Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
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
2.
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
3.
Malar J ; 16(1): 259, 2017 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-28662722

RESUMEN

BACKGROUND: Yeast cells represent an established bioreactor to produce recombinant proteins for subunit vaccine development. In addition, delivery of vaccine antigens directly within heat-inactivated yeast cells is attractive due to the adjuvancy provided by the yeast cell. In this study, Pichia pastoris yeast lysates carrying the nucleoprotein (N) from the measles vaccine virus were evaluated as a novel subunit vaccine platform to deliver the circumsporozoite surface antigen (CS) of Plasmodium. When expressed in Pichia pastoris yeast, the N protein auto-assembles into highly multimeric ribonucleoparticles (RNPs). The CS antigen from Plasmodium berghei (PbCS) was expressed in Pichia pastoris yeast in fusion with N, generating recombinant PbCS-carrying RNPs in the cytoplasm of yeast cells. RESULTS: When evaluated in mice after 3-5 weekly subcutaneous injections, yeast lysates containing N-PbCS RNPs elicited strong anti-PbCS humoral responses, which were PbCS-dose dependent and reached a plateau by the pre-challenge time point. Protective efficacy of yeast lysates was dose-dependent, although anti-PbCS antibody titers were not predictive of protection. Multimerization of PbCS on RNPs was essential for providing benefit against infection, as immunization with monomeric PbCS delivered in yeast lysates was not protective. Three weekly injections with N-PbCS yeast lysates in combination with alum adjuvant produced sterile protection in two out of six mice, and significantly reduced parasitaemia in the other individuals from the same group. This parasitaemia decrease was of the same extent as in mice immunized with non-adjuvanted N-PbCS yeast lysates, providing evidence that the yeast lysate formulation did not require accessory adjuvants for eliciting efficient parasitaemia reduction. CONCLUSIONS: This study demonstrates that yeast lysates are an attractive auto-adjuvant and efficient platform for delivering multimeric PbCS on measles N-based RNPs. By combining yeast lysates that carry RNPs with a large panel of Plasmodium antigens, this technology could be applied to developing a multivalent vaccine against malaria.


Asunto(s)
Vacunas contra la Malaria/inmunología , Malaria/prevención & control , Nucleoproteínas , Pichia/fisiología , Plasmodium berghei/inmunología , Proteínas Protozoarias/inmunología , Proteínas Virales , Animales , Femenino , Ratones , Proteínas de la Nucleocápside , Nucleoproteínas/inmunología , Vacunas de Subunidad/inmunología , Proteínas Virales/inmunología
4.
J Immunol ; 182(9): 5740-7, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19380821

RESUMEN

Bid, a proapoptotic member of Bcl-2 family, is involved in Fas receptor signaling. Fas activation promotes human eosinophil cell death and is believed to accelerate the resolution of pulmonary Th2-driven allergic reaction in mice. We hypothesized that Bid would regulate eosinophil apoptosis and Ag-induced airway inflammation, particularly eosinophilia. C57BL/6 Bid(-/-) and wild-type mice were immunized and repeatedly challenged with OVA, and bronchoalveolar lavage (BAL) fluid, lung, and spleen were collected 4-240 h after the final challenge. Cultured BAL eosinophils from Bid-deficient mice showed resistance to Fas-mediated apoptotic DNA fragmentation, phosphatidylserine exposure, mitochondria depolarization, and caspase-3 activity. In addition, OVA-challenged Bid(-/-) mice had higher BAL eosinophilia and a lower proportion of BAL apoptotic eosinophils than Bid(+/+) mice. This was accompanied by augmented BAL levels of the eosinophilotactic cytokine, IL-5, and of the eosinophil-associated mediators, TGF-beta1 and fibronectin. Finally, cultured OVA-stimulated lung mononuclear cells and splenocytes from Bid-deficient mice showed increased release of the Th2-type cytokines, IL-4 and IL-5, but no change in cell number. We conclude that Bid modulates BAL eosinophilia by regulating both eosinophil apoptosis and Th2-type cytokine production.


