Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.413
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(43): e2408509121, 2024 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-39413137

RESUMEN

CRISPR-associated (Cas) endonucleases and their derivatives are widespread tools for the targeted genetic modification of both prokaryotic and eukaryotic genomes. A critical step of all CRISPR-Cas technologies is the delivery of the Cas endonuclease to the target cell. Here, we investigate the possibility of using bacterial conjugation to translocate Cas proteins into recipient bacteria. Conjugative relaxases are translocated through a type IV secretion system into the recipient cell, covalently attached to the transferred DNA strand. We fused relaxase R388-TrwC with the endonuclease Cas12a and confirmed that it can be transported through a T4SS. The fusion protein maintained its activity upon translocation by conjugation into the recipient cell, as evidenced by the induction of the SOS signal resulting from DNA breaks produced by the endonuclease in the recipient cell, and the detection of mutations at the target position. We further show how a template DNA provided on the transferred DNA can be used to introduce specific mutations. The guide RNA can also be encoded by the transferred DNA, enabling its production in the recipient cells where it can form a complex with the Cas nuclease transferred as a protein. This self-contained setup enables to target wild-type bacterial cells. Finally, we extended this strategy to the delivery of relaxases fused to base editors. Using TrwC and MobA relaxases as drivers, we achieved precise editing of transconjugants. Thus, conjugation provides a delivery system for Cas-derived editing tools, bypassing the need to deliver and express a cas gene in the target cells.


Asunto(s)
Sistemas CRISPR-Cas , Conjugación Genética , Sistemas de Secreción Tipo IV , Sistemas de Secreción Tipo IV/metabolismo , Sistemas de Secreción Tipo IV/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Asociadas a CRISPR/metabolismo , Proteínas Asociadas a CRISPR/genética , Nucleoproteínas/metabolismo , Nucleoproteínas/genética , Endodesoxirribonucleasas/metabolismo , Endodesoxirribonucleasas/genética , Edición Génica/métodos , ARN Guía de Sistemas CRISPR-Cas/metabolismo , ARN Guía de Sistemas CRISPR-Cas/genética
2.
Front Immunol ; 15: 1445338, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39247192

RESUMEN

Background: Defective ribosomal products (DRiPs) are non-functional proteins rapidly degraded during or after translation being an essential source for MHC class I ligands. DRiPs are characterized to derive from a substantial subset of nascent gene products that degrade more rapidly than their corresponding native retiree pool. So far, mass spectrometry analysis revealed that a large number of HLA class I peptides derive from DRiPs. However, a specific viral DRiP on protein level was not described. In this study, we aimed to characterize and identify DRiPs derived from a viral protein. Methods: Using the nucleoprotein (NP) of the lymphocytic choriomeningitis virus (LCMV) which is conjugated N-terminally to ubiquitin, or the ubiquitin-like modifiers FAT10 or ISG15 the occurrence of DRiPs was studied. The formation and degradation of DRiPs was monitored by western blot with the help of a FLAG tag. Flow cytometry and cytotoxic T cells were used to study antigen presentation. Results: We identified several short lived DRiPs derived from LCMV-NP. Of note, these DRiPs could only be observed when the LCMV-NP was modified with ubiquitin or ubiquitin-like modifiers, but not in the wild type form. Using proteasome inhibitors, we could show that degradation of LCMV-NP derived DRiPs were proteasome dependent. Interestingly, the synthesis of DRiPs could be enhanced when cells were stressed with the help of FCS starvation. An enhanced NP118-126 presentation was observed when the LCMV-NP was modified with ubiquitin or ubiquitin-like modifiers, or under FCS starvation. Conclusion: Taken together, we visualize for the first time DRiPs derived from a viral protein. Furthermore, DRiPs formation, and therefore MHC-I presentation, is enhanced under cellular stress conditions. Our investigations on DRiPs in MHC class I antigen presentation open up new approaches for the development of vaccination strategies.


Asunto(s)
Presentación de Antígeno , Antígenos de Histocompatibilidad Clase I , Virus de la Coriomeningitis Linfocítica , Presentación de Antígeno/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Virus de la Coriomeningitis Linfocítica/inmunología , Animales , Humanos , Estrés Fisiológico/inmunología , Linfocitos T Citotóxicos/inmunología , Ratones , Ubiquitinas/metabolismo , Ubiquitinas/genética , Proteínas Ribosómicas/metabolismo , Proteínas Ribosómicas/inmunología , Proteolisis , Nucleoproteínas/inmunología , Nucleoproteínas/metabolismo
3.
Nat Commun ; 15(1): 8292, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39333100

RESUMEN

BRCA2 is essential for DNA repair by homologous recombination in mitosis and meiosis. It interacts with recombinases RAD51 and DMC1 to facilitate the formation of nucleoprotein filaments on resected DNA ends that catalyse recombination-mediated repair. BRCA2's BRC repeats bind and disrupt RAD51 and DMC1 filaments, whereas its PhePP motifs bind recombinases and stabilise their nucleoprotein filaments. However, the mechanism of filament stabilisation has hitherto remained unknown. Here, we report the crystal structure of a BRCA2-DMC1 complex, revealing how core interaction sites of PhePP motifs bind to recombinases. The interaction mode is conserved for RAD51 and DMC1, which selectively bind to BRCA2's two distinct PhePP motifs via subtly divergent binding pockets. PhePP motif sequences surrounding their core interaction sites protect nucleoprotein filaments from BRC-mediated disruption. Hence, we report the structural basis of how BRCA2's PhePP motifs stabilise RAD51 and DMC1 nucleoprotein filaments for their essential roles in mitotic and meiotic recombination.


Asunto(s)
Proteína BRCA2 , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Unión Proteica , Recombinasa Rad51 , Recombinasa Rad51/metabolismo , Recombinasa Rad51/química , Proteína BRCA2/metabolismo , Proteína BRCA2/química , Proteína BRCA2/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Humanos , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Nucleoproteínas/metabolismo , Nucleoproteínas/química , Nucleoproteínas/genética , Cristalografía por Rayos X , Meiosis , Sitios de Unión , Secuencias de Aminoácidos , Modelos Moleculares , Mitosis
4.
Med Microbiol Immunol ; 213(1): 20, 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39320473

RESUMEN

Crimean-Congo Hemorrhagic Fever Virus (CCHFV) is a globally significant vector-borne pathogen with no internationally-licensed preventative and therapeutic interventions. Hazara virus (HAZV), on the other hand, a related Orthonairovirus, has not been reported as a human pathogen. HAZV has been proposed as a surrogate model for studying CCHFV, bisosafety level 4 (BSL-4) agent. Previously, we investigated the humoral immune responses between NPs of these viruses and in this study, we extended the scrutiny to cellular immune responses elicited by NPs of CCHFV and HAZV. Here, mice were immunized with recombinant CCHFV NP and HAZV NP to evaluate the correlates of cell-mediated immunity (CMI). Delayed-type hypersensitivity (DTH) responses were assessed by challenging immunized mice with CCHFV-rNP or HAZV-rNP on the footpad and lymphocyte proliferation assays (LPAs) were performed by stimulating splenocytes in vitro with CCHFV-rNP or HAZV-rNP to compare cellular immune responses. In all test groups, strong DTH and LPA responses were detected against homologous and heterologous challenging antigens. To assess the cytokine response, an RT-qPCR -specific for cytokine mRNAs was utilized. Interestingly, CCHFV NP stimulated groups exhibited a significantly elevated mRNA level of interleukin 17 A (IL-17) compared to HAZV NP, indicating a notable difference in immune responses. This study presents comparison between CMI elicited by NPs of CCHFV and HAZV and contributes to the understanding of a highly pathogenic virus, particularly in the context of the declaration of CCHFV by World Health Organization's (WHO) as a major viral threat to the world.


Asunto(s)
Citocinas , Virus de la Fiebre Hemorrágica de Crimea-Congo , Inmunidad Celular , Animales , Virus de la Fiebre Hemorrágica de Crimea-Congo/inmunología , Citocinas/metabolismo , Ratones , Nucleoproteínas/inmunología , Ratones Endogámicos BALB C , Femenino , Hipersensibilidad Tardía/inmunología , Proliferación Celular , Bazo/inmunología
5.
Cell ; 187(20): 5587-5603.e19, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39293445

RESUMEN

Filoviruses, including the Ebola and Marburg viruses, cause hemorrhagic fevers with up to 90% lethality. The viral nucleocapsid is assembled by polymerization of the nucleoprotein (NP) along the viral genome, together with the viral proteins VP24 and VP35. We employed cryo-electron tomography of cells transfected with viral proteins and infected with model Ebola virus to illuminate assembly intermediates, as well as a 9 Å map of the complete intracellular assembly. This structure reveals a previously unresolved third and outer layer of NP complexed with VP35. The intrinsically disordered region, together with the C-terminal domain of this outer layer of NP, provides the constant width between intracellular nucleocapsid bundles and likely functions as a flexible tether to the viral matrix protein in the virion. A comparison of intracellular nucleocapsids with prior in-virion nucleocapsid structures reveals that the nucleocapsid further condenses vertically in the virion. The interfaces responsible for nucleocapsid assembly are highly conserved and offer targets for broadly effective antivirals.


Asunto(s)
Ebolavirus , Tomografía con Microscopio Electrónico , Nucleocápside , Ensamble de Virus , Ebolavirus/ultraestructura , Ebolavirus/química , Ebolavirus/metabolismo , Ebolavirus/fisiología , Nucleocápside/metabolismo , Nucleocápside/ultraestructura , Nucleocápside/química , Humanos , Microscopía por Crioelectrón/métodos , Proteínas de la Nucleocápside/química , Proteínas de la Nucleocápside/metabolismo , Proteínas de la Nucleocápside/ultraestructura , Nucleoproteínas/química , Nucleoproteínas/metabolismo , Nucleoproteínas/ultraestructura , Animales , Proteínas Virales/metabolismo , Proteínas Virales/química , Proteínas Virales/ultraestructura , Modelos Moleculares , Virión/ultraestructura , Virión/metabolismo , Fiebre Hemorrágica Ebola/virología , Chlorocebus aethiops
6.
J Cell Sci ; 137(19)2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39292070

RESUMEN

Lipid droplets (LDs) are organelles involved in lipid storage, maintenance of energy homeostasis, protein sequestration, signaling events and inter-organelle interactions. Recently, LDs have been shown to favor the replication of members from different viral families, such as the Flaviviridae and Coronaviridae. In this work, we show that LDs are essential organelles for members of the Arenaviridae family. A virus-driven reduction of LD number was observed in cultures infected with Junín mammarenavirus (JUNV), caused in part by action of the viral nucleoprotein. Notably, we identified a new pool of nucleoprotein and viral RNA that localizes in the vicinity of LDs, suggesting that LDs play a role during the viral replication cycle. Regarding the mechanism behind LD exhaustion, we found evidence that lipophagy is involved in LD degradation with the resulting fatty acids being substrates of fatty acid ß-oxidation, which fuels viral multiplication. This work highlights the importance of LDs during the replication cycle of JUNV, contributing to the knowledge of the metabolic changes these mammarenaviruses cause in their hosts.


Asunto(s)
Virus Junin , Gotas Lipídicas , Nucleoproteínas , Replicación Viral , Virus Junin/metabolismo , Gotas Lipídicas/metabolismo , Gotas Lipídicas/virología , Nucleoproteínas/metabolismo , Animales , Chlorocebus aethiops , Células Vero , ARN Viral/metabolismo , ARN Viral/genética , Humanos , Ácidos Grasos/metabolismo
7.
J Virol Methods ; 329: 115009, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39142521

RESUMEN

Bovine ephemeral fever virus (BEFV) is a member of the genus Ephemerovirus in the family Rhabdoviridae. It is an arthropod-borne virus transmitted by many species of midges and mosquitoes. It can cause severe economic consequences due to losses in milk production and the general condition of cattle and water buffalo. BEF occurs in some tropical, subtropical and warm temperate regions of Africa, Australia, the Middle East and Asia with seasonal outbreaks, but its possible spread to other areas (e.g. Europe) cannot be excluded. Therefore, using and developing rapid diagnostic methods with optimal performance is essential for identifying emerging pathogens and their control. In the present study, we developed two competitive serological ELISAs based on monoclonal antibodies (mAbs), designed by using BEFV inactivated antigen and the BEF recombinant nucleoprotein (N), respectively. A panel of 77 BEF-positive and 338 BEF-negative sera was used to evaluate the two tests. With a diagnostic sensitivity of 97.4 % using the inactivated virus and 98.7 % using the recombinant N, and a diagnostic specificity of 100 % using both antigens, our results suggest that these tests are suitable for the serological diagnosis of BEF.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Antivirales , Antígenos Virales , Ensayo de Inmunoadsorción Enzimática , Virus de la Fiebre Efímera Bovina , Fiebre Efímera , Sensibilidad y Especificidad , Animales , Virus de la Fiebre Efímera Bovina/inmunología , Virus de la Fiebre Efímera Bovina/aislamiento & purificación , Bovinos , Fiebre Efímera/diagnóstico , Fiebre Efímera/virología , Fiebre Efímera/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Monoclonales/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Ensayo de Inmunoadsorción Enzimática/veterinaria , Antígenos Virales/inmunología , Pruebas Serológicas/métodos , Nucleoproteínas/inmunología
8.
Nat Commun ; 15(1): 7602, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39217162

RESUMEN

Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic pathogen. Its RNA genome consists of two negative-sense segments (L and M) with one gene each, and one ambisense segment (S) with two opposing genes separated by the noncoding "intergenic region" (IGR). These vRNAs and the complementary cRNAs are encapsidated by nucleoprotein (N). Using iCLIP2 (individual-nucleotide resolution UV crosslinking and immunoprecipitation) to map all N-vRNA and N-cRNA interactions, we detect N coverage along the L and M segments. However, the S segment vRNA and cRNA each contain approximately 100 non-encapsidated nucleotides stretching from the IGR into the 5'-adjacent reading frame. These exposed regions are RNase-sensitive and predicted to form stem-loop structures with the mRNA transcription termination motif positioned near the top. Moreover, optimal S segment transcription and replication requires the entire exposed region rather than only the IGR. Thus, the RVFV S segment contains a central, non-encapsidated RNA region with a functional role.


Asunto(s)
ARN Viral , Virus de la Fiebre del Valle del Rift , Virus de la Fiebre del Valle del Rift/genética , ARN Viral/genética , Animales , ADN Intergénico/genética , Genoma Viral , Replicación Viral/genética , Fiebre del Valle del Rift/virología , Fiebre del Valle del Rift/transmisión , Conformación de Ácido Nucleico , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Humanos , Transcripción Genética
9.
Emerg Microbes Infect ; 13(1): 2387910, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39087696

RESUMEN

Nuclear export of the viral ribonucleoprotein (vRNP) is a critical step in the influenza A virus (IAV) life cycle and may be an effective target for the development of anti-IAV drugs. The host factor ras-related nuclear protein (RAN) is known to participate in the life cycle of several viruses, but its role in influenza virus replication remains unknown. In the present study, we aimed to determine the function of RAN in influenza virus replication using different cell lines and subtype strains. We found that RAN is essential for the nuclear export of vRNP, as it enhances the binding affinity of XPO1 toward the viral nuclear export protein NS2. Depletion of RAN constrained the vRNP complex in the nucleus and attenuated the replication of various subtypes of influenza virus. Using in silico compound screening, we identified that bepotastine could dissociate the RAN-XPO1-vRNP trimeric complex and exhibit potent antiviral activity against influenza virus both in vitro and in vivo. This study demonstrates the important role of RAN in IAV replication and suggests its potential use as an antiviral target.


Asunto(s)
Transporte Activo de Núcleo Celular , Antivirales , Proteína Exportina 1 , Virus de la Influenza A , Carioferinas , Replicación Viral , Proteína de Unión al GTP ran , Replicación Viral/efectos de los fármacos , Humanos , Proteína de Unión al GTP ran/metabolismo , Proteína de Unión al GTP ran/genética , Antivirales/farmacología , Animales , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/fisiología , Carioferinas/metabolismo , Carioferinas/antagonistas & inhibidores , Perros , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Células de Riñón Canino Madin Darby , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Ratones , Piperidinas/farmacología , Gripe Humana/virología , Células A549 , Nucleoproteínas/metabolismo , Nucleoproteínas/genética , Células HEK293 , Línea Celular , Núcleo Celular/metabolismo , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas/genética
10.
Structure ; 32(8): 1027-1028, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39121836

RESUMEN

The genome of segmented negative-sense single-stranded RNA viruses, such as influenza virus and bunyaviruses, is coated by viral nucleoproteins (NPs), forming a ribonucleoprotein (RNP). In this issue of Structure, Dick et al.1 expand our knowledge on the RNPs of these viruses by solving the structures of Thogoto virus NP and RNP.


Asunto(s)
Ribonucleoproteínas , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo , ARN Viral/química , ARN Viral/metabolismo , ARN Viral/genética , Thogotovirus/química , Thogotovirus/metabolismo , Virus ARN/genética , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Virales/genética , Modelos Moleculares , Nucleoproteínas/química , Nucleoproteínas/metabolismo
11.
Methods Mol Biol ; 2824: 281-318, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39039419

RESUMEN

Rift Valley fever virus (RVFV; genus Phlebovirus, family Phenuiviridae, order Bunyavirales) is a mosquito-borne zoonotic pathogen endemic in Africa. Its negative-stranded genomic RNA (vRNA) is divided into three segments termed L, M, and S. Both vRNAs and antigenomic cRNAs are encapsidated by viral nucleoprotein (N) to form nucleocapsids, which constitute the template for genome transcription and replication. Based on a number of electron microscopy and structural studies, the viral RNAs of negative-strand RNA viruses, including phleboviruses, are commonly considered to be entirely and uniformly covered by N protein. However, high resolution data supporting this notion was missing to date.Here, we describe a method how to globally map all N-RNA interactions of RVFV by using iCLIP (individual-nucleotide resolution UV cross-linking and immunoprecipitation). The protocol is based on covalent cross-linking of direct protein-RNA interactions by UV irradiation. Following sample lysis, a selective isolation of N in complex with its RNA targets is achieved by immunoprecipitation. Then, N-RNA complexes are separated by SDS-PAGE, and after membrane transfer, RNA is isolated and subjected to library preparation and high-throughput sequencing. We explain how the standard iCLIP protocol can be adapted to RVFV N-RNA interaction studies. The protocol describes mapping of all N interactions with the vRNAs and cRNAs derived either from RVFV particles or from infected cells.


Asunto(s)
Genoma Viral , Nucleoproteínas , ARN Viral , Virus de la Fiebre del Valle del Rift , Virus de la Fiebre del Valle del Rift/genética , ARN Viral/genética , ARN Viral/metabolismo , Nucleoproteínas/metabolismo , Nucleoproteínas/genética , Mapeo Nucleótido/métodos , Inmunoprecipitación/métodos , Humanos , Fiebre del Valle del Rift/virología , Fiebre del Valle del Rift/metabolismo , Animales
12.
J Virol ; 98(8): e0071124, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39082839

RESUMEN

Cytotoxic T lymphocytes (CTLs) mediate host defense against viral and intracellular bacterial infections and tumors. However, the magnitude of CTL response and their function needed to confer heterosubtypic immunity against influenza virus infection are unknown. We addressed the role of CD8+ T cells in the absence of any cross-reactive antibody responses to influenza viral proteins using an adenoviral vector expressing a 9mer amino acid sequence recognized by CD8+ T cells. Our results indicate that both CD8+ T cell frequency and function are crucial for heterosubtypic immunity. Low morbidity, lower viral lung titers, low to minimal lung pathology, and better survival upon heterosubtypic virus challenge correlated with the increased frequency of NP-specific CTLs. NP-CD8+ T cells induced by differential infection doses displayed distinct RNA transcriptome profiles and functional properties. CD8+ T cells induced by a high dose of influenza virus secreted significantly higher levels of IFN-γ and exhibited higher levels of cytotoxic function. The mice that received NP-CD8+ T cells from the high-dose virus recipients through adoptive transfer had lower viral titers following viral challenge than those induced by the low dose of virus, suggesting differential cellular programming by antigen dose. Enhanced NP-CD8+ T-cell functions induced by a higher dose of influenza virus strongly correlated with the increased expression of cellular and metabolic genes, indicating a shift to a more glycolytic metabolic phenotype. These findings have implications for developing effective T cell vaccines against infectious diseases and cancer. IMPORTANCE: Cytotoxic T lymphocytes (CTLs) are an important component of the adaptive immune system that clears virus-infected cells or tumor cells. Hence, developing next-generation vaccines that induce or recall CTL responses against cancer and infectious diseases is crucial. However, it is not clear if the frequency, function, or both are essential in conferring protection, as in the case of influenza. In this study, we demonstrate that both CTL frequency and function are crucial for providing heterosubtypic immunity to influenza by utilizing an Ad-viral vector expressing a CD8 epitope only to rule out the role of antibodies, single-cell RNA-seq analysis, as well as adoptive transfer experiments. Our findings have implications for developing T cell vaccines against infectious diseases and cancer.


Asunto(s)
Linfocitos T CD8-positivos , Infecciones por Orthomyxoviridae , Linfocitos T Citotóxicos , Animales , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Ratones , Linfocitos T CD8-positivos/inmunología , Linfocitos T Citotóxicos/inmunología , Ratones Endogámicos C57BL , Femenino , Traslado Adoptivo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Proteínas de la Nucleocápside/inmunología , Pulmón/inmunología , Pulmón/virología , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/genética , Nucleoproteínas/inmunología , Nucleoproteínas/genética , Proteínas del Núcleo Viral/inmunología , Proteínas del Núcleo Viral/genética
13.
J Biol Chem ; 300(8): 107456, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38866325

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a lipid-enveloped virus that acquires its lipid bilayer from the host cell it infects. SARS-CoV-2 can spread from cell to cell or from patient to patient by undergoing assembly and budding to form new virions. The assembly and budding of SARS-CoV-2 is mediated by several structural proteins known as envelope (E), membrane (M), nucleoprotein (N), and spike (S), which can form virus-like particles (VLPs) when co-expressed in mammalian cells. Assembly and budding of SARS-CoV-2 from the host ER-Golgi intermediate compartment is a critical step in the virus acquiring its lipid bilayer. To date, little information is available on how SARS-CoV-2 assembles and forms new viral particles from host membranes. In this study, we used several lipid binding assays and found the N protein can strongly associate with anionic lipids including phosphoinositides and phosphatidylserine. Moreover, we show lipid binding occurs in the N protein C-terminal domain, which is supported by extensive in silico analysis. We demonstrate anionic lipid binding occurs for both the free and the N oligomeric forms, suggesting N can associate with membranes in the nucleocapsid form. Based on these results, we present a lipid-dependent model based on in vitro, cellular, and in silico data for the recruitment of N to assembly sites in the lifecycle of SARS-CoV-2.


Asunto(s)
SARS-CoV-2 , SARS-CoV-2/metabolismo , Humanos , Proteínas de la Nucleocápside de Coronavirus/metabolismo , Proteínas de la Nucleocápside de Coronavirus/química , Proteínas de la Nucleocápside de Coronavirus/genética , COVID-19/metabolismo , COVID-19/virología , Lípidos de la Membrana/metabolismo , Ensamble de Virus , Nucleoproteínas/metabolismo , Nucleoproteínas/química , Fosfatidilserinas/metabolismo , Fosfatidilserinas/química , Aniones/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/química , Membrana Celular/metabolismo , Betacoronavirus/metabolismo
14.
Int J Mol Sci ; 25(12)2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38928101

RESUMEN

In our prior investigations, we elucidated the role of the tryptophan-to-tyrosine substitution at the 61st position in the nonstructural protein NSsW61Y in diminishing the interaction between nonstructural proteins (NSs) and nucleoprotein (NP), impeding viral replication. In this study, we focused on the involvement of NSs in replication via the modulation of autophagosomes. Initially, we examined the impact of NP expression levels, a marker for replication, upon the infection of HeLa cells with severe fever thrombocytopenia syndrome virus (SFTSV), with or without the inhibition of NP binding. Western blot analysis revealed a reduction in NP levels in NSsW61Y-expressing conditions. Furthermore, the expression levels of the canonical autophagosome markers p62 and LC3 decreased in HeLa cells expressing NSsW61Y, revealing the involvement of individual viral proteins on autophagy. Subsequent experiments confirmed that NSsW61Y perturbs autophagy flux, as evidenced by reduced levels of LC3B and p62 upon treatment with chloroquine, an inhibitor of autophagosome-lysosome fusion. LysoTracker staining demonstrated a decrease in lysosomes in cells expressing the NS mutant compared to those expressing wild-type NS. We further explored the mTOR-associated regulatory pathway, a key regulator affected by NS mutant expression. The observed inhibition of replication could be linked to conformational changes in the NSs, impairing their binding to NP and altering mTOR regulation, a crucial upstream signaling component in autophagy. These findings illuminate the intricate interplay between NSsW61Y and the suppression of host autophagy machinery, which is crucial for the generation of autophagosomes to facilitate viral replication.


Asunto(s)
Autofagosomas , Autofagia , Phlebovirus , Triptófano , Tirosina , Proteínas no Estructurales Virales , Replicación Viral , Humanos , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Replicación Viral/genética , Autofagosomas/metabolismo , Células HeLa , Phlebovirus/genética , Phlebovirus/fisiología , Phlebovirus/metabolismo , Autofagia/genética , Tirosina/metabolismo , Triptófano/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Mutación , Sustitución de Aminoácidos , Síndrome de Trombocitopenia Febril Grave/metabolismo , Síndrome de Trombocitopenia Febril Grave/virología , Síndrome de Trombocitopenia Febril Grave/genética , Lisosomas/metabolismo , Nucleoproteínas/metabolismo , Nucleoproteínas/genética
15.
Sci Rep ; 14(1): 14099, 2024 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890308

RESUMEN

We report the first cryoEM structure of the Hendra henipavirus nucleoprotein in complex with RNA, at 3.5 Å resolution, derived from single particle analysis of a double homotetradecameric RNA-bound N protein ring assembly exhibiting D14 symmetry. The structure of the HeV N protein adopts the common bi-lobed paramyxoviral N protein fold; the N-terminal and C-terminal globular domains are bisected by an RNA binding cleft containing six RNA nucleotides and are flanked by the N-terminal and C-terminal arms, respectively. In common with other paramyxoviral nucleocapsids, the lateral interface between adjacent Ni and Ni+1 protomers involves electrostatic and hydrophobic interactions mediated primarily through the N-terminal arm and globular domains with minor contribution from the C-terminal arm. However, the HeV N multimeric assembly uniquely identifies an additional protomer-protomer contact between the Ni+1 N-terminus and Ni-1 C-terminal arm linker. The model presented here broadens the understanding of RNA-bound paramyxoviral nucleocapsid architectures and provides a platform for further insight into the molecular biology of HeV, as well as the development of antiviral interventions.


Asunto(s)
Microscopía por Crioelectrón , Virus Hendra , Nucleocápside , Nucleoproteínas , Virus Hendra/química , Nucleoproteínas/química , Nucleoproteínas/ultraestructura , Nucleoproteínas/metabolismo , Nucleocápside/química , Nucleocápside/ultraestructura , Nucleocápside/metabolismo , Modelos Moleculares , ARN Viral/química , ARN Viral/metabolismo , ARN Viral/genética , Proteínas de la Nucleocápside/química , Proteínas de la Nucleocápside/ultraestructura , Proteínas de la Nucleocápside/metabolismo
16.
J Virol ; 98(7): e0020224, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38842318

RESUMEN

Nucleoprotein (N) is well known for its function in the encapsidation of the genomic RNAs of negative-strand RNA viruses, which leads to the formation of ribonucleoproteins that serve as templates for viral transcription and replication. However, the function of the N protein in other aspects during viral infection is far from clear. In this study, the N protein of snakehead vesiculovirus (SHVV), a kind of fish rhabdovirus, was proved to be ubiquitinated mainly via K63-linked ubiquitination. We identified nine host E3 ubiquitin ligases that interacted with SHVV N, among which seven E3 ubiquitin ligases facilitated ubiquitination of the N protein. Further investigation revealed that only two E3 ubiquitin ligases, Siah E3 ubiquitin protein ligase 2 (Siah2) and leucine-rich repeat and sterile alpha motif containing 1 (LRSAM1), mediated K63-linked ubiquitination of the N protein. SHVV infection upregulated the expression of Siah2 and LRSAM1, which maintained the stability of SHVV N. Besides, overexpression of Siah2 or LRSAM1 promoted SHVV replication, while knockdown of Siah2 or LRSAM1 inhibited SHVV replication. Deletion of the ligase domain of Siah2 or LRSAM1 did not affect their interactions with SHVV N but reduced the K63-linked ubiquitination of SHVV N and SHVV replication. In summary, Siah2 and LRSAM1 mediate K63-linked ubiquitination of SHVV N to facilitate SHVV replication, which provides novel insights into the role of the N proteins of negative-strand RNA viruses. IMPORTANCE: Ubiquitination of viral protein plays an important role in viral replication. However, the ubiquitination of the nucleoprotein (N) of negative-strand RNA viruses has rarely been investigated. This study aimed at investigating the ubiquitination of the N protein of a fish rhabdovirus SHVV (snakehead vesiculovirus), identifying the related host E3 ubiquitin ligases, and determining the role of SHVV N ubiquitination and host E3 ubiquitin ligases in viral replication. We found that SHVV N was ubiquitinated mainly via K63-linked ubiquitination, which was mediated by host E3 ubiquitin ligases Siah2 (Siah E3 ubiquitin protein ligase 2) and LRSAM1 (leucine-rich repeat and sterile alpha motif containing 1). The data suggested that Siah2 and LRSAM1 were hijacked by SHVV to ubiquitinate the N protein for viral replication, which exhibited novel anti-SHVV targets for drug design.


Asunto(s)
Nucleoproteínas , Ubiquitina-Proteína Ligasas , Ubiquitinación , Vesiculovirus , Replicación Viral , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Animales , Nucleoproteínas/metabolismo , Nucleoproteínas/genética , Vesiculovirus/fisiología , Vesiculovirus/metabolismo , Vesiculovirus/genética , Humanos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Células HEK293 , Proteínas Virales/metabolismo , Proteínas Virales/genética , Línea Celular , Infecciones por Rhabdoviridae/virología , Infecciones por Rhabdoviridae/metabolismo , Enfermedades de los Peces/virología , Enfermedades de los Peces/metabolismo
17.
EBioMedicine ; 104: 105153, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38805853

RESUMEN

BACKGROUND: The development of a universal influenza virus vaccine, to protect against both seasonal and pandemic influenza A viruses, is a long-standing public health goal. The conserved stalk domain of haemagglutinin (HA) is a promising vaccine target. However, the stalk is immunosubdominant. As such, innovative approaches are required to elicit robust immunity against this domain. In a previously reported observer-blind, randomised placebo-controlled phase I trial (NCT03300050), immunisation regimens using chimeric HA (cHA)-based immunogens formulated as inactivated influenza vaccines (IIV) -/+ AS03 adjuvant, or live attenuated influenza vaccines (LAIV), elicited durable HA stalk-specific antibodies with broad reactivity. In this study, we sought to determine if these vaccines could also boost T cell responses against HA stalk, and nucleoprotein (NP). METHODS: We measured interferon-γ (IFN-γ) responses by Enzyme-Linked ImmunoSpot (ELISpot) assay at baseline, seven days post-prime, pre-boost and seven days post-boost following heterologous prime:boost regimens of LAIV and/or adjuvanted/unadjuvanted IIV-cHA vaccines. FINDINGS: Our findings demonstrate that immunisation with adjuvanted cHA-based IIVs boost HA stalk-specific and NP-specific T cell responses in humans. To date, it has been unclear if HA stalk-specific T cells can be boosted in humans by HA-stalk focused universal vaccines. Therefore, our study will provide valuable insights for the design of future studies to determine the precise role of HA stalk-specific T cells in broad protection. INTERPRETATION: Considering that cHA-based vaccines also elicit stalk-specific antibodies, these data support the further clinical advancement of cHA-based universal influenza vaccine candidates. FUNDING: This study was funded in part by the Bill and Melinda Gates Foundation (BMGF).


Asunto(s)
Anticuerpos Antivirales , Glicoproteínas Hemaglutininas del Virus de la Influenza , Inmunidad Celular , Vacunas contra la Influenza , Gripe Humana , Humanos , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Gripe Humana/prevención & control , Gripe Humana/inmunología , Anticuerpos Antivirales/inmunología , Femenino , Adulto , Masculino , Linfocitos T/inmunología , Inmunización Secundaria , Interferón gamma/metabolismo , Nucleoproteínas/inmunología , Adulto Joven , Virus de la Influenza A/inmunología
18.
Microbiol Spectr ; 12(6): e0379623, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38712963

RESUMEN

Cyclic GMP-AMP synthase (cGAS) is an important DNA pattern recognition receptor that senses double-stranded DNA derived from invading pathogens or self DNA in cytoplasm, leading to an antiviral interferon response. A tick-borne Bunyavirus, severe fever with thrombocytopenia syndrome virus (SFTSV), is an RNA virus that causes a severe emerging viral hemorrhagic fever in Asia with a high case fatality rate of up to 30%. However, it is unclear whether cGAS interacts with SFTSV infection. In this study, we found that SFTSV infection upregulated cGAS RNA transcription and protein expression, indicating that cGAS is an important innate immune response against SFTSV infection. The mechanism of cGAS recognizing SFTSV is by cGAS interacting with misplaced mitochondrial DNA in the cytoplasm. Depletion of mitochondrial DNA significantly inhibited cGAS activation under SFTSV infection. Strikingly, we found that SFTSV nucleoprotein (N) induced cGAS degradation in a dose-dependent manner. Mechanically, N interacted with the 161-382 domain of cGAS and linked the cGAS to LC3. The cGAS-N-LC3 trimer was targeted to N-induced autophagy, and the cGAS was degraded in autolysosome. Taken together, our study discovered a novel antagonistic mechanism of RNA viruses, SFTSV is able to suppress the cGAS-dependent antiviral innate immune responses through N-hijacking cGAS into N-induced autophagy. Our results indicated that SFTSV N is an important virulence factor of SFTSV in mediating host antiviral immune responses. IMPORTANCE: Severe fever with thrombocytopenia syndrome virus (SFTSV) is a tick-borne RNA virus that is widespread in East and Southeast Asian countries with a high fatality rate of up to 30%. Up to now, many cytoplasmic pattern recognition receptors, such as RIG-I, MDA5, and SAFA, have been reported to recognize SFTSV genomic RNA and trigger interferon-dependent antiviral responses. However, current knowledge is not clear whether SFTSV can be recognized by DNA sensor cyclic GMP-AMP synthase (cGAS). Our study demonstrated that cGAS could recognize SFTSV infection via ectopic mitochondrial DNA, and the activated cGAS-stimulator of interferon genes signaling pathway could significantly inhibit SFTSV replication. Importantly, we further uncovered a novel mechanism of SFTSV to inhibit innate immune responses by the degradation of cGAS. cGAS was degraded in N-induced autophagy. Collectively, this study illustrated a novel virulence factor of SFTSV to suppress innate immune responses through autophagy-dependent cGAS degradation.


Asunto(s)
Inmunidad Innata , Nucleoproteínas , Nucleotidiltransferasas , Phlebovirus , Phlebovirus/genética , Phlebovirus/inmunología , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Humanos , Nucleoproteínas/metabolismo , Nucleoproteínas/genética , Nucleoproteínas/inmunología , Células HEK293 , Síndrome de Trombocitopenia Febril Grave/virología , Síndrome de Trombocitopenia Febril Grave/inmunología , Síndrome de Trombocitopenia Febril Grave/metabolismo , Autofagia , Animales , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Interferones/metabolismo , Interferones/inmunología , Interferones/genética , Proteínas Virales/metabolismo , Proteínas Virales/genética
19.
Structure ; 32(8): 1068-1078.e5, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38749445

RESUMEN

Orthomyxoviruses, such as influenza and thogotoviruses, are important human and animal pathogens. Their segmented viral RNA genomes are wrapped by viral nucleoproteins (NPs) into helical ribonucleoprotein complexes (RNPs). NP structures of several influenza viruses have been reported. However, there are still contradictory models of how orthomyxovirus RNPs are assembled. Here, we characterize the crystal structure of Thogoto virus (THOV) NP and found striking similarities to structures of influenza viral NPs, including a two-lobed domain architecture, a positively charged RNA-binding cleft, and a tail loop important for trimerization and viral transcription. A low-resolution cryo-electron tomography reconstruction of THOV RNPs elucidates a left-handed double helical assembly. By providing a model for RNP assembly of THOV, our study suggests conserved NP assembly and RNA encapsidation modes for thogoto- and influenza viruses.


Asunto(s)
Modelos Moleculares , ARN Viral , Ribonucleoproteínas , Thogotovirus , ARN Viral/metabolismo , ARN Viral/química , Thogotovirus/metabolismo , Thogotovirus/química , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas/química , Cristalografía por Rayos X , Microscopía por Crioelectrón , Nucleoproteínas/química , Nucleoproteínas/metabolismo , Unión Proteica , Sitios de Unión , Humanos , Ensamble de Virus
20.
J Virol ; 98(6): e0057824, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38767352

RESUMEN

The mammarenavirus Lassa virus (LASV) causes the life-threatening hemorrhagic fever disease, Lassa fever. The lack of licensed medical countermeasures against LASV underscores the urgent need for the development of novel LASV vaccines, which has been hampered by the requirement for a biosafety level 4 facility to handle live LASV. Here, we investigated the efficacy of mRNA-lipid nanoparticle (mRNA-LNP)-based vaccines expressing the LASV glycoprotein precursor (LASgpc) or nucleoprotein (LCMnp) of the prototypic mammarenavirus, lymphocytic choriomeningitis virus (LCMV), in mice. Two doses of LASgpc- or LCMnp-mRNA-LNP administered intravenously (i.v.) protected C57BL/6 mice from a lethal challenge with a recombinant (r) LCMV expressing a modified LASgpc (rLCMV/LASgpc2m) inoculated intracranially. Intramuscular (i.m.) immunization with two doses of LASgpc- or LCMnp-mRNA-LNP significantly reduced the viral load in C57BL/6 mice inoculated i.v. with rLCMV/LASgpc2m. High levels of viremia and lethality were observed in CBA mice inoculated i.v. with rLCMV/LASgpc2m, which were abrogated by i.m. immunization with two doses of LASgpc-mRNA-LNP. The protective efficacy of two i.m. doses of LCMnp-mRNA-LNP was confirmed in a lethal hemorrhagic disease model of FVB mice i.v. inoculated with wild-type rLCMV. In all conditions tested, negligible and high levels of LASgpc- and LCMnp-specific antibodies were detected in mRNA-LNP-immunized mice, respectively, but robust LASgpc- and LCMnp-specific CD8+ T cell responses were induced. Accordingly, plasma from LASgpc-mRNA-LNP-immunized mice did not exhibit neutralizing activity. Our findings and surrogate mouse models of LASV infection, which can be studied at a reduced biocontainment level, provide a critical foundation for the rapid development of mRNA-LNP-based LASV vaccines.IMPORTANCELassa virus (LASV) is a highly pathogenic mammarenavirus responsible for several hundred thousand infections annually in West African countries, causing a high number of lethal Lassa fever (LF) cases. Despite its significant impact on human health, clinically approved, safe, and effective medical countermeasures against LF are not available. The requirement of a biosafety level 4 facility to handle live LASV has been one of the main obstacles to the research and development of LASV countermeasures. Here, we report that two doses of mRNA-lipid nanoparticle-based vaccines expressing the LASV glycoprotein precursor (LASgpc) or nucleoprotein (LCMnp) of lymphocytic choriomeningitis virus (LCMV), a mammarenavirus genetically closely related to LASV, conferred protection to recombinant LCMV-based surrogate mouse models of lethal LASV infection. Notably, robust LASgpc- and LCMnp-specific CD8+ T cell responses were detected in mRNA-LNP-immunized mice, whereas no virus-neutralizing activity was observed.


Asunto(s)
Fiebre de Lassa , Virus Lassa , Virus de la Coriomeningitis Linfocítica , Nanopartículas , Vacunas Virales , Animales , Femenino , Ratones , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Glicoproteínas/inmunología , Glicoproteínas/genética , Fiebre de Lassa/prevención & control , Fiebre de Lassa/inmunología , Virus Lassa/inmunología , Virus Lassa/genética , Liposomas , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/genética , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Nucleoproteínas/inmunología , Nucleoproteínas/genética , ARN Mensajero/genética , ARN Mensajero/inmunología , Carga Viral , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA