Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
Methods Mol Biol ; 2808: 89-103, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743364

RESUMEN

The study of virus-host interactions is essential to achieve a comprehensive understanding of the viral replication process. The commonly used methods are yeast two-hybrid approach and transient expression of a single tagged viral protein in host cells followed by affinity purification of interacting cellular proteins and mass spectrometry analysis (AP-MS). However, by these approaches, virus-host protein-protein interactions are detected in the absence of a real infection, not always correctly compartmentalized, and for the yeast two-hybrid approach performed in a heterologous system. Thus, some of the detected protein-protein interactions may be artificial. Here we describe a new strategy based on recombinant viruses expressing tagged viral proteins to capture both direct and indirect protein partners during the infection (AP-MS in viral context). This way, virus-host protein-protein interacting co-complexes can be purified directly from infected cells for further characterization.


Asunto(s)
Interacciones Huésped-Patógeno , Virus del Sarampión , Genética Inversa , Proteínas Virales , Virus del Sarampión/genética , Humanos , Interacciones Huésped-Patógeno/genética , Genética Inversa/métodos , Proteínas Virales/metabolismo , Proteínas Virales/genética , Técnicas del Sistema de Dos Híbridos , Replicación Viral , Espectrometría de Masas , Mapeo de Interacción de Proteínas/métodos , Sarampión/virología , Sarampión/metabolismo , Animales , Unión Proteica
2.
Microbes Infect ; : 105321, 2024 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-38461968

RESUMEN

Rabies virus (RABV) is a lethal neurotropic virus that causes 60,000 human deaths every year globally. RABV infection is characterized by the suppression of the interferon (IFN)-mediated antiviral response. However, molecular mechanisms leading to RABV sensing by RIG-I-like receptors (RLR) that initiates IFN signaling currently remain elusive. Here, we showed that RABV RNAs are primarily recognized by the RIG-I RLR, resulting in an IFN response in the infected cells, but this response varied according to the type of RABV used. Pathogenic RABV strain RNAs, Tha, were poorly detected in the cytosol by RIG-I and therefore caused a weak antiviral response. However, we revealed a strong IFN activity triggered by the attenuated RABV vaccine strain RNAs, SAD, mediated by RIG-I. We characterized two major 5' copy-back defective interfering (5'cb DI) genomes generated during SAD replication. Furthermore, we identified an interaction between 5'cb DI genomes, and RIG-I correlated with a high stimulation of the type I IFN signaling. This study indicates that wild-type RABV RNAs poorly activate the RIG-I pathway, while the presence of 5'cb DIs in the live-attenuated vaccine strain serves as an intrinsic adjuvant that strengthens its efficiency by enhancing RIG-I detection thus strongly stimulates the IFN response.

3.
Cells ; 13(2)2024 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-38247817

RESUMEN

The membrane (M) glycoprotein of coronaviruses (CoVs) serves as the nidus for virion assembly. Using a yeast two-hybrid screen, we identified the interaction of the cytosolic tail of Murine Hepatitis Virus (MHV-CoV) M protein with Myosin Vb (MYO5B), specifically with the alternative splice variant of cellular MYO5B including exon D (MYO5B+D), which mediates interaction with Rab10. When co-expressed in human lung epithelial A549 and canine kidney epithelial MDCK cells, MYO5B+D co-localized with the MHV-CoV M protein, as well as with the M proteins from Porcine Epidemic Diarrhea Virus (PEDV-CoV), Middle East Respiratory Syndrome (MERS-CoV) and Severe Acute Respiratory Syndrome 2 (SARS-CoV-2). Co-expressed M proteins and MYO5B+D co-localized with endogenous Rab10 and Rab11a. We identified point mutations in MHV-CoV M that blocked the interaction with MYO5B+D in yeast 2-hybrid assays. One of these point mutations (E121K) was previously shown to block MHV-CoV virion assembly and its interaction with MYO5B+D. The E to K mutation at homologous positions in PEDV-CoV, MERS-CoV and SARS-CoV-2 M proteins also blocked colocalization with MYO5B+D. The knockdown of Rab10 blocked the co-localization of M proteins with MYO5B+D and was rescued by re-expression of CFP-Rab10. Our results suggest that CoV M proteins traffic through Rab10-containing systems, in association with MYO5B+D.