Asunto(s)
Apoptosis/inmunología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/fisiología , Eosinófilos/inmunología , Eosinófilos/patología , Pulmón/patología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/patología , Animales , Apoptosis/genética , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/deficiencia , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Células Cultivadas , Citocinas/biosíntesis , Eosinófilos/metabolismo , Mediadores de Inflamación/fisiología , Pulmón/inmunología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hipersensibilidad Respiratoria/metabolismo , Células Th2/inmunología , Células Th2/metabolismo
5.
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.

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.
PLoS Pathog ; 3(2): e23, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17305428

RESUMEN

Theiler's virus, a picornavirus, persists for life in the central nervous system of mouse and causes a demyelinating disease that is a model for multiple sclerosis. The virus infects neurons first but persists in white matter glial cells, mainly oligodendrocytes and macrophages. The mechanism, by which the virus traffics from neurons to glial cells, and the respective roles of oligodendrocytes and macrophages in persistence are poorly understood. We took advantage of our previous finding that the shiverer mouse, a mutant with a deletion in the myelin basic protein gene (Mbp), is resistant to persistent infection to examine the role of myelin in persistence. Using immune chimeras, we show that resistance is not mediated by immune responses or by an efficient recruitment of inflammatory cells into the central nervous system. With both in vivo and in vitro experiments, we show that the mutation does not impair the permissiveness of neurons, oligodendrocytes, and macrophages to the virus. We demonstrate that viral antigens are present in cytoplasmic channels of myelin during persistent infection of wild-type mice. Using the optic nerve as a model, we show that the virus traffics from the axons of retinal ganglion cells to the cytoplasmic channels of myelin, and that this traffic is impaired by the shiverer mutation. These results uncover an unsuspected axon to myelin traffic of Theiler's virus and the essential role played by the infection of myelin/oligodendrocyte in persistence.


Asunto(s)
Encéfalo/virología , Vaina de Mielina/fisiología , Theilovirus/crecimiento & desarrollo , Animales , Antígenos Virales/análisis , Células de la Médula Ósea/fisiología , Infecciones por Cardiovirus/inmunología , Células Cultivadas , Ratones , Ratones Endogámicos C3H , Mutación , Vaina de Mielina/genética , Oligodendroglía/virología , Theilovirus/patogenicidad
8.
NPJ Vaccines ; 4: 12, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30820355

RESUMEN

Following the RTS,S malaria vaccine, which showed only partial protection with short-term memory, there is strong support to develop second-generation malaria vaccines that yield higher efficacy with longer duration. The use of replicating viral vectors to deliver subunit vaccines is of great interest due to their capacity to induce efficient cellular immune responses and long-term memory. The measles vaccine virus offers an efficient and safe live viral vector that could easily be implemented in the field. Here, we produced recombinant measles viruses (rMV) expressing malaria "gold standard" circumsporozoïte antigen (CS) of Plasmodium berghei (Pb) and Plasmodium falciparum (Pf) to test proof of concept of this delivery strategy. Immunization with rMV expressing PbCS or PfCS induced high antibody responses in mice that did not decrease for at least 22 weeks post-prime, as well as rapid development of cellular immune responses. The observed long-term memory response is key for development of second-generation malaria vaccines. Sterile protection was achieved in 33% of immunized mice, as usually observed with the CS antigen, and all other immunized animals were clinically protected from severe and lethal Pb ANKA-induced cerebral malaria. Further rMV-vectored malaria vaccine candidates expressing additional pre-erythrocytic and blood-stage antigens in combination with rMV expressing PfCS may provide a path to development of next generation malaria vaccines with higher efficacy.

10.
JMM Case Rep ; 5(5): e005145, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29896405

RESUMEN

Introduction: Although the current Zika virus (ZIKV) epidemic is a major public health concern, most reports have focused on congenital ZIKV syndrome, its most devastating manifestation. Severe ocular complications associated with ZIKV infections and possible pathogenetic factors are rarely described. Here, we describe three Venezuelan patients who developed severe ocular manifestations following ZIKV infections. We also analyse their serological response to ZIKV and dengue virus (DENV). Case presentation: One adult with bilateral optic neuritis, a child of 4 years of age with retrobulbar neuritis [corrected]. and a newborn with bilateral congenital glaucoma had a recent history of an acute exanthematous infection consistent with ZIKV infection. The results of ELISA tests indicated that all patients were seropositive for ZIKV and four DENV serotypes. Conclusion: Patients with ZIKV infection can develop severe ocular complications. Anti-DENV antibodies from previous infections could play a role in the pathogenesis of these complications. Well-designed epidemiological studies are urgently needed to measure the risk of ZIKV ocular complications and confirm whether they are associated with the presence of anti-flaviviral antibodies.

11.
J Nutr ; 137(12): 2730-6, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18029491

RESUMEN

Signaling of all-trans retinoic acid (ATRA) through nuclear retinoid acid (RA) receptors regulates several biological functions in airway epithelial cells, eosinophils, and immune cells, yet its impact on different in vivo aspects of pulmonary allergic reaction remains elusive. We compared the effect of a treatment with liposomally encapsulated ATRA (Lipo-ATRA) in a mouse model of ovalbumin (OVA)-induced T helper (Th) 2-type responses and airway remodeling. Daily intraperitoneal injections of 10 mg/kg Lipo-ATRA, at the time of each of the 2 systemic sensitizing injections, increased OVA-induced Immunoglobulin E synthesis, bronchoalveolar lavage (BAL) eosinophilia, and accumulation of IL-5, transforming-growth factor beta1, fibronectin, eotaxin/chemokine (C-C motif) ligand 11 (eotaxin/CCL11) and regulated upon activation, normal T expressed and secreted chemokine (C-C motif) ligand 5. In contrast, Lipo-ATRA, administered during each of the 4 intranasal OVA challenges, did not affect these variables. Regardless of the treatment regimen, Lipo-ATRA augmented mucin levels in BAL fluid and reduced lung total collagen content. In vitro incubation of mouse splenocytes or purified spleen cluster of differentiation (CD) 4-positive T lymphocytes, with ATRA, increased, respectively, OVA- and anti-CD 3 antibody-induced IL-4 and IL-5 production and inhibited IFNgamma release. These findings demonstrate that, when given during systemic sensitization, Lipo-ATRA exacerbates allergic immune and inflammatory responses, most likely by promoting Th2 development.


Asunto(s)
Asma/tratamiento farmacológico , Liposomas , Tretinoina/uso terapéutico , Animales , Asma/inducido químicamente , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Linfocitos T CD4-Positivos , Colágeno/metabolismo , Citocinas/análisis , Inmunoglobulina E , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ovalbúmina , Retinoides/sangre , Bazo/citología , Bazo/efectos de los fármacos , Factores de Tiempo , Tretinoina/administración & dosificación
12.
Sci Rep ; 7(1): 2561, 2017 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-28566766

RESUMEN

The type I interferon response plays a pivotal role in host defense against infectious agents and tumors, and promising therapeutic approaches rely on small molecules designed to boost this system. To identify such compounds, we developed a high-throughput screening assay based on HEK-293 cells expressing luciferase under the control of Interferon-Stimulated Response Elements (ISRE). An original library of 10,000 synthetic compounds was screened, and we identified a series of 1H-benzimidazole-4-carboxamide compounds inducing the ISRE promoter sequence, specific cellular Interferon-Stimulated Genes (ISGs), and the phosphorylation of Interferon Regulatory Factor (IRF) 3. ISRE induction by ChX710, a prototypical member of this chemical series, was dependent on the adaptor MAVS and IRF1, but was IRF3 independent. Although it was unable to trigger type I IFN secretion per se, ChX710 efficiently primed cellular response to transfected plasmid DNA as assessed by potent synergistic effects on IFN-ß secretion and ISG expression levels. This cellular response was dependent on STING, a key adaptor involved in the sensing of cytosolic DNA and immune activation by various pathogens, stress signals and tumorigenesis. Our results demonstrate that cellular response to cytosolic DNA can be boosted with a small molecule, and potential applications in antimicrobial and cancer therapies are discussed.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Factor 3 Regulador del Interferón/genética , Interferón Tipo I/química , Bibliotecas de Moléculas Pequeñas/farmacología , Citosol/química , ADN/química , ADN/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Elementos de Respuesta/genética , Bibliotecas de Moléculas Pequeñas/química , Transfección
13.
Eur Cytokine Netw ; 13(3): 340-9, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12231478

RESUMEN

IL-5 is present in the lung and in the circulation following allergenic challenges in humans and in animals, but its role in bronchopulmonary hyperreactivity (BHR) and lung and bronchoalveolar lavage fluid (BALF) eosinophilia remains unclear. Because compartmentalization of IL-5 is recognized, the anti-IL-5 monoclonal antibody TRFK-5 or its isotype control GL113 were delivered selectively intranasally (i.n.) and/or intravenously (i.v.) before the prior i.n. challenge with 10 mug OVA in BALB/c and BP2 "Biozzi" mice immunized according to optimized protocols with read-outs taken 24 h later. IL-5 in the BALF was suppressed by i.n. TRFK-5, whereas its production persisted in the serum. Conversely, i.v. TRFK-5 suppressed IL-5 in the serum but not in the BALF. IL-5 was suppressed in conditioned medium from lung explants from mice treated with i.n. TRFK-5, which did not affect the other Th2 cytokines, IL-4 and IL-13. IL-5 is thus present in the alveolar, pulmonary and circulatory compartments following an i.n. allergenic challenge. When specific anti-IL-5 antibodies were delivered by the same i.n. route, BALF eosinophilia was markedly reduced, whereas BHR and lung eosinophil sequestration persisted totally or mostly, in both strains. The passage of eosinophils from lungs to alveoli depends on IL-5 released into the BALF, but not into circulation, whereas their lung sequestration and BHR are mostly IL-5-independent. IL-5 alone does not account for the complexities of BHR or of eosinophil tissue trapping, and lung-targeted immunobiologicals should be delivered into the appropriate compartment in order to assess the role of specific mediators in experimental airways/lung allergy.


Asunto(s)
Hiperreactividad Bronquial/fisiopatología , Movimiento Celular/fisiología , Eosinófilos/citología , Hipersensibilidad/fisiopatología , Interleucina-5/fisiología , Pulmón/fisiopatología , Alveolos Pulmonares/citología , Animales , Líquido del Lavado Bronquioalveolar , Compartimento Celular , Hipersensibilidad/patología , Interleucina-4/metabolismo , Pulmón/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Especificidad de la Especie
14.
PLoS One ; 9(1): e86658, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24475165

RESUMEN

Yeasts are largely used as bioreactors for vaccine production. Usually, antigens are produced in yeast then purified and mixed with adjuvants before immunization. However, the purification costs and the safety concerns recently raised by the use of new adjuvants argue for alternative strategies. To this end, the use of whole yeast as both production and delivery system appears attractive. Here, we evaluated Pichia pastoris yeast as an alternative vaccine production and delivery system for the circumsporozoite protein (CS) of Plasmodium, the etiologic agent of malaria. The CS protein from Plasmodium berghei (Pb) was selected given the availability of the stringent C57Bl/6 mouse model of infection by Pb sporozoites, allowing the evaluation of vaccine efficacy in vivo. PbCS was multimerized by fusion to the measles virus (MV) nucleoprotein (N) known to auto-assemble in yeast in large-size ribonucleoprotein rods (RNPs). Expressed in P. pastoris, the N-PbCS protein generated highly multimeric and heterogenic RNPs bearing PbCS on their surface. Electron microscopy and immunofluorescence analyses revealed the shape of these RNPs and their localization in peripheral cytoplasmic inclusions. Subcutaneous immunization of C57Bl/6 mice with heat-inactivated whole P. pastoris expressing N-PbCS RNPs provided significant reduction of parasitemia after intradermal challenge with a high dose of parasites. Thus, in the absence of accessory adjuvants, a very low amount of PbCS expressed in whole yeast significantly decreased clinical damages associated with Pb infection in a highly stringent challenge model, providing a proof of concept of the intrinsic adjuvancy of this vaccine strategy. In addition to PbCS multimerization, the N protein contributed by itself to parasitemia delay and long-term mice survival. In the future, mixtures of whole recombinant yeasts expressing relevant Plasmodium antigens would provide a multivalent formulation applicable for antigen combination screening and possibly for large-scale production, distribution and delivery of a malaria vaccine in developing countries.


Asunto(s)
Reactores Biológicos , Sistemas de Liberación de Medicamentos/métodos , Vacunas contra la Malaria/biosíntesis , Pichia/metabolismo , Plasmodium berghei/química , Proteínas Protozoarias/metabolismo , Animales , Descubrimiento de Drogas , Técnica del Anticuerpo Fluorescente , Vacunas contra la Malaria/administración & dosificación , Virus del Sarampión/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Nucleoproteínas/metabolismo , Proteínas Protozoarias/aislamiento & purificación , Ribonucleoproteínas/biosíntesis
15.
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
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
19.
PLoS One ; 2(12): e1331, 2007 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18159229

RESUMEN

We showed previously that Theiler's virus, a neurotropic non-enveloped picornavirus of mouse, traffics from the axon of infected neurons into the surrounding myelin. When this traffic is interrupted, as in the shiverer mouse which bears a mutation in the myelin basic protein gene, the virus is unable to persist in the central nervous system. In the present work, we used the Wld(s) mutant mouse, a strain in which axonal degeneration is considerably slowed down, to show that axon to myelin traffic takes place in the absence of axon degeneration. Our results suggest the existence of a mechanism of transfer of axonal cytoplasm into the myelin which Theiler's virus might exploit to ensure its persistence.


Asunto(s)
Axones , Vaina de Mielina/fisiología , Theilovirus/fisiología , Animales , Femenino , Ratones , Ratones Mutantes , Nervio Óptico/virología , Retina/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Immunity ; 22(1): 31-42, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15664157

RESUMEN

Serum IgA is considered a discrete housekeeper of the immune system with multiple anti-inflammatory functions, whereas IgA-immune complexes mediate inflammatory responses. Here, we identify FcalphaRI as a molecular device that determines the nature of IgA responses. In the absence of sustained aggregation, receptor targeting by serum IgA or anti-FcalphaRI Fab inhibits activating responses of heterologous FcgammaR or FcepsilonRI. The inhibitory mechanism involves recruitment of tyrosine phosphatase SHP-1 to FcalphaRI and impairment of Syk, LAT, and ERK phosphorylation induced by FcepsilonRI engagement. SHP-1 recruitment is dependent on ERK. Conversely, sustained aggregation of FcalphaRI by multimeric ligands stimulates cell activation by recruiting high amounts of Syk and aborting SHP-1 binding. Both types of signals require the FcRgamma-ITAM motif. Anti-FcalphaRI Fab treatment suppresses manifestations of allergic asthma in FcalphaRI transgenic mice. These findings redefine FcalphaRI as a bifunctional inhibitory/activating receptor of the immune system that mediates both anti- and proinflammatory functions of IgA.


Asunto(s)
Antígenos CD/fisiología , Inmunoglobulina A/fisiología , Inflamación/inmunología , Receptores Fc/fisiología , Animales , Antígenos CD/metabolismo , Línea Celular , Citometría de Flujo , Humanos , Inmunoglobulina A/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Cinética , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fagocitosis , Fosforilación , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/metabolismo , Receptores Fc/metabolismo , Receptores de IgE/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA