Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Virol ; 98(5): e0023924, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38647327

RESUMEN

Dengue virus (DENV) represents a significant global health burden, with 50% of the world's population at risk of infection, and there is an urgent need for next-generation vaccines. Virus-like particle (VLP)-based vaccines, which mimic the antigenic structure of the virus but lack the viral genome, are an attractive approach. Here, we describe a dengue VLP (DENVLP) vaccine which generates a neutralizing antibody response against all four DENV serotypes in 100% of immunized non-human primates for up to 1 year. Additionally, DENVLP vaccination produced no ADE response against any of four DENV serotypes in vitro. DENVLP vaccination reduces viral replication in a non-human primate challenge model. We also show that transfer of purified IgG from immunized monkeys into immunodeficient mice protects against subsequent lethal DENV challenge, indicating a humoral mechanism of protection. These results indicate that this DENVLP vaccine is immunogenic and can be considered for clinical evaluation. Immunization of non-human primates with a tetravalent DENVLP vaccine induces high levels of neutralizing antibodies and reduces the severity of infection for all four dengue serotypes.IMPORTANCEDengue is a viral disease that infects nearly 400 million people worldwide and causes dengue hemorrhagic fever, which is responsible for 10,000 deaths each year. Currently, there is no therapeutic drug licensed to treat dengue infection, which makes the development of an effective vaccine essential. Virus-like particles (VLPs) are a safe and highly immunogenic platform that can be used in young children, immunocompromised individuals, as well as healthy adults. In this study, we describe the development of a dengue VLP vaccine and demonstrate that it induces a robust immune response against the dengue virus for over 1 year in monkeys. The immunity induced by this vaccine reduced live dengue infection in both murine and non-human primate models. These results indicate that our dengue VLP vaccine is a promising vaccine candidate.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Vacunas contra el Dengue , Virus del Dengue , Dengue , Vacunas de Partículas Similares a Virus , Replicación Viral , Animales , Anticuerpos Neutralizantes/inmunología , Virus del Dengue/inmunología , Vacunas contra el Dengue/inmunología , Vacunas contra el Dengue/administración & dosificación , Dengue/prevención & control , Dengue/inmunología , Dengue/virología , Anticuerpos Antivirales/inmunología , Ratones , Vacunas de Partículas Similares a Virus/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación , Humanos , Vacunación , Serogrupo , Inmunoglobulina G/inmunología , Modelos Animales de Enfermedad , Macaca fascicularis , Femenino , Macaca mulatta
2.
iScience ; 27(2): 108964, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38352232

RESUMEN

Continuing emergence of variants of concern resulting in reduced SARS-CoV-2 vaccine efficacy necessitates additional prevention strategies. The structure of VLPCOV-01, a lipid nanoparticle-encapsulated, self-amplifying RNA COVID-19 vaccine with a comparable immune response to BNT162b2, was revised by incorporating a modified base, 5-methylcytosine, to reduce reactogenicity, and an updated receptor-binding domain derived from the Brazil (gamma) variant. Interim analyses of a phase 1 dose-escalation booster vaccination study with the resulting construct, VLPCOV-02, in healthy, previously vaccinated Japanese individuals (N = 96) are reported (jRCT2051230005). A dose-related increase in solicited local and systemic adverse events was observed, which were generally rated mild or moderate. The most commonly occurring events were tenderness, pain, fatigue, and myalgia. Serum SARS-CoV-2 immunoglobulin titers increased during the 4 weeks post-immunization. VLPCOV-02 demonstrated a favorable safety profile compared with VLPCOV-01, with reduced adverse events and fewer fever events at an equivalent dose. These findings support further study of VLPCOV-02.

3.
bioRxiv ; 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37961509

RESUMEN

In order to improve vaccine effectiveness and safety profile of existing synthetic RNA-based vaccines, we have developed a self-amplifying RNA (saRNA)-based vaccine expressing membrane-anchored receptor binding domain (RBD) of SARS-CoV-2 S protein (S-RBD) and have demonstrated that a minimal dose of this saRNA vaccine elicits robust immune responses. Results from a recent clinical trial with 5-methylcytidine (5mC) incorporating saRNA vaccine demonstrated reduced vaccine-induced adverse effects while maintaining robust humoral responses. In this study, we investigate the mechanisms accounting for induction of efficient innate and adaptive immune responses and attenuated adverse effects induced by the 5mC-incorporated saRNA. We show that the 5mC-incorporating saRNA platform leads to prolonged and robust expression of antigen, while induction of type-I interferon (IFN-I), a key driver of reactogenicity, is attenuated in peripheral blood mononuclear cells (PBMCs), but not in macrophages and dendritic cells. Interestingly, we find that the major cellular source of IFN-I production in PBMCs is plasmacytoid dendritic cells (pDCs), which is attenuated upon 5mC incorporation in saRNA. In addition, we demonstrate that monocytes also play an important role in amplifying proinflammatory responses. Furthermore, we show that the detection of saRNA is mediated by a host cytosolic RNA sensor, RIG-I. Importantly, 5mC-incorporating saRNA vaccine candidate produced robust IgG responses against S-RBD upon injection in mice, thus providing strong support for the potential clinical use of 5mC-incorporating saRNA vaccines.

4.
Cell Rep Med ; 4(8): 101134, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37586325

RESUMEN

VLPCOV-01 is a lipid nanoparticle-encapsulated self-amplifying RNA (saRNA) vaccine that expresses a membrane-anchored receptor-binding domain (RBD) derived from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. A phase 1 study of VLPCOV-01 is conducted (jRCT2051210164). Participants who completed two doses of the BNT162b2 mRNA vaccine previously are randomized to receive one intramuscular vaccination of 0.3, 1.0, or 3.0 µg VLPCOV-01, 30 µg BNT162b2, or placebo. No serious adverse events have been reported. VLPCOV-01 induces robust immunoglobulin G (IgG) titers against the RBD protein that are maintained up to 26 weeks in non-elderly participants, with geometric means ranging from 5,037 (95% confidence interval [CI] 1,272-19,940) at 0.3 µg to 12,873 (95% CI 937-17,686) at 3 µg compared with 3,166 (95% CI 1,619-6,191) with 30 µg BNT162b2. Neutralizing antibody titers against all variants of SARS-CoV-2 tested are induced. VLPCOV-01 is immunogenic following low-dose administration. These findings support the potential for saRNA as a vaccine platform.


Asunto(s)
COVID-19 , Vacunas , Humanos , Persona de Mediana Edad , Vacunas contra la COVID-19/efectos adversos , Vacuna BNT162 , SARS-CoV-2/genética , ARN , COVID-19/prevención & control , Vacunas de ARNm
5.
Nat Commun ; 14(1): 2810, 2023 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-37208330

RESUMEN

Several vaccines have been widely used to counteract the global pandemic caused by SARS-CoV-2. However, due to the rapid emergence of SARS-CoV-2 variants of concern (VOCs), further development of vaccines that confer broad and longer-lasting protection against emerging VOCs are needed. Here, we report the immunological characteristics of a self-amplifying RNA (saRNA) vaccine expressing the SARS-CoV-2 Spike (S) receptor binding domain (RBD), which is membrane-anchored by fusing with an N-terminal signal sequence and a C-terminal transmembrane domain (RBD-TM). Immunization with saRNA RBD-TM delivered in lipid nanoparticles (LNP) efficiently induces T-cell and B-cell responses in non-human primates (NHPs). In addition, immunized hamsters and NHPs are protected against SARS-CoV-2 challenge. Importantly, RBD-specific antibodies against VOCs are maintained for at least 12 months in NHPs. These findings suggest that this saRNA platform expressing RBD-TM will be a useful vaccine candidate inducing durable immunity against emerging SARS-CoV-2 strains.


Asunto(s)
COVID-19 , Vacunas , Animales , Cricetinae , Humanos , SARS-CoV-2/genética , COVID-19/prevención & control , Motivo de Reconocimiento de ARN , Glicoproteína de la Espiga del Coronavirus/genética , Anticuerpos Neutralizantes , Anticuerpos Antivirales
6.
PLoS Negl Trop Dis ; 14(1): e0007860, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31999695

RESUMEN

In the past two decades there has been a significant expansion in the number of new therapeutic monoclonal antibodies (mAbs) that are approved by regulators. The discovery of these new medicines has been driven primarily by new approaches in inflammatory diseases and oncology, especially in immuno-oncology. Other recent successes have included new antibodies for use in viral diseases, including HIV. The perception of very high costs associated with mAbs has led to the assumption that they play no role in prophylaxis for diseases of poverty. However, improvements in antibody-expression yields and manufacturing processes indicate this is a cost-effective option for providing protection from many types of infection that should be revisited. Recent technology developments also indicate that several months of protection could be achieved with a single dose. Moreover, new methods in B cell sorting now enable the systematic identification of high-quality antibodies from humanized mice, or patients. This Review discusses the potential for passive immunization against schistosomiasis, fungal infections, dengue, and other neglected diseases.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Enfermedades Desatendidas/tratamiento farmacológico , Animales , Dengue/tratamiento farmacológico , Desarrollo de Medicamentos , Humanos , Inmunización Pasiva , Ratones , Micosis/tratamiento farmacológico , Esquistosomiasis/tratamiento farmacológico , Medicina Tropical
7.
Sci Transl Med ; 11(492)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092692

RESUMEN

Western, Eastern, and Venezuelan equine encephalitis viruses (WEEV, EEEV, and VEEV, respectively) are important mosquito-borne agents that pose public health and bioterrorism threats. Despite considerable advances in understanding alphavirus replication, there are currently no available effective vaccines or antiviral treatments against these highly lethal pathogens. To develop a potential countermeasure for viral encephalitis, we generated a trivalent, or three-component, EEV vaccine composed of virus-like particles (VLPs). Monovalent VLPs elicited neutralizing antibody responses and protected mice and nonhuman primates (NHPs) against homologous challenges, but they were not cross-protective. In contrast, NHPs immunized with trivalent VLPs were completely protected against aerosol challenge by each of these three EEVs. Passive transfer of IgG from immunized NHPs protected mice against aerosolized EEV challenge, demonstrating that the mechanism of protection was humoral. Because they are replication incompetent, these trivalent VLPs represent a potentially safe and effective vaccine that can protect against diverse encephalitis viruses.


Asunto(s)
Virus de la Encefalitis/inmunología , Encefalitis por Arbovirus/inmunología , Encefalitis por Arbovirus/prevención & control , Vacunas de Partículas Similares a Virus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Encefalitis por Arbovirus/patología , Encefalitis por Arbovirus/virología , Inmunización , Inmunoglobulina G/inmunología , Macaca fascicularis , Ratones Endogámicos BALB C , Vacunas de Partículas Similares a Virus/ultraestructura
8.
Proc Natl Acad Sci U S A ; 114(52): 13703-13707, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29203665

RESUMEN

Cleavage of the alphavirus precursor glycoprotein p62 into the E2 and E3 glycoproteins before assembly with the nucleocapsid is the key to producing fusion-competent mature spikes on alphaviruses. Here we present a cryo-EM, 6.8-Å resolution structure of an "immature" Chikungunya virus in which the cleavage site has been mutated to inhibit proteolysis. The spikes in the immature virus have a larger radius and are less compact than in the mature virus. Furthermore, domains B on the E2 glycoproteins have less freedom of movement in the immature virus, keeping the fusion loops protected under domain B. In addition, the nucleocapsid of the immature virus is more compact than in the mature virus, protecting a conserved ribosome-binding site in the capsid protein from exposure. These differences suggest that the posttranslational processing of the spikes and nucleocapsid is necessary to produce infectious virus.


Asunto(s)
Virus Chikungunya/química , Virus Chikungunya/ultraestructura , Glicoproteínas/química , Proteínas del Envoltorio Viral/química , Virus Chikungunya/metabolismo , Microscopía por Crioelectrón , Glicoproteínas/metabolismo , Dominios Proteicos , Estructura Secundaria de Proteína , Proteínas del Envoltorio Viral/metabolismo
9.
J Virol ; 91(23)2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28956764

RESUMEN

Dengue viruses (DENV) infect 50 to 100 million people each year. The spread of DENV-associated infections is one of the most serious public health problems worldwide, as there is no widely available vaccine or specific therapeutic for DENV infections. To address this, we developed a novel tetravalent dengue vaccine by utilizing virus-like particles (VLPs). We created recombinant DENV1 to -4 (DENV1-4) VLPs by coexpressing precursor membrane (prM) and envelope (E) proteins, with an F108A mutation in the fusion loop structure of E to increase the production of VLPs in mammalian cells. Immunization with DENV1-4 VLPs as individual, monovalent vaccines elicited strong neutralization activity against each DENV serotype in mice. For use as a tetravalent vaccine, DENV1-4 VLPs elicited high levels of neutralization activity against all four serotypes simultaneously. The neutralization antibody responses induced by the VLPs were significantly higher than those with DNA or recombinant E protein immunization. Moreover, antibody-dependent enhancement (ADE) was not observed against any serotype at a 1:10 serum dilution. We also demonstrated that the Zika virus (ZIKV) VLP production level was enhanced by introducing the same F108A mutation into the ZIKV envelope protein. Taken together, these results suggest that our strategy for DENV VLP production is applicable to other flavivirus VLP vaccine development, due to the similarity in viral structures, and they describe the promising development of an effective tetravalent vaccine against the prevalent flavivirus.IMPORTANCE Dengue virus poses one of the most serious public health problems worldwide, and the incidence of diseases caused by the virus has increased dramatically. Despite decades of effort, there is no effective treatment against dengue. A safe and potent vaccine against dengue is still needed. We developed a novel tetravalent dengue vaccine by using virus-like particles (VLPs), which are noninfectious because they lack the viral genome. Previous attempts of other groups to use dengue VLPs resulted in generally poor yields. We found that a critical amino acid mutation in the envelope protein enhances the production of VLPs. Our tetravalent vaccine elicited potent neutralizing antibody responses against all four DENV serotypes. Our findings can also be applied to vaccine development against other flaviviruses, such as Zika virus or West Nile virus.


Asunto(s)
Vacunas contra el Dengue/química , Flavivirus/inmunología , Vacunas de Partículas Similares a Virus/química , Vacunas de Partículas Similares a Virus/inmunología , Proteínas del Envoltorio Viral/genética , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Acrecentamiento Dependiente de Anticuerpo , Dengue/inmunología , Vacunas contra el Dengue/administración & dosificación , Vacunas contra el Dengue/inmunología , Virus del Dengue/genética , Flavivirus/genética , Inmunogenicidad Vacunal , Ratones , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Serogrupo , Vacunas de Partículas Similares a Virus/administración & dosificación , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/inmunología , Virus Zika/inmunología , Infección por el Virus Zika/inmunología
10.
Clin Vaccine Immunol ; 24(7)2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28515133

RESUMEN

Virus-like particles (VLPs) are noninfectious multiprotein structures that are engineered to self-assemble from viral structural proteins. Here, we developed a novel VLP-based vaccine platform utilizing VLPs from the chikungunya virus. We identified two regions within the envelope protein, a structural component of chikungunya, where foreign antigens can be inserted without compromising VLP structure. Our VLP displays 480 copious copies of an inserted antigen on the VLP surface in a highly symmetric manner and is thus capable of inducing strong immune responses against any inserted antigen. Furthermore, by mimicking the structure of the immature form of the virus, we altered our VLP's in vivo dynamics and enhanced its immunogenicity. We used the circumsporozoite protein (CSP) of the Plasmodium falciparum malaria parasite as an antigen and demonstrated that our VLP-based vaccine elicits strong immune responses against CSP in animals. The sera from immunized monkeys protected mice from malaria infection. Likewise, mice vaccinated with P. yoelii CSP-containing VLPs were protected from an infectious sporozoite challenge. Hence, our uniquely engineered VLP platform can serve as a blueprint for the development of vaccines against other pathogens and diseases.


Asunto(s)
Virus Chikungunya/genética , Portadores de Fármacos , Vacunas contra la Malaria/inmunología , Malaria/prevención & control , Proteínas Protozoarias/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Macaca mulatta , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Masculino , Ratones Endogámicos BALB C , Plasmodium falciparum/genética , Plasmodium falciparum/inmunología , Plasmodium yoelii/genética , Plasmodium yoelii/inmunología , Proteínas Protozoarias/genética , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/genética
11.
J Virol ; 90(3): 1169-77, 2016 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26537684

RESUMEN

UNLABELLED: Chikungunya virus is a positive-stranded RNA alphavirus. Structures of chikungunya virus-like particles in complex with strongly neutralizing antibody Fab fragments (8B10 and 5F10) were determined using cryo-electron microscopy and X-ray crystallography. By fitting the crystallographically determined structures of these Fab fragments into the cryo-electron density maps, we show that Fab fragments of antibody 8B10 extend radially from the viral surface and block receptor binding on the E2 glycoprotein. In contrast, Fab fragments of antibody 5F10 bind the tip of the E2 B domain and lie tangentially on the viral surface. Fab 5F10 fixes the B domain rigidly to the surface of the virus, blocking exposure of the fusion loop on glycoprotein E1 and therefore preventing the virus from becoming fusogenic. Although Fab 5F10 can neutralize the wild-type virus, it can also bind to a mutant virus without inhibiting fusion or attachment. Although the mutant virus is no longer able to propagate by extracellular budding, it can, however, enter the next cell by traveling through junctional complexes without being intercepted by a neutralizing antibody to the wild-type virus, thus clarifying how cell-to-cell transmission can occur. IMPORTANCE: Alphaviral infections are transmitted mainly by mosquitoes. Chikungunya virus (CHIKV), which belongs to the Alphavirus genus, has a wide distribution in the Old World that has expanded in recent years into the Americas. There are currently no vaccines or drugs against alphaviral infections. Therefore, a better understanding of