Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
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.
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
4.
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.

5.
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
6.
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
7.
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.

8.
Cell Host Microbe ; 26(6): 823-835.e11, 2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31761719

RESUMEN

RNA-binding proteins (RBPs) perform key cellular activities by controlling the function of bound RNAs. The widely held assumption that RBPs are strictly intracellular has been challenged by the discovery of secreted RBPs. However, extracellular RBPs have been described in eukaryotes, while secreted bacterial RBPs have not been reported. Here, we show that the bacterial pathogen Listeria monocytogenes secretes a small RBP that we named Zea. We show that Zea binds a subset of L. monocytogenes RNAs, causing their accumulation in the extracellular medium. Furthermore, during L. monocytogenes infection, Zea binds RIG-I, the non-self-RNA innate immunity sensor, potentiating interferon-ß production. Mouse infection studies reveal that Zea affects L. monocytogenes virulence. Together, our results unveil that bacterial RNAs can be present extracellularly in association with RBPs, acting as "social RNAs" to trigger a host response during infection.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Listeria monocytogenes/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Proteína 58 DEAD Box/inmunología , Células HEK293 , Interacciones Microbiota-Huesped , Humanos , Inmunidad Innata , Interferón beta/metabolismo , Listeria monocytogenes/inmunología , Listeria monocytogenes/patogenicidad , Ratones , ARN Bacteriano/metabolismo , Transducción de Señal/inmunología , Virulencia/inmunología
9.
Sci Signal ; 12(601)2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31575732

RESUMEN

The retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) RIG-I, MDA5, and LGP2 stimulate inflammatory and antiviral responses by sensing nonself RNA molecules produced during viral replication. Here, we investigated how LGP2 regulates the RIG-I- and MDA5-dependent induction of type I interferon (IFN) signaling and showed that LGP2 interacted with different components of the RNA-silencing machinery. We identified a direct protein-protein interaction between LGP2 and the IFN-inducible, double-stranded RNA binding protein PACT. The LGP2-PACT interaction was mediated by the regulatory C-terminal domain of LGP2 and was necessary for inhibiting RIG-I-dependent responses and for amplifying MDA5-dependent responses. We described a point mutation within LGP2 that disrupted the LGP2-PACT interaction and led to the loss of LGP2-mediated regulation of RIG-I and MDA5 signaling. These results suggest a model in which the LGP2-PACT interaction regulates the inflammatory responses mediated by RIG-I and MDA5 and enables the cellular RNA-silencing machinery to coordinate with the innate immune response.


Asunto(s)
Antivirales/metabolismo , Proteína 58 DEAD Box/metabolismo , Helicasa Inducida por Interferón IFIH1/metabolismo , ARN Helicasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Chlorocebus aethiops , Proteína 58 DEAD Box/genética , Enterovirus Humano B/genética , Enterovirus Humano B/fisiología , Células HEK293 , Células HeLa , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Mengovirus/genética , Mengovirus/fisiología , Unión Proteica , ARN Helicasas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Receptores Inmunológicos , Transducción de Señal/genética , Células Vero
10.
Cancer Immunol Immunother ; 68(4): 533-544, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30656384

RESUMEN

Cancer immunotherapy is seeing an increasing focus on vaccination with tumor-associated antigens (TAAs). Human telomerase (hTERT) is a TAA expressed by most tumors to overcome telomere shortening. Tolerance to hTERT can be easily broken both naturally and experimentally and hTERT DNA vaccine candidates have been introduced in clinical trials. DNA prime/boost strategies have been widely developed to immunize efficiently against infectious diseases. We explored the use of a recombinant measles virus (MV) hTERT vector to boost DNA priming as recombinant live attenuated measles virus has an impressive safety and efficacy record. Here, we show that a MV-TERT vector can rapidly and strongly boost DNA hTERT priming in MV susceptible IFNAR/CD46 mouse models. The cellular immune responses were Th1 polarized. No humoral responses were elicited. The 4 kb hTERT transgene did not impact MV replication or induction of cell-mediated responses. These findings validate the MV-TERT vector to boost cell-mediated responses following DNA priming in humans.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Epítopos de Linfocito T/inmunología , Vectores Genéticos , Inmunidad Celular , Virus del Sarampión , Linfocitos T/inmunología , Telomerasa/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/genética , Línea Celular , Chlorocebus aethiops , Citocinas/metabolismo , Citotoxicidad Inmunológica , Vectores Genéticos/genética , Humanos , Inmunización , Inmunización Secundaria , Virus del Sarampión/genética , Ratones , Ratones Transgénicos , Telomerasa/genética , Vacunas de ADN , Células Vero
11.
Cell Rep ; 24(2): 320-328, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29996094

RESUMEN

The flavivirus genus comprises major human pathogens, such as Dengue (DENV) and Zika (ZIKV) viruses. RIG-I and MDA5 are key cytoplasmic pathogen recognition receptors that are implicated in detecting viral RNAs. Here, we show that RNAs that co-purified with RIG-I during DENV infection are immuno-stimulatory, whereas RNAs bound to MDA5 are not. An affinity purification method combined with next-generation sequencing (NGS) revealed that the 5' region of the DENV genome is recognized by RIG-I. No DENV RNA was bound to MDA5. In vitro production of fragments of the DENV genome confirmed the NGS data and revealed that the 5' end of the genome, when bearing 5'-triphosphates, is the RIG-I ligand. The 5' region of the ZIKV genome is also a RIG-I agonist. We propose that RIG-I binds to the highly structured and conserved 5' region of flavivirus nascent transcripts before capping and that this mechanism leads to interferon secretion by infected cells.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Virus del Dengue/genética , Genoma Viral , Virus Zika/genética , Células Dendríticas/inmunología , Células Dendríticas/virología , Dengue/virología , Células HEK293 , Humanos , Receptores Inmunológicos , Replicación Viral/fisiología
12.
J Virol ; 91(20)2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28768856

RESUMEN

Attenuated measles virus (MV) is one of the most effective and safe vaccines available, making it an attractive candidate vector for preventing other infectious diseases. Yet the great capacity of this vaccine still needs to be understood at the molecular level. MV vaccine strains have different type I interferon (IFN)-inducing abilities that partially depend on the presence of 5' copy-back defective interfering genomes (DI-RNAs). DI-RNAs are pathogen-associated molecular patterns recognized by RIG-I-like receptors (RLRs) (RIG-I, MDA5, and LGP2) that activate innate immune signaling and shape the adaptive immune response. In this study, we characterized the DI-RNAs produced by various modified recombinant MVs (rMVs), including vaccine candidates, as well as wild-type MV. All tested rMVs produced 5' copy-back DI-RNAs that were different in length and nucleotide sequence but still respected the so-called "rule of six." We correlated the presence of DI-RNAs with a larger stimulation of the IFN-ß pathway and compared their immunostimulatory potentials. Importantly, we revealed that encapsidation of DI-RNA molecules within the MV nucleocapsid abolished their immunoactive properties. Furthermore, we identified specific interactions of DI-RNAs with both RIG-I and LGP2 but not MDA5. Our results suggest that DI-RNAs produced by rMV vaccine candidates may indeed strengthen their efficiency by triggering RLR signaling.IMPORTANCE Having been administered to hundreds of millions of children, the live attenuated measles virus (MV) vaccine is the safest and most widely used human vaccine, providing high protection with long-term memory. Additionally, recombinant MVs carrying heterologous antigens are promising vectors for new vaccines. The great capacity of this vaccine still needs to be elucidated at the molecular level. Here we document that recombinant MVs produce defective interfering genomes that have high immunostimulatory properties via their binding to RIG-I and LGP2 proteins, both of which are cytosolic nonself RNA sensors of innate immunity. Defective interfering genome production during viral replication should be considered of great importance due to the immunostimulatory properties of these genomes as intrinsic adjuvants produced by the vector that increase recognition by the innate immune system.


Asunto(s)
Genoma Viral , Helicasa Inducida por Interferón IFIH1/metabolismo , Virus del Sarampión/genética , ARN Helicasas/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Receptores de Ácido Retinoico/metabolismo , Línea Celular , Humanos , Inmunidad Innata , Interferón beta/metabolismo , Sarampión/virología , Vacuna Antisarampión/genética , Vacuna Antisarampión/inmunología , Virus del Sarampión/patogenicidad , Nucleocápside/metabolismo , ARN Viral/inmunología , Transducción de Señal
13.
J Virol ; 88(8): 4161-72, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24478441

RESUMEN

UNLABELLED: Lentiviral RNA genomes present a strong bias in their nucleotide composition with extremely high frequencies of A nucleotide in human immunodeficiency virus type 1 (HIV-1) and simian immunodeficiency virus (SIV). Based on the observation that human optimization of RNA virus gene fragments may abolish their ability to stimulate the type I interferon (IFN-I) response, we identified the most biased sequences along the SIV genome and showed that they are the most potent IFN-I stimulators. With the aim of designing an attenuated SIV genome based on a reduced capacity to activate the IFN-I response, we synthesized artificial SIV genomes whose biased sequences were optimized toward macaque average nucleotide composition without altering their regulatory elements or amino acid sequences. A synthetic SIV optimized with 169 synonymous mutations in gag and pol genes showed a 100-fold decrease in replicative capacity. Interestingly, a synthetic SIV optimized with 70 synonymous mutations in pol had a normal replicative capacity. Its ability to stimulate IFN-I was reduced when infected cells were cocultured with reporter cells. IFN regulatory factor 3 (IRF3) transcription factor was required for IFN-I stimulation, implicating cytosolic sensors in the detection of SIV-biased RNA in infected cells. No reversion of introduced mutations was observed for either of the optimized viruses after 10 serial passages. In conclusion, we have designed large-scale nucleotide-modified SIVs that may display attenuated pathogenic potential. IMPORTANCE: In this study, we synthesized artificial SIV genomes in which the most hyperbiased sequences were optimized to bring them closer to the nucleotide composition of the macaque SIV host. Interestingly, we generated a stable synthetic SIV optimized with 70 synonymous mutations in pol gene, which had a normal replicative capacity but a reduced ability to stimulate type I IFN. This demonstrates the possibility to rationally change viral nucleotide composition to design replicative and genetically stable lentiviruses with attenuated pathogenic potentials.


Asunto(s)
Interferón Tipo I/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/genética , Animales , Secuencia de Bases , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/inmunología , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Proteínas del Virus de la Inmunodeficiencia Humana/inmunología , Humanos , Interferón Tipo I/genética , Macaca mulatta , Datos de Secuencia Molecular , Mutación , Vacunas contra el SIDAS/genética , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/inmunología
14.
Vaccine ; 31(36): 3718-25, 2013 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-23742993

RESUMEN

Chikungunya virus (CHIKV), a mosquito-transmitted alphavirus, recently reemerged in the Indian Ocean, India and Southeast Asia, causing millions of cases of severe polyarthralgia. No specific treatment to prevent disease or vaccine to limit epidemics is currently available. Here we describe a recombinant live-attenuated measles vaccine (MV) expressing CHIKV virus-like particles comprising capsid and envelope structural proteins from the recent CHIKV strain La Reunion. Immunization of mice susceptible to measles virus induced high titers of CHIKV antibodies that neutralized several primary isolates. Specific cellular immune responses were also elicited. A single immunization with this vaccine candidate protected all mice from a lethal CHIKV challenge, and passive transfer of immune sera conferred protection to naïve mice. Measles vaccine is one of the safest and most effective human vaccines. A recombinant MV-CHIKV virus could make a safe and effective vaccine against chikungunya that deserves to be further tested in human trials.


Asunto(s)
Infecciones por Alphavirus/prevención & control , Virus Chikungunya/inmunología , Vacuna Antisarampión/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Proteínas de la Cápside/inmunología , Fiebre Chikungunya , Chlorocebus aethiops , Reacciones Cruzadas , Sueros Inmunes/inmunología , Inmunidad Celular , Inmunización Pasiva , Ratones , Ratones Transgénicos , Vacunas Atenuadas/inmunología , Células Vero , Proteínas del Envoltorio Viral/inmunología
15.
PLoS One ; 7(4): e33502, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22529893

RESUMEN

The genome of human immunodeficiency virus (HIV) has an average nucleotide composition strongly biased as compared to the human genome. The consequence of such nucleotide composition on HIV pathogenicity has not been investigated yet. To address this question, we analyzed the role of nucleotide bias of HIV-derived nucleic acids in stimulating type-I interferon response in vitro. We found that the biased nucleotide composition of HIV is detected in human cells as compared to humanized sequences, and triggers a strong innate immune response, suggesting the existence of cellular immune mechanisms able to discriminate RNA sequences according to their nucleotide composition or to detect specific secondary structures or linear motifs within biased RNA sequences. We then extended our analysis to the entire genome scale by testing more than 1300 HIV-1 complete genomes to look for an association between nucleotide composition of HIV-1 group M subtypes and their pathogenicity. We found that subtype D, which has an increased pathogenicity compared to the other subtypes, has the most divergent nucleotide composition relative to the human genome. These data support the hypothesis that the biased nucleotide composition of HIV-1 may be related to its pathogenicity.


Asunto(s)
VIH-1/genética , VIH-1/patogenicidad , Interferón Tipo I/biosíntesis , ARN Viral/química , Composición de Base , Línea Celular , Genes Virales , Genoma Viral , VIH-1/clasificación , Humanos
16.
Virology ; 419(2): 117-25, 2011 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-21890164

RESUMEN

We generated a replicating chimeric measles virus in which the hemagglutinin and fusion surface glycoproteins were replaced with the gp160 envelope glycoprotein of simian immunodeficiency virus (SIVmac239). Based on a previously cloned live-attenuated Schwarz vaccine strain of measles virus (MV), this chimera was rescued at high titers using reverse genetics in CD4+ target cells. Cytopathic effect consisted in the presence of large cell aggregates evolving to form syncytia, as observed during SIV infection. The morphology of the chimeric virus was identical to that of the parent MV particles. The presence of SIV gp160 as the only envelope protein on chimeric particles surface altered the cell tropism of the new virus from CD46+ to CD4+ cells. Used as an HIV candidate vaccine, this MV/SIVenv chimeric virus would mimic transient HIV-like infection, benefiting both from HIV-like tropism and the capacity of MV to replicate in dendritic cells, macrophages and lymphocytes.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Productos del Gen env/genética , Virus del Sarampión/genética , Virus del Sarampión/fisiología , Receptores CCR5/metabolismo , Virus de la Inmunodeficiencia de los Simios/genética , Replicación Viral , Animales , Western Blotting , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , Chlorocebus aethiops , Técnica del Anticuerpo Fluorescente , Productos del Gen env/metabolismo , Células Gigantes , Hemaglutininas Virales/genética , Humanos , Macaca mulatta , Receptores CCR5/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Virus de la Inmunodeficiencia de los Simios/metabolismo , Proteínas Virales de Fusión/genética
17.
Vaccine ; 28(41): 6730-9, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20688034

RESUMEN

Dengue disease is an increasing global health problem that threatens one-third of the world's population. To control this emerging arbovirus, an efficient preventive vaccine is still needed. Because four serotypes of dengue virus (DV) coexist and antibody-dependent enhanced infection may occur, most strategies developed so far rely on the administration of tetravalent formulations of four live attenuated or chimeric viruses. Here, we evaluated a new strategy based on the expression of a single minimal tetravalent DV antigen by a single replicating viral vector derived from pediatric live-attenuated measles vaccine (MV). We generated a recombinant MV vector expressing a DV construct composed of the four envelope domain III (EDIII) from the four DV serotypes fused with the ectodomain of the membrane protein (ectoM). After two injections in mice susceptible to MV infection, the recombinant vector induced neutralizing antibodies against the four serotypes of dengue virus. When immunized mice were further inoculated with live DV from each serotype, a strong memory neutralizing response was raised against all four serotypes. A combined measles-dengue vaccine might be attractive to immunize infants against both diseases where they co-exist.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Vacunas contra el Dengue/inmunología , Dengue/prevención & control , Vacuna Antisarampión/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/inmunología , Chlorocebus aethiops , Dengue/inmunología , Virus del Dengue/inmunología , Drosophila/citología , Escherichia coli/metabolismo , Vectores Genéticos , Inmunidad Humoral , Memoria Inmunológica , Masculino , Ratones , Ratones Transgénicos , Pruebas de Neutralización , Plásmidos , Proteínas Recombinantes/inmunología , Células Vero
18.
Virology ; 388(1): 191-203, 2009 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-19345390

RESUMEN

Although a live attenuated HIV vaccine is not currently considered for safety reasons, a strategy inducing both T cells and neutralizing antibodies to native assembled HIV-1 particles expressed by a replicating virus might mimic the advantageous characteristics of live attenuated vaccine. To this aim, we generated a live attenuated recombinant measles vaccine expressing HIV-1 Gag virus-like particles (VLPs) covered with gp160DeltaV1V2 Env protein. The measles-HIV virus replicated efficiently in cell culture and induced the intense budding of HIV particles covered with Env. In mice sensitive to MV infection, this recombinant vaccine stimulated high levels of cellular and humoral immunity to both MV and HIV with neutralizing activity. The measles-HIV virus infected human professional antigen-presenting cells, such as dendritic cells and B cells, and induced efficient presentation of HIV-1 epitopes and subsequent activation of human HIV-1 Gag-specific T cell clones. This candidate vaccine will be next tested in non-human primates. As a pediatric vaccine, it might protect children and adolescents simultaneously from measles and HIV.


Asunto(s)
Vacunas contra el SIDA/inmunología , Vacuna Antisarampión/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología , Animales , Anticipación Genética , Chlorocebus aethiops , Humanos , Virus del Sarampión , Proteína Cofactora de Membrana/genética , Ratones , Vacunas Sintéticas , Células Vero , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...