Asunto(s)
Proteínas M de Coronavirus , Animales , Perros , Humanos , Células de Riñón Canino Madin Darby/metabolismo , Células de Riñón Canino Madin Darby/virología , Coronavirus del Síndrome Respiratorio de Oriente Medio , Miosinas , Proteínas de Unión al GTP rab/genética , Saccharomyces cerevisiae , Porcinos , Proteínas de la Matriz Viral , SARS-CoV-2/metabolismo , Virus de la Hepatitis Murina/metabolismo , Células A549/metabolismo , Células A549/virología , Virus de la Diarrea Epidémica Porcina/metabolismo
4.
Nat Commun ; 14(1): 8440, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38114531

RESUMEN

Autophagy receptor NDP52 triggers bacterial autophagy against infection. However, the ability of NDP52 to protect against viral infection has not been established. We show that NDP52 binds to envelope proteins of hepatitis B virus (HBV) and triggers a degradation process that promotes HBV clearance. Inactivating NDP52 in hepatocytes results in decreased targeting of viral envelopes in the lysosome and increased levels of viral replication. NDP52 inhibits HBV at both viral entry and late replication stages. In contrast to NDP52-mediated bacterial autophagy, lysosomal degradation of HBV envelopes is independent of galectin 8 and ATG5. NDP52 forms complex with Rab9 and viral envelope proteins and links HBV to Rab9-dependent lysosomal degradation pathway. These findings reveal that NDP52 acts as a sensor for HBV infection, which mediates a unique antiviral response to eliminate the virus. This work also suggests direct roles for autophagy receptors in other lysosomal degradation pathways than canonical autophagy.


Asunto(s)
Virus de la Hepatitis B , Hepatitis B , Humanos , Virus de la Hepatitis B/fisiología , Hepatocitos/metabolismo , Autofagia/fisiología , Lisosomas/metabolismo , Antivirales/farmacología , Antivirales/uso terapéutico , Antivirales/metabolismo , Replicación Viral/fisiología
5.
Int J Mol Sci ; 24(19)2023 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-37834238

RESUMEN

Infection with Ebola virus (EBOV) is responsible for hemorrhagic fever in humans with a high mortality rate. Combined efforts of prevention and therapeutic intervention are required to tackle highly variable RNA viruses, whose infections often lead to outbreaks. Here, we have screened the 2P2I3D chemical library using a nanoluciferase-based protein complementation assay (NPCA) and isolated two compounds that disrupt the interaction of the EBOV protein fragment VP35IID with the N-terminus of the dsRNA-binding proteins PKR and PACT, involved in IFN response and/or intrinsic immunity, respectively. The two compounds inhibited EBOV infection in cell culture as well as infection by measles virus (MV) independently of IFN induction. Consequently, we propose that the compounds are antiviral by restoring intrinsic immunity driven by PACT. Given that PACT is highly conserved across mammals, our data support further testing of the compounds in other species, as well as against other negative-sense RNA viruses.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Humanos , Animales , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/metabolismo , Ebolavirus/fisiología , Antivirales/farmacología , Antivirales/uso terapéutico , Mamíferos
6.
J Infect Dis ; 228(12): 1675-1679, 2023 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-37562051

RESUMEN

The use of soluble recombinant angiotensin-converting enzyme 2 (rACE2) as a decoy capable of blocking SARS-CoV-2 entry into cells has been envisaged as a therapeutic strategy to reduce viral loads in patients with severe COVID-19. We engineered a novel form of rACE2, fused to the Epstein-Barr virus antigen P18F3 (rACE2-P18F3), to reorient a preexisting humoral response toward Epstein-Barr virus against SARS-CoV-2 particles. Recombinant ACE2-P18F3 was able to bind to the SARS-CoV-2 spike protein, neutralize viral entry into cells, and promote the phagocytosis of spheres coated with different spike variants by monocytic cells. The results position rACE2-P18F3 as a promising therapeutic candidate to universally block coronavirus cell entry and clear viral particles.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Anticuerpos Antivirales , COVID-19 , Herpesvirus Humano 4 , Glicoproteína de la Espiga del Coronavirus , Humanos , Enzima Convertidora de Angiotensina 2/metabolismo , Anticuerpos Antivirales/inmunología , Herpesvirus Humano 4/inmunología , Peptidil-Dipeptidasa A/genética , Unión Proteica , Proteínas Recombinantes/metabolismo , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/inmunología
7.
Cancer Immunol Immunother ; 72(10): 3309-3322, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37466668

RESUMEN

Antitumor virotherapy stimulates the antitumor immune response during tumor cell lysis induced by oncolytic viruses (OVs). OV can be modified to express additional transgenes that enhance their therapeutic potential. In this study, we armed the spontaneously oncolytic Schwarz strain of measles viruses (MVs) with the gene encoding the cancer/testis antigen NY-ESO-1 to obtain MVny. We compared MV and MVny oncolytic activity and ability to induce NY-ESO-1 expression in six human melanoma cell lines. After MVny infection, we measured the capacity of melanoma cells to present NY-ESO-1 peptides to CD4 + and CD8 + T cell clones specific for this antigen. We assessed the ability of MVny to induce NY-ESO-1 expression and presentation in monocyte-derived dendritic cells (DCs). Our results show that MVny and MV oncolytic activity are similar with a faster cell lysis induced by MVny. We also observed that melanoma cell lines and DC expressed the NY-ESO-1 protein after MVny infection. In addition, MVny-infected melanoma cells and DCs were able to stimulate NY-ESO-1-specific CD4 + and CD8 + T cells. Finally, MVny was able to induce DC maturation. Altogether, these results show that MVny could be an interesting candidate to stimulate NY-ESO-1-specific T cells in melanoma patients with NY-ESO-1-expressing tumor cells.


Asunto(s)
Sarampión , Melanoma , Virus Oncolíticos , Masculino , Humanos , Virus Oncolíticos/genética , Proteínas de la Membrana , Virus del Sarampión/genética , Melanoma/metabolismo , Linfocitos T CD8-positivos , Antígenos de Neoplasias , Anticuerpos/metabolismo , Células Dendríticas , Sarampión/metabolismo
8.
Nat Commun ; 14(1): 1352, 2023 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-36906645

RESUMEN

Lassa fever hits West African countries annually in the absence of licensed vaccine to limit the burden of this viral hemorrhagic fever. We previously developed MeV-NP, a single-shot vaccine protecting cynomolgus monkeys against divergent strains one month or more than a year before Lassa virus infection. Given the limited dissemination area during outbreaks and the risk of nosocomial transmission, a vaccine inducing rapid protection could be useful to protect exposed people during outbreaks in the absence of preventive vaccination. Here, we test whether the time to protection can be reduced after immunization by challenging measles virus pre-immune male cynomolgus monkeys sixteen or eight days after a single shot of MeV-NP. None of the immunized monkeys develop disease and they rapidly control viral replication. Animals immunized eight days before the challenge are the best controllers, producing a strong CD8 T-cell response against the viral glycoprotein. A group of animals was also vaccinated one hour after the challenge, but was not protected and succumbed to the disease as the control animals. This study demonstrates that MeV-NP can induce a rapid protective immune response against Lassa fever in the presence of MeV pre-existing immunity but can likely not be used as therapeutic vaccine.


Asunto(s)
Fiebre de Lassa , Fiebre de Lassa/inmunología , Fiebre de Lassa/prevención & control , Virus Lassa/inmunología , Masculino , Animales , Macaca fascicularis , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Nucleoproteínas/inmunología , Inmunidad Humoral , Replicación Viral , Linfocitos T/inmunología , Células Asesinas Naturales/inmunología , Transcriptoma
9.
PLoS Pathog ; 19(2): e1011170, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36802406

