid, FCV VP2 forms a portal-like assembly, which might provide a channel for RNA release. However, the process of calicivirus RNA release is not yet fully understood. Our findings suggest that the separation of the FCV capsid from its genome RNA (gRNA) occurs rapidly in the early endosomes of infected cells. Using a liposome model decorated with the FCV cell receptor fJAM-A, we demonstrate that FCV releases its gRNA into the liposomes by penetrating membranes under low pH conditions. Furthermore, we found that VP2, which is rich in hydrophobic residues at its N-terminus, functions as the pore-forming protein. When we substituted the VP2 N-terminal hydrophobic residues, the gRNA release efficacy of the FCV mutants decreased. In conclusion, our results suggest that in the acidic environment of early endosomes, FCV VP2 functions as the pore-forming protein to mediate gRNA release into the cytoplasm of infected cells. This provides insight into the mechanism of calicivirus genome release.IMPORTANCEResearch on the biology and pathogenicity of certain caliciviruses, such as Norovirus and Sapovirus, is hindered by the lack of easy-to-use cell culture system. Feline calicivirus (FCV), which grows effectively in cell lines, is used as a substitute. At present, there is limited understanding of the genome release mechanism in caliciviruses. Our findings suggest that FCV uses VP2 to pierce the endosome membrane for genome release and provide new insights into the calicivirus gRNA release mechanism.


Assuntos
Calicivirus Felino , Proteínas do Capsídeo , Endossomos , Genoma Viral , RNA Viral , Calicivirus Felino/genética , Calicivirus Felino/metabolismo , Calicivirus Felino/fisiologia , Gatos , Endossomos/virologia , Endossomos/metabolismo , Animais , RNA Viral/metabolismo , RNA Viral/genética , Linhagem Celular , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Infecções por Caliciviridae/virologia , Infecções por Caliciviridae/metabolismo , Liberação de Vírus , Capsídeo/metabolismo , Lipossomos/metabolismo
2.
J Med Virol ; 96(4): e29620, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38647027

RESUMO

Vertical transmission has been described following monkeypox virus (MPXV) infection in pregnant women. The presence of MPXV has been reported in the placenta from infected women, but whether pathogens colonize placenta remains unexplored. We identify trophoblasts as a target cell for MPXV replication. In a pan-microscopy approach, we decipher the specific infectious cycle of MPXV and inner cellular structures in trophoblasts. We identified the formation of a specialized region for viral morphogenesis and replication in placental cells. We also reported infection-induced cellular remodeling. We found that MPXV stimulates cytoskeleton reorganization with intercellular extensions for MPXV cell spreading specifically to trophoblastic cells. Altogether, the specific infectious cycle of MPXV in trophoblast cells and these protrusions that were structurally and morphologically similar to filopodia reveal new insights into the infection of MPXV.


Assuntos
Monkeypox virus , Pseudópodes , Trofoblastos , Trofoblastos/virologia , Humanos , Pseudópodes/virologia , Feminino , Gravidez , Monkeypox virus/fisiologia , Liberação de Vírus , Replicação Viral , Citoesqueleto/virologia , Placenta/virologia , Placenta/citologia , Vírion/ultraestrutura , Microscopia/métodos , Linhagem Celular
3.
PLoS Pathog ; 20(4): e1012133, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38662794

RESUMO

The discovery that extracellular vesicles (EVs) serve as carriers of virus particles calls for a reevaluation of the release strategies of non-enveloped viruses. Little is currently known about the molecular mechanisms that determine the release and composition of EVs produced by virus-infected cells, as well as conservation of these mechanisms among viruses. We previously described an important role for the Leader protein of the picornavirus encephalomyocarditis virus (EMCV) in the induction of virus-carrying EV subsets with distinct molecular and physical properties. EMCV L acts as a 'viral security protein' by suppressing host antiviral stress and type-I interferon (IFN) responses. Here, we tested the ability of functionally related picornavirus proteins of Theilers murine encephalitis virus (TMEV L), Saffold virus (SAFV L), and coxsackievirus B3 (CVB3 2Apro), to rescue EV and EV-enclosed virus release when introduced in Leader-deficient EMCV. We show that all viral security proteins tested were able to promote virus packaging in EVs, but that only the expression of EMCV L and CVB3 2Apro increased overall EV production. We provide evidence that one of the main antiviral pathways counteracted by this class of picornaviral proteins, i.e. the inhibition of PKR-mediated stress responses, affected EV and EV-enclosed virus release during infection. Moreover, we show that the enhanced capacity of the viral proteins EMCV L and CVB3 2Apro to promote EV-enclosed virus release is linked to their ability to simultaneously promote the activation of the stress kinase P38 MAPK. Taken together, we demonstrate that cellular stress pathways involving the kinases PKR and P38 are modulated by the activity of non-structural viral proteins to increase the release EV-enclosed viruses during picornavirus infections. These data shed new light on the molecular regulation of EV production in response to virus infection.


Assuntos
Vesículas Extracelulares , Picornaviridae , Proteínas Virais , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/virologia , Humanos , Picornaviridae/metabolismo , Picornaviridae/fisiologia , Proteínas Virais/metabolismo , Proteínas Virais/genética , Animais , eIF-2 Quinase/metabolismo , Liberação de Vírus/fisiologia , Camundongos , Theilovirus/metabolismo , Infecções por Cardiovirus/virologia , Infecções por Cardiovirus/metabolismo , Vírus da Encefalomiocardite/metabolismo , Vírus da Encefalomiocardite/fisiologia
4.
PLoS Pathog ; 20(1): e1011936, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38227586

RESUMO

Nuclear egress is an essential process in herpesvirus replication whereby nascent capsids translocate from the nucleus to the cytoplasm. This initial step of nuclear egress-budding at the inner nuclear membrane-is coordinated by the nuclear egress complex (NEC). Composed of the viral proteins UL31 and UL34, NEC deforms the membrane around the capsid as the latter buds into the perinuclear space. NEC oligomerization into a hexagonal membrane-bound lattice is essential for budding because NEC mutants designed to perturb lattice interfaces reduce its budding ability. Previously, we identified an NEC suppressor mutation capable of restoring budding to a mutant with a weakened hexagonal lattice. Using an established in-vitro budding assay and HSV-1 infected cell experiments, we show that the suppressor mutation can restore budding to a broad range of budding-deficient NEC mutants thereby acting as a universal suppressor. Cryogenic electron tomography of the suppressor NEC mutant lattice revealed a hexagonal lattice reminiscent of wild-type NEC lattice instead of an alternative lattice. Further investigation using x-ray crystallography showed that the suppressor mutation promoted the formation of new contacts between the NEC hexamers that, ostensibly, stabilized the hexagonal lattice. This stabilization strategy is powerful enough to override the otherwise deleterious effects of mutations that destabilize the NEC lattice by different mechanisms, resulting in a functional NEC hexagonal lattice and restoration of membrane budding.


Assuntos
Herpesviridae , Herpesvirus Humano 1 , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Supressão Genética , Núcleo Celular/metabolismo , Membrana Nuclear/metabolismo , Herpesviridae/metabolismo , Liberação de Vírus
5.
J Virol ; 98(2): e0178523, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38193690

RESUMO

The human pathogen herpes simplex virus 1 (HSV-1) produces a lifelong infection in the majority of the world's population. While the generalities of alpha herpesvirus assembly and egress pathways are known, the precise molecular and spatiotemporal details remain unclear. In order to study this aspect of HSV-1 infection, we engineered a recombinant HSV-1 strain expressing a pH-sensitive reporter, gM-pHluorin. Using a variety of fluorescent microscopy modalities, we can detect individual virus particles undergoing intracellular transport and exocytosis at the plasma membrane. We show that particles exit from epithelial cells individually, not bulk release of many particles at once, as has been reported for other viruses. In multiple cell types, HSV-1 particles accumulate over time at the cell periphery and cell-cell contacts. We show that this accumulation effect is the result of individual particles undergoing exocytosis at preferential sites and that these egress sites can contribute to cell-cell spread. We also show that the viral membrane proteins gE, gI, and US9, which have important functions in intracellular transport in neurons, are not required for preferential egress and clustering in non-neuronal cells. Importantly, by comparing HSV-1 to a related alpha herpesvirus, pseudorabies virus, we show that this preferential exocytosis and clustering effect are cell type dependent, not virus dependent. This preferential egress and clustering appear to be the result of the arrangement of the microtubule cytoskeleton, as virus particles co-accumulate at the same cell protrusions as an exogenous plus end-directed kinesin motor.IMPORTANCEAlpha herpesviruses produce lifelong infections in their human and animal hosts. The majority of people in the world are infected with herpes simplex virus 1 (HSV-1), which typically causes recurrent oral or genital lesions. However, HSV-1 can also spread to the central nervous system, causing severe encephalitis, and might also contribute to the development of neurodegenerative diseases. Many of the steps of how these viruses infect and replicate inside host cells are known in depth, but the final step, exiting from the infected cell, is not fully understood. In this study, we engineered a novel variant of HSV-1 that allows us to visualize how individual virus particles exit from infected cells. With this imaging assay, we investigated preferential egress site formation in certain cell types and their contribution to the cell-cell spread of HSV-1.


Assuntos
Exocitose , Herpes Simples , Herpesvirus Humano 1 , Liberação de Vírus , Animais , Humanos , Transporte Biológico , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Neurônios
6.
J Virol ; 98(2): e0189923, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38294245

RESUMO

After Epstein-Barr virus (EBV) genome replication and encapsidation in the nucleus, nucleocapsids are translocated into the cytoplasm for subsequent tegumentation and maturation. The EBV BGLF4 kinase, which induces partial disassembly of the nuclear lamina, and the nuclear egress complex BFRF1/BFLF2 coordinately facilitate the nuclear egress of nucleocapsids. Here, we demonstrate that within EBV reactivated epithelial cells, viral capsids, tegument proteins, and glycoproteins are clustered in the juxtanuclear concave region, accompanied by redistributed cytoplasmic organelles and the cytoskeleton regulator IQ-domain GTPase-activation protein 1 (IQGAP1), close to the microtubule-organizing center (MTOC). The assembly compartment (AC) structure was diminished in BGLF4-knockdown TW01-EBV cells and BGLF4-knockout bacmid-carrying TW01 cells, suggesting that the formation of AC structure is BGLF4-dependent. Notably, glycoprotein gp350/220 was observed by confocal imaging to be distributed in the perinuclear concave region and surrounded by the endoplasmic reticulum (ER) membrane marker calnexin, indicating that the AC may be located within a globular structure derived from ER membranes, adjacent to the outer nuclear membrane. Moreover, the viral capsid protein BcLF1 and tegument protein BBLF1 were co-localized with IQGAP1 near the cytoplasmic membrane in the late stage of replication. Knockdown of IQGAP1 did not affect the AC formation but decreased virion release from both TW01-EBV and Akata+ cells, suggesting IQGAP1-mediated trafficking regulates EBV virion release. The data presented here show that BGLF4 is required for cytoskeletal rearrangement, coordination with the redistribution of cytoplasmic organelles and IQGAP1 for virus maturation, and subsequent IQGAP1-dependent virion release.IMPORTANCEEBV genome is replicated and encapsidated in the nucleus, and the resultant nucleocapsids are translocated to the cytoplasm for subsequent virion maturation. We show that a cytoplasmic AC, containing viral proteins, markers of the endoplasmic reticulum, Golgi, and endosomes, is formed in the juxtanuclear region of epithelial and B cells during EBV reactivation. The viral BGLF4 kinase contributes to the formation of the AC. The cellular protein IQGAP1 is also recruited to the AC and partially co-localizes with the virus capsid protein BcLF1 and tegument protein BBLF1 in EBV-reactivated cells, dependent on the BGLF4-induced cytoskeletal rearrangement. In addition, virion release was attenuated in IQGAP1-knockdown epithelial and B cells after reactivation, suggesting that IQGAP1-mediated trafficking may regulate the efficiency of virus maturation and release.


Assuntos
Citoplasma , Herpesvirus Humano 4 , Proteínas Serina-Treonina Quinases , Proteínas Virais , Vírion , Montagem de Vírus , Liberação de Vírus , Proteínas Ativadoras de ras GTPase , Humanos , Proteínas do Capsídeo/metabolismo , Citoplasma/metabolismo , Citoplasma/virologia , Infecções por Vírus Epstein-Barr/metabolismo , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/química , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/crescimento & desenvolvimento , Herpesvirus Humano 4/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Proteínas Virais/metabolismo , Vírion/química , Vírion/crescimento & desenvolvimento , Vírion/metabolismo , Montagem de Vírus/fisiologia , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Complexo de Golgi/metabolismo
7.
J Virol ; 98(2): e0190023, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38289107

RESUMO

The endosomal sorting complex required for transport (ESCRT) is a conserved protein machine mediating membrane remodeling and scission. In the context of viral infection, different components of the ESCRT-III complex, which serve as the core machinery to catalyze membrane fission, are involved in diverse viruses' entry, replication, and/or budding. However, the interplay between ESCRT-III and viral factors in the virus life cycle, especially for that of large enveloped DNA viruses, is largely unknown. Recently, the ESCRT-III components Vps2B, Vps20, Vps24, Snf7, Vps46, and Vps60 were determined for entry and/or egress of the baculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV). Here, we identified the final three ESCRT-III components Chm7, Ist1, and Vps2A of Spodoptera frugiperda. Overexpression of the dominant-negative forms of these proteins or RNAi downregulation of their transcripts significantly reduced infectious budded viruses (BVs) production of AcMNPV. Quantitative PCR together with confocal and transmission electron microscopy analysis revealed that these proteins were required for internalization and trafficking of BV during entry and egress of nucleocapsids. In infected Sf9 cells, nine ESCRT-III components were distributed on the nuclear envelope and plasma membrane, and except for Chm7, the other components were also localized to the intranuclear ring zone. Y2H and BiFC analysis revealed that 42 out of 64 BV-related proteins including 35 BV structural proteins and 7 non-BV structural proteins interacted with single or multiple ESCRT-III components. By further mapping the interactome of 64 BV-related proteins, we established the interaction networks of ESCRT-III and the viral protein complexes involved in BV entry and egress.IMPORTANCEFrom archaea to eukaryotes, the endosomal sorting complex required for transport (ESCRT)-III complex is hijacked by many enveloped and nonenveloped DNA or RNA viruses for efficient replication. However, the mechanism of ESCRT-III recruitment, especially for that of large enveloped DNA viruses, remains elusive. Recently, we found the ESCRT-III components Vps2B, Vps20, Vps24, Snf7, Vps46, and Vps60 are necessary for the entry and/or egress of budded viruses (BVs) of Autographa californica multiple nucleopolyhedrovirus. Here, we demonstrated that the other three ESCRT-III components Chm7, Ist1, and Vps2A play similar roles in BV infection. By determining the subcellular localization of ESCRT-III components in infected cells and mapping the interaction of nine ESCRT-III components and 64 BV-related proteins, we built the interaction networks of ESCRT-III and the viral protein complexes involved in BV entry and egress. These studies provide a fundamental basis for understanding the mechanism of the ESCRT-mediated membrane remodeling for replication of baculoviruses.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte , Interações entre Hospedeiro e Microrganismos , Nucleopoliedrovírus , Spodoptera , Proteínas Virais , Internalização do Vírus , Liberação de Vírus , Animais , Complexos Endossomais de Distribuição Requeridos para Transporte/química , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/ultraestrutura , Nucleopoliedrovírus/metabolismo , Nucleopoliedrovírus/fisiologia , Nucleopoliedrovírus/ultraestrutura , Spodoptera/citologia , Spodoptera/metabolismo , Spodoptera/ultraestrutura , Spodoptera/virologia , Proteínas Virais/química , Proteínas Virais/metabolismo , Proteínas Virais/ultraestrutura , Replicação Viral , Transporte Biológico , Células Sf9
8.
Antiviral Res ; 221: 105786, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38147902

RESUMO

The endosomal sorting complex required for transport (ESCRT) machinery plays a significant role in the spread of human viruses. However, our understanding of how the host ESCRT machinery responds to viral infection remains limited. Emerging evidence suggests that the ESCRT machinery can be hijacked by viruses of different families to enhance their replication. Throughout their life cycle, these viruses can interfere with or exploit ESCRT-mediated physiological processes to increase their chances of infecting the host. In contrast, to counteract virus infection, the interferon-stimulated gene 15 (ISG15) or the E3 ISG15-protein ligase (HERC5) system within the infected cells is activated to degrade the ESCRT proteins. Many retroviral and RNA viral proteins have evolved "late (L) domain" motifs, which enable them to recruit host ESCRT subunit proteins to facilitate virus transport, replication, budding, mature, and even endocytosis, Therefore, the L domain motifs and ESCRT subunit proteins could serve as promising drug targets for antiviral therapy. This review investigated the composition and essential functions of the ESCRT, shedding light on the impact of ESCRT subunits and viral L domain motifs on the replication of viruses. Furthermore, the antiviral effects facilitated by the ESCRT machinery have been investigated, aiming to provide valuable insights to guide the development and utilization of antiviral drugs.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte , Viroses , Humanos , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Transporte Proteico , Proteínas Virais/metabolismo , Interferons/metabolismo , Ubiquitina-Proteína Ligases , Replicação Viral , Liberação de Vírus
9.
Viruses ; 15(12)2023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-38140670

RESUMO

Hepatitis C virus (HCV) is a positive-sense, single-stranded RNA virus that causes chronic hepatitis, liver cirrhosis and hepatocellular carcinoma. The release of infectious HCV particles from infected hepatocytes is a crucial step in viral dissemination and disease progression. While the exact mechanisms of HCV particle release remain poorly understood, emerging evidence suggests that HCV utilizes intracellular membrane trafficking and secretory pathways. These pathways include the Golgi secretory pathway and the endosomal trafficking pathways, such as the recycling endosome pathway and the endosomal sorting complex required for transport (ESCRT)-dependent multivesicular bodies (MVBs) pathway. This review provides an overview of recent advances in understanding the release of infectious HCV particles, with a particular focus on the involvement of the host cell factors that participate in HCV particle release. By summarizing the current knowledge in this area, this review aims to contribute to a better understanding of endosomal pathways involved in the extracellular release of HCV particles and the development of novel antiviral strategies.


Assuntos
Hepatite A , Hepatite C , Humanos , Hepacivirus/metabolismo , Endossomos/metabolismo , Vírion/metabolismo , Liberação de Vírus , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo
10.
Viruses ; 15(12)2023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-38140530

RESUMO

HIV-1 budding as well as many other cellular processes require the Endosomal Sorting Complex Required for Transport (ESCRT) machinery. Understanding the architecture of the native ESCRT-III complex at HIV-1 budding sites is limited due to spatial resolution and transient ESCRT-III recruitment. Here, we developed a drug-inducible transient HIV-1 budding inhibitory tool to enhance the ESCRT-III lifetime at budding sites. We generated autocleavable CHMP2A, CHMP3, and CHMP4B fusion proteins with the hepatitis C virus NS3 protease. We characterized the CHMP-NS3 fusion proteins in the absence and presence of protease inhibitor Glecaprevir with regard to expression, stability, localization, and HIV-1 Gag VLP budding. Immunoblotting experiments revealed rapid and stable accumulation of CHMP-NS3 fusion proteins. Notably, upon drug administration, CHMP2A-NS3 and CHMP4B-NS3 fusion proteins substantially decrease VLP release while CHMP3-NS3 exerted no effect but synergized with CHMP2A-NS3. Localization studies demonstrated the relocalization of CHMP-NS3 fusion proteins to the plasma membrane, endosomes, and Gag VLP budding sites. Through the combined use of transmission electron microscopy and video-microscopy, we unveiled drug-dependent accumulation of CHMP2A-NS3 and CHMP4B-NS3, causing a delay in HIV-1 Gag-VLP release. Our findings provide novel insight into the functional consequences of inhibiting ESCRT-III during HIV-1 budding and establish new tools to decipher the role of ESCRT-III at HIV-1 budding sites and other ESCRT-catalyzed cellular processes.


Assuntos
HIV-1 , HIV-1/fisiologia , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Liberação de Vírus/fisiologia
11.
PLoS One ; 18(11): e0292833, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37922253

RESUMO

The innate immune response is a first-line defense mechanism triggered by rabies virus (RABV). Interferon (IFN) signaling and ISG products have been shown to confer resistance to RABV at various stages of the virus's life cycle. Human tetherin, also known as bone marrow stromal cell antigen 2 (hBST2), is a multifunctional transmembrane glycoprotein induced by IFN that has been shown to effectively counteract many viruses through diverse mechanisms. Here, we demonstrate that hBST2 inhibits RABV budding by tethering new virions to the cell surface. It was observed that release of virus-like particles (VLPs) formed by RABV G (RABV-G VLPs), but not RABV M (RABV-G VLPs), were suppressed by hBST2, indicating that RABV-G has a specific effect on the hBST2-mediated restriction of RABV. The ability of hBST2 to prevent the release of RABV-G VLPs and impede RABV growth kinetics is retained even when hBST2 has mutations at dimerization and/or glycosylation sites, making hBST2 an antagonist to RABV, with multiple mechanisms possibly contributing to the hBST2-mediated suppression of RABV. Our findings expand the knowledge of host antiviral mechanisms that control RABV infection.


Assuntos
Vírus da Raiva , Raiva , Humanos , Vírus da Raiva/fisiologia , Raiva/prevenção & controle , Glicosilação , Asparagina/metabolismo , Cisteína/metabolismo , Dimerização , Liberação de Vírus , Antígeno 2 do Estroma da Médula Óssea/genética , Antígenos CD/metabolismo , Proteínas Ligadas por GPI/metabolismo
12.
J Virol ; 97(10): e0083623, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37787529

RESUMO

IMPORTANCE: Herpesviruses are able to disseminate in infected hosts despite development of a strong immune response. Their ability to do this relies on a specialized process called cell-to-cell spread in which newly assembled virus particles are trafficked to plasma membrane surfaces that abut adjacent uninfected cells. The mechanism of cell-to-cell spread is obscure, and little is known about whether or how it is regulated in different cells. We show here that a viral protein with a well-characterized role in promoting spread from neurons has an opposite, inhibitory role in other cells.


Assuntos
Estruturas da Membrana Celular , Núcleo Celular , Células Epiteliais , Herpesvirus Humano 1 , Peptídeos e Proteínas de Sinalização Intracelular , Lipoproteínas , Mutação , Proteínas Virais , Liberação de Vírus , Transporte Biológico , Estruturas da Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipoproteínas/metabolismo , Neurônios/metabolismo , Neurônios/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vírion/genética , Vírion/metabolismo
13.
Sheng Wu Gong Cheng Xue Bao ; 39(10): 3948-3965, 2023 Oct 25.
Artigo em Chinês | MEDLINE | ID: mdl-37877384

RESUMO

Endosomal sorting complex required for transport (ESCRT) system drives various cellular processes, including endosome sorting, organelle biogenesis, vesicle transport, maintenance of plasma membrane integrity, membrane fission during cytokinesis, nuclear membrane reformation after mitosis, closure of autophagic vacuoles, and enveloped virus budding. Increasing evidence suggests that the ESCRT system can be hijacked by different family viruses for their proliferation. At different stages of the virus life cycle, viruses can interfere with or exploit ESCRT-mediated physiological processes in various ways to maximize their chance of infecting the host. In addition, many retroviral and RNA viral proteins possess "late domain" motifs, which can recruit host ESCRT subunit proteins to assist in virus endocytosis, transport, replicate, budding and efflux. Therefore, the "late domain" motifs of viruses and ESCRT subunit proteins could serve as promising drug targets in antiviral therapy. This review focuses on the composition and functions of the ESCRT system, the effects of ESCRT subunits and virus "late domain" motifs on viral replication, and the antiviral effects mediated by the ESCRT system, aiming to provide a reference for the development and utilization of antiviral drugs.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte , Vírus , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Vírus/metabolismo , Transporte Proteico , Replicação Viral , Endossomos/metabolismo , Liberação de Vírus
14.
Sci Adv ; 9(42): eadj4198, 2023 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-37862421

RESUMO

Virus-induced changes in host lipid metabolism are an important but poorly understood aspect of viral pathogenesis. By combining nontargeted lipidomics analyses of infected cells and purified extracellular quasi-enveloped virions with high-throughput RNA sequencing and genetic depletion studies, we show that hepatitis A virus, an hepatotropic picornavirus, broadly manipulates the host cell lipid environment, enhancing synthesis of ceramides and other sphingolipids and transcriptionally activating acyl-coenzyme A synthetases and fatty acid elongases to import and activate long-chain fatty acids for entry into the fatty acid elongation cycle. Phospholipids with very-long-chain acyl tails (>C22) are essential for genome replication, whereas increases in sphingolipids support assembly and release of quasi-enveloped virions wrapped in membranes highly enriched for sphingomyelin and very-long-chain ceramides. Our data provide insight into how a pathogenic virus alters lipid flux in infected hepatocytes and demonstrate a distinction between lipid species required for viral RNA synthesis versus nonlytic quasi-enveloped virus release.


Assuntos
Hepatovirus , RNA Viral , Hepatovirus/metabolismo , RNA Viral/genética , Replicação do RNA , Liberação de Vírus , Replicação Viral/fisiologia , Ácidos Graxos/metabolismo , Esfingolipídeos , Ceramidas
15.
EMBO Rep ; 24(12): e57224, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-37818801

RESUMO

The antiviral restriction factor, tetherin, blocks the release of several different families of enveloped viruses, including the Coronaviridae. Tetherin is an interferon-induced protein that forms parallel homodimers between the host cell and viral particles, linking viruses to the surface of infected cells and inhibiting their release. We demonstrate that SARS-CoV-2 infection causes tetherin downregulation and that tetherin depletion from cells enhances SARS-CoV-2 viral titres. We investigate the potential viral proteins involved in abrogating tetherin function and find that SARS-CoV-2 ORF3a reduces tetherin localisation within biosynthetic organelles where Coronaviruses bud, and increases tetherin localisation to late endocytic organelles via reduced retrograde recycling. We also find that expression of Spike protein causes a reduction in cellular tetherin levels. Our results confirm that tetherin acts as a host restriction factor for SARS-CoV-2 and highlight the multiple distinct mechanisms by which SARS-CoV-2 subverts tetherin function.


Assuntos
Antígeno 2 do Estroma da Médula Óssea , COVID-19 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Liberação de Vírus , Humanos , Antígeno 2 do Estroma da Médula Óssea/antagonistas & inibidores , Antígeno 2 do Estroma da Médula Óssea/metabolismo , COVID-19/virologia , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus/genética
16.
J Virol ; 97(10): e0042623, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37830820

RESUMO

IMPORTANCE: Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), the virus responsible for coronavirus disease 2019 (COVID-19), has caused a global public health crisis. The E protein, a structural protein found in this virus particle, is also known to be a viroporin. As such, it forms oligomeric ion channels or pores in the host cell membrane. However, the relationship between these two functions is poorly understood. In this study, we showed that the roles of E protein in virus particle and viroporin formation are distinct. This study contributes to the development of drugs that inhibit SARS-CoV-2 virus particle formation. Additionally, we designed a highly sensitive and high-throughput virus-like particle detection system using the HiBiT tag, which is a useful tool for studying the release of SARS-CoV-2.


Assuntos
Proteínas do Envelope de Coronavírus , SARS-CoV-2 , Humanos , COVID-19 , Lisossomos/metabolismo , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/metabolismo , Proteínas Viroporinas/metabolismo , Proteínas do Envelope de Coronavírus/metabolismo , Motivos de Aminoácidos , Liberação de Vírus
17.
Viruses ; 15(9)2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37766274

RESUMO

The endosomal sorting complex required for transport (ESCRT) machinery is essential for the budding of retroviruses such as human immunodeficiency virus (HIV) and bovine foamy virus (BFV), which rely on their late domain to recruit ESCRT complexes to facilitate budding. However, the impact of intracellular host proteins on BFV budding remains poorly understood. In this study, we aimed to investigate the impact of CCL2 on BFV budding and interactions with key host proteins. Our results indicate that CCL2 promotes BFV budding in an ALG-2-interacting protein X (Alix)-dependent manner by enhancing the interaction between Alix and BFV Gag (BGag). Notably, we found a link between Alix, BGag and CCL2, with Alix mediating the interaction between the latter two. Furthermore, we observed that natural host bovine CCL2 also has a facilitating role in the budding process of BFV, similar to human CCL2. Taken together, these results demonstrate that CCL2 promotes BFV budding by enhancing the Alix-BGag association.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte , Spumavirus , Humanos , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Spumavirus/metabolismo , Ligantes , Proteínas de Ciclo Celular/metabolismo , Quimiocinas/metabolismo , Liberação de Vírus
18.
Educ. med. super ; 37(3)sept. 2023. ilus
Artigo em Espanhol | LILACS, CUMED | ID: biblio-1528556

RESUMO

Introducción: La viruela símica es una enfermedad zoonótica que también se trasmite de persona a persona por contacto estrecho. En el brote actual hasta el 31 de agosto de 2022 se reportaban 50 496 casos diagnosticados en 101 países, por lo que se consideró una situación preocupante por la Organización Mundial de la Salud. Objetivo: Exponer información actualizada sobre la viruela símica en el contexto sanitario actual. Métodos: Se realizó una búsqueda de literatura científica en las bases de datos ScienceDirect, PubMed/Medline, SciELO y Google Académico, mediante los descriptores o palabras relacionadas con la enfermedad, para encontrar revisiones, comunicados, informes, distintos artículos de revistas, entre otros documentos especializados de producción científica. Se seleccionó un total de 30 citas, actualizadas en su totalidad. Desarrollo: Desde su identificación en humanos se han reportado brotes de viruela símica en varios países; el más preocupante, ha sido el de reciente declaración en 2022, debido a la presencia de casos en países no endémicos, con un alcance geográfico extenso. Las manifestaciones clínicas pueden cursar con síntomas leves, como erupciones en la cara y el resto del cuerpo, fiebre, cefalea, mialgias y fatiga, por lo que no constituye una enfermedad potencialmente mortal; sin embargo, de presentarse comorbilidades la evolución podría ser tórpida. Conclusiones: La presencia de casos de viruela símica en humanos se ha mantenido desde su aparición, sin encontrar un tratamiento específico y vacunas autorizadas para su administración, lo que podría generar un aumento de contagios y fallecidos(AU)


Introduction: Mpox is a zoonotic disease also transmitted from person to person by close contact. The current outbreak, up to August 31, 2022, reported 50 496 diagnosed cases from 101 countries; therefore; it was considered a situation of concern by the World Health Organization. Objective: To present updated information on Mpox in the current health context. Methods: A scientific literature search was carried out in the databases ScienceDirect, PubMed/Medline, SciELO and Google Scholar, using descriptors or words related to the disease, in order to find reviews, communications, reports, different journal articles, among other specialized documents of scientific production. A total of 30 entirely updated citations were selected. Development: Since Mpox was identified in humans, outbreaks of the disease have been reported in several countries; the most worrisome has been reported recently in 2022, due to the presence of cases in nonendemic countries, with an extensive geographical scope. The clinical manifestations may occur with mild symptoms, such as rash on the face or the rest of the body, fever, headache, myalgia and fatigue; therefore, it is not a potentially mortal disease. However, in case of comorbidity, the evolution could be torpid. Conclusions: Mpox cases in humans has been present since its appearance, without any specific treatment or vaccines authorized to be administered, which could generate an increase in contagions and deaths(AU)


Assuntos
Humanos , Conhecimentos, Atitudes e Prática em Saúde , Mpox/diagnóstico , Mpox/história , Mpox/mortalidade , Mpox/prevenção & controle , Mpox/transmissão , Liberação de Vírus , Orthopoxvirus
19.
J Cell Biol ; 222(9)2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37516914

RESUMO

Herpes simplex virus (HSV-1) progeny form in the nucleus and exit to successfully infect other cells. Newly formed capsids navigate complex chromatin architecture to reach the inner nuclear membrane (INM) and egress. Here, we demonstrate by transmission electron microscopy (TEM) that HSV-1 capsids traverse heterochromatin associated with trimethylation on histone H3 lysine 27 (H3K27me3) and the histone variant macroH2A1. Through chromatin profiling during infection, we revealed global redistribution of these marks whereby massive host genomic regions bound by macroH2A1 and H3K27me3 correlate with decreased host transcription in active compartments. We found that the loss of these markers resulted in significantly lower viral titers but did not impact viral genome or protein accumulation. Strikingly, we discovered that loss of macroH2A1 or H3K27me3 resulted in nuclear trapping of capsids. Finally, by live-capsid tracking, we quantified this decreased capsid movement. Thus, our work demonstrates that HSV-1 takes advantage of the dynamic nature of host heterochromatin formation during infection for efficient nuclear egress.


Assuntos
Herpesvirus Humano 1 , Heterocromatina , Liberação de Vírus , Núcleo Celular/virologia , Cromatina , Herpesvirus Humano 1/genética , Heterocromatina/genética , Histonas/genética , Capsídeo/ultraestrutura
20.
J Virol ; 97(6): e0043723, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37195206

RESUMO

Enveloped viruses undergo a complex multistep process of assembly, maturation, and release into the extracellular space utilizing host secretory machinery. Several studies of the herpesvirus subfamily have shown that secretory vesicles derived from the trans-Golgi network (TGN) or endosomes transport virions into the extracellular space. However, the regulatory mechanism underlying the release of Epstein-Barr virus, a human oncovirus, remains unclear. We demonstrate that disruption of BBLF1, a tegument component, suppressed viral release and resulted in the accumulation of viral particles on the inner side of the vesicular membrane. Organelle separation revealed the accumulation of infectious viruses in fractions containing vesicles derived from the TGN and late endosomes. Deficiency of an acidic amino acid cluster in BBLF1 reduced viral secretion. Moreover, truncational deletion of the C-terminal region of BBLF1 increased infectious virus production. These findings suggest that BBLF1 regulates the viral release pathway and reveal a new aspect of tegument protein function. IMPORTANCE Several viruses have been linked to the development of cancer in humans. Epstein-Barr virus (EBV), the first identified human oncovirus, causes a wide range of cancers. Accumulating literature has demonstrated the role of viral reactivation in tumorigenesis. Elucidating the functions of viral lytic genes induced by reactivation, and the mechanisms of lytic infection, is essential to understanding pathogenesis. Progeny viral particles synthesized during lytic infection are released outside the cell after the assembly, maturation, and release steps, leading to further infection. Through functional analysis using BBLF1-knockout viruses, we demonstrated that BBLF1 promotes viral release. The acidic amino acid cluster in BBLF1 was also important for viral release. Conversely, mutants lacking the C terminus exhibited more efficient virus production, suggesting that BBLF1 is involved in the fine-tuning of progeny release during the EBV life cycle.


Assuntos
Herpesvirus Humano 4 , Vesículas Secretórias , Proteínas Virais , Liberação de Vírus , Replicação Viral , Humanos , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/fisiologia , Vesículas Secretórias/metabolismo , Vesículas Secretórias/virologia , Vírion/fisiologia , Replicação Viral/fisiologia , Células HEK293 , Proteínas Virais/metabolismo , Liberação de Vírus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...