RESUMEN

Viruses have evolved countless mechanisms to subvert and impair the host innate immune response. Measles virus (MeV), an enveloped, non-segmented, negative-strand RNA virus, alters the interferon response through different mechanisms, yet no viral protein has been described as directly targeting mitochondria. Among the crucial mitochondrial enzymes, 5'-aminolevulinate synthase (ALAS) is an enzyme that catalyzes the first step in heme biosynthesis, generating 5'-aminolevulinate from glycine and succinyl-CoA. In this work, we demonstrate that MeV impairs the mitochondrial network through the V protein, which antagonizes the mitochondrial enzyme ALAS1 and sequesters it to the cytosol. This re-localization of ALAS1 leads to a decrease in mitochondrial volume and impairment of its metabolic potential, a phenomenon not observed in MeV deficient for the V gene. This perturbation of the mitochondrial dynamics demonstrated both in culture and in infected IFNAR-/- hCD46 transgenic mice, causes the release of mitochondrial double-stranded DNA (mtDNA) in the cytosol. By performing subcellular fractionation post infection, we demonstrate that the most significant source of DNA in the cytosol is of mitochondrial origin. Released mtDNA is then recognized and transcribed by the DNA-dependent RNA polymerase III. The resulting double-stranded RNA intermediates will be captured by RIG-I, ultimately initiating type I interferon production. Deep sequencing analysis of cytosolic mtDNA editing divulged an APOBEC3A signature, primarily analyzed in the 5'TpCpG context. Finally, in a negative feedback loop, APOBEC3A an interferon inducible enzyme will orchestrate the catabolism of mitochondrial DNA, decrease cellular inflammation, and dampen the innate immune response.


Asunto(s)
Interferones , Mitocondrias , Ratones , Animales , Mitocondrias/metabolismo , Virus del Sarampión , 5-Aminolevulinato Sintetasa/genética , 5-Aminolevulinato Sintetasa/metabolismo , ADN Mitocondrial
10.
Vaccines (Basel) ; 10(9)2022 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-36146633

RESUMEN

The recent surge of COVID-19 related to the Omicron variant emergence has thrown a harsh light upon epidemic control in the near future. This should lead the scientific and medical community to question the long-term vaccine strategy for SARS-CoV-2 control. We provide here a critical point of view regarding the virological evolution, epidemiological aspects, and immunological drivers for COVID-19 control, including a vaccination strategy. Overall, we need more innovations in vaccine development to reduce the COVID-19 burden long term. The most adequate answer might be better cooperation between universities, biotech and pharmaceutical companies.

11.
J Mol Biol ; 434(19): 167763, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-35907573

RESUMEN

Human RSV is the leading cause of infantile bronchiolitis in the world and one of the major causes of childhood deaths in resource-poor settings. It is a major unmet target for vaccines and anti-viral drugs. Respiratory syncytial virus has evolved a unique strategy to evade host immune response by coding for two non-structural proteins NS1 and NS2. Recently it was shown that in infected cells, nuclear NS1 could be involved in transcription regulation of host genes linked to innate immune response, via interactions with chromatin and the Mediator complex. Here we identified the MED25 Mediator subunit as an NS1 interactor in a yeast two-hybrid screen. We demonstrate that NS1 directly interacts with MED25 in vitro and in cellula, and that this interaction involves the MED25 transactivator binding ACID domain on the one hand, and the C-terminal α3 helix of NS1, with an additional contribution of the globular domain of NS1, on the other hand. By NMR we show that the NS1 α3 sequence primarily binds to the MED25 ACID H2 face, similarly to the α-helical transactivation domains (TADs) of transcription regulators such as Herpex simplex VP16 and ATF6α, a master regulator of ER stress response activated upon viral infection. Moreover, we found out that the NS1 could compete with ATF6α TAD for binding to MED25. These findings point to a mechanism of NS1 interfering with innate immune response by impairing recruitment by cellular TADs of the Mediator via MED25 and hence transcription of specific genes by RNA polymerase II.


Asunto(s)
Complejo Mediador , Virus Sincitial Respiratorio Humano , Transactivadores , Proteínas no Estructurales Virales , Cromatina/química , Humanos , Complejo Mediador/química , Unión Proteica , Dominios Proteicos , ARN Polimerasa II/metabolismo , Virus Sincitial Respiratorio Humano/genética , Transactivadores/química , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética
12.
iScience ; 25(7): 104599, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35789859

RESUMEN

Pattern recognition receptors (PRRs) protect against microbial invasion by detecting specific molecular patterns found in pathogens and initiating an immune response. Although microbial-derived PRR ligands have been extensively characterized, the contribution and relevance of endogenous ligands to PRR activation remains overlooked. Here, we characterize the landscape of endogenous ligands that engage RIG-I-like receptors (RLRs) upon infection by different RNA viruses. In each infection, several RNAs transcribed by RNA polymerase III (Pol3) specifically engaged RLRs, particularly the family of Y RNAs. Sensing of Y RNAs was dependent on their mimicking of viral secondary structure and their 5'-triphosphate extremity. Further, we found that HIV-1 triggered a VPR-dependent downregulation of RNA triphosphatase DUSP11 in vitro and in vivo, inducing a transcriptome-wide change of cellular RNA 5'-triphosphorylation that licenses Y RNA immunogenicity. Overall, our work uncovers the contribution of endogenous RNAs to antiviral immunity and demonstrates the importance of this pathway in HIV-1 infection.

13.
RNA ; 28(3): 277-289, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34937774

RESUMEN

Coronavirus RNA-dependent RNA polymerases produce subgenomic RNAs (sgRNAs) that encode viral structural and accessory proteins. User-friendly bioinformatic tools to detect and quantify sgRNA production are urgently needed to study the growing number of next-generation sequencing (NGS) data of SARS-CoV-2. We introduced sgDI-tector to identify and quantify sgRNA in SARS-CoV-2 NGS data. sgDI-tector allowed detection of sgRNA without initial knowledge of the transcription-regulatory sequences. We produced NGS data and successfully detected the nested set of sgRNAs with the ranking M > ORF3a > N>ORF6 > ORF7a > ORF8 > S > E>ORF7b. We also compared the level of sgRNA production with other types of viral RNA products such as defective interfering viral genomes.


Asunto(s)
Biología Computacional/métodos , Genoma Viral , ARN Viral/genética , SARS-CoV-2/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Sistemas de Lectura Abierta
14.
J Mol Biol ; 434(6): 167277, 2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-34599939

RESUMEN

Establishment of the interferon (IFN)-mediated antiviral state provides a crucial initial line of defense against viral infection. Numerous genes that contribute to this antiviral state remain to be identified. Using a loss-of-function strategy, we screened an original library of 1156 siRNAs targeting 386 individual curated human genes in stimulated microglial cells infected with Zika virus (ZIKV), an emerging RNA virus that belongs to the flavivirus genus. The screen recovered twenty-one potential host proteins that modulate ZIKV replication in an IFN-dependent manner, including the previously known IFITM3 and LY6E. Further characterization contributed to delineate the spectrum of action of these genes towards other pathogenic RNA viruses, including Hepatitis C virus and SARS-CoV-2. Our data revealed that APOL3 acts as a proviral factor for ZIKV and several other related and unrelated RNA viruses. In addition, we showed that MTA2, a chromatin remodeling factor, possesses potent flavivirus-specific antiviral functions induced by IFN. Our work identified previously unrecognized genes that modulate the replication of RNA viruses in an IFN-dependent manner, opening new perspectives to target weakness points in the life cycle of these viruses.


Asunto(s)
Flavivirus , Interferones , Replicación Viral , Apolipoproteínas L/genética , Apolipoproteínas L/metabolismo , Flavivirus/fisiología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Interferones/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , SARS-CoV-2/fisiología , Virus Zika/fisiología
15.
Med Sci (Paris) ; 38(12): 1052-1060, 2022 Dec.
Artículo en Francés | MEDLINE | ID: mdl-36692265

RESUMEN

Viruses have been used as tools to prevent viral infections themselves for more than two centuries with impressive success. After the empirical discoveries of the first vaccines, today the development of genetic engineering, molecular virology, reverse genetics, the manipulation of viral genomes, their high-throughput sequencing and their chemical synthesis, the mastery of cell culture and purification methods have greatly benefited the development of viral vaccines. Since smallpox and rabies, the history of vaccinology has followed in the footsteps of the history of virology. New mRNA or viral vector vaccines have emerged in recent years. They were developed and distributed to the population in record time in the face of the Covid pandemic. Viruses in the service of health have a bright future ahead of them, whether to prevent other pandemics, to treat cancer, or to finally control HIV and malaria.


Title: Les virus au service de la santé : la vaccination. Abstract: Depuis plus de deux siècles, les virus sont utilisés, avec un succès impressionnant, comme outils de prévention des infections virales. Depuis la variole et la rage, l'histoire de la vaccinologie a suivi les pas de l'histoire de la virologie. Après les découvertes empiriques des premiers vaccins, le développement du génie génétique, de la virologie moléculaire, de la génétique inverse, la manipulation des génomes viraux, leur séquençage à haut débit et leur synthèse chimique, la maîtrise de la culture cellulaire et des méthodes de purification, ont considérablement contribué au développement de nouveaux vaccins viraux. Des vaccins à ARN messager ou à vecteur viral ont ainsi vu le jour ces dernières années et, face à la pandémie de Covid-19, ont été développés et distribués à la population en un temps record. Les virus au service de la santé ont un bel avenir devant eux, que cela soit pour prévenir d'autres pandémies, pour traiter le cancer, ou contrôler, enfin, le VIH ou le Plasmodium, l'agent du paludisme.


Asunto(s)
COVID-19 , Vacunas Virales , Virosis , Virus , Humanos , COVID-19/prevención & control , Vacunación/historia , Virosis/prevención & control
16.
Nat Commun ; 12(1): 6277, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34725327

RESUMEN

Several COVID-19 vaccines have now been deployed to tackle the SARS-CoV-2 pandemic, most of them based on messenger RNA or adenovirus vectors.The duration of protection afforded by these vaccines is unknown, as well as their capacity to protect from emerging new variants. To provide sufficient coverage for the world population, additional strategies need to be tested. The live pediatric measles vaccine (MV) is an attractive approach, given its extensive safety and efficacy history, along with its established large-scale manufacturing capacity. We develop an MV-based SARS-CoV-2 vaccine expressing the prefusion-stabilized, membrane-anchored full-length S antigen, which proves to be efficient at eliciting strong Th1-dominant T-cell responses and high neutralizing antibody titers. In both mouse and golden Syrian hamster models, these responses protect the animals from intranasal infectious challenge. Additionally, the elicited antibodies efficiently neutralize in vitro the three currently circulating variants of SARS-CoV-2.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Vectores Genéticos , Inmunidad , Adenoviridae , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/prevención & control , Vacunas contra la COVID-19/administración & dosificación , Cricetinae , Citocinas , Femenino , Inmunización , Inmunización Secundaria , Masculino , Vacuna Antisarampión/inmunología , Mesocricetus , Ratones , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología
17.
NPJ Vaccines ; 6(1): 123, 2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34686669

RESUMEN

Replicative vectors derived from live-attenuated measles virus (MV) carrying additional non-measles vaccine antigens have long demonstrated safety and immunogenicity in humans despite pre-existing immunity to measles. Here, we report the vaccination of cynomolgus macaques with MV replicative vectors expressing simian-human immunodeficiency virus Gag, Env, and Nef antigens (MV-SHIV Wt) either wild type or mutated in the immunosuppressive (IS) domains of Nef and Env antigens (MV-SHIV Mt). We found that the inactivation of Nef and Env IS domains by targeted mutations led to the induction of significantly enhanced post-prime cellular immune responses. After repeated challenges with low doses of SHIV-SF162p3, vaccinees were protected against high viremia, resulting in a 2-Log reduction in peak viremia, accelerated viral clearance, and a decrease -even complete protection for nearly half of the monkeys- in reservoir cell infection. This study demonstrates the potential of a replicative viral vector derived from the safe and widely used measles vaccine in the development of a future human vaccine against HIV-1.

18.
J Virol ; 95(22): e0091221, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34431698

RESUMEN

Respiratory syncytial virus (RSV) is the main cause of acute respiratory infections in young children and also has a major impact on the elderly and immunocompromised people. In the absence of a vaccine or efficient treatment, a better understanding of RSV interactions with the host antiviral response during infection is needed. Previous studies revealed that cytoplasmic inclusion bodies (IBs), where viral replication and transcription occur, could play a major role in the control of innate immunity during infection by recruiting cellular proteins involved in the host antiviral response. We recently showed that the morphogenesis of IBs relies on a liquid-liquid-phase separation mechanism depending on the interaction between viral nucleoprotein (N) and phosphoprotein (P). These scaffold proteins are expected to play a central role in the recruitment of cellular proteins to IBs. Here, we performed a yeast two-hybrid screen using RSV N protein as bait and identified the cellular protein TAX1BP1 as a potential partner of this viral protein. This interaction was validated by pulldown and immunoprecipitation assays. We showed that TAX1BP1 suppression has only a limited impact on RSV infection in cell cultures. However, RSV replication is decreased in TAX1BP1-deficient (TAX1BP1 knockout [TAX1BP1KO]) mice, whereas the production of inflammatory and antiviral cytokines is enhanced. In vitro infection of wild-type or TAX1BP1KO alveolar macrophages confirmed that the innate immune response to RSV infection is enhanced in the absence of TAX1BP1. Altogether, our results suggest that RSV could hijack TAX1BP1 to restrain the host immune response during infection. IMPORTANCE Respiratory syncytial virus (RSV), which is the leading cause of lower respiratory tract illness in infants, remains a medical problem in the absence of a vaccine or efficient treatment. This virus is also recognized as a main pathogen in the elderly and immunocompromised people, and the occurrence of coinfections (with other respiratory viruses and bacteria) amplifies the risks of developing respiratory distress. In this context, a better understanding of the pathogenesis associated with viral respiratory infections, which depends on both viral replication and the host immune response, is needed. The present study reveals that the cellular protein TAX1BP1, which interacts with the RSV nucleoprotein N, participates in the control of the innate immune response during RSV infection, suggesting that the N-TAX1BP1 interaction represents a new target for the development of antivirals.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteínas de Neoplasias/inmunología , Proteínas de la Nucleocápside/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Animales , Línea Celular , Cricetinae , Humanos , Inmunidad Innata , Ratones , Ratones Noqueados , Replicación Viral
19.
Sci Transl Med ; 13(597)2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34108251

RESUMEN

A safe and protective Lassa virus vaccine is crucially needed in Western Africa to stem the recurrent outbreaks of Lassa virus infections in Nigeria and the emergence of Lassa virus in previously unaffected countries, such as Benin and Togo. Major challenges in developing a Lassa virus vaccine include the high diversity of circulating strains and their reemergence from 1 year to another. To address each of these challenges, we immunized cynomolgus monkeys with a measles virus vector expressing the Lassa virus glycoprotein and nucleoprotein of the prototypic Lassa virus strain Josiah (MeV-NP). To evaluate vaccine efficacy against heterologous strains of Lassa virus, we challenged the monkeys a month later with heterologous strains from lineage II or lineage VII, finding that the vaccine was protective against these strains. A second cohort of monkeys was challenged 1 year later with the homologous Josiah strain, finding that a single dose of MeV-NP was sufficient to protect all vaccinated monkeys. These studies demonstrate that MeV-NP can generate both long-lasting immune responses and responses that are able to protect against diverse strains of Lassa virus.


Asunto(s)
Fiebre de Lassa , Vacunas Virales/inmunología , África Occidental , Animales , Fiebre de Lassa/prevención & control , Virus Lassa , Macaca fascicularis , Nucleoproteínas
20.
Mol Ecol Resour ; 21(5): 1620-1640, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33586292

RESUMEN

To salvage marine ecosystems from fishery overexploitation, sustainable and efficient aquaculture must be emphasized. The knowledge obtained from available genome sequence of marine organisms has accelerated marine aquaculture in many cases. The black tiger shrimp (Penaeus monodon) is one of the most prominent cultured penaeid shrimps (Crustacean) with an average annual global production of half a million tons in the last decade. However, its currently available genome assemblies lack the contiguity and completeness required for accurate genome annotation due to the highly repetitive nature of the genome and technical difficulty in extracting high-quality, high-molecular weight DNA. Here, we report the first chromosome-level whole-genome assembly of P. monodon. The combination of long-read Pacific Biosciences (PacBio) and long-range Chicago and Hi-C technologies enabled a successful assembly of this first high-quality genome sequence. The final assembly covered 2.39 Gb (92.3% of the estimated genome size) and contained 44 pseudomolecules, corresponding to the haploid chromosome number. Repetitive elements occupied a substantial portion of the assembly (62.5%), the highest of the figures reported among crustacean species. The availability of this high-quality genome assembly enabled the identification of genes associated with rapid growth in the black tiger shrimp through the comparison of hepatopancreas transcriptome of slow-growing and fast-growing shrimps. The results highlighted several growth-associated genes. Our high-quality genome assembly provides an invaluable resource for genetic improvement and breeding penaeid shrimp in aquaculture. The availability of P. monodon genome enables analyses of ecological impact, environment adaptation and evolution, as well as the role of the genome to protect the ecological resources by promoting sustainable shrimp farming.


Asunto(s)
Genoma , Penaeidae , Animales , Acuicultura , Cromosomas , Penaeidae/genética , Penaeidae/crecimiento & desarrollo , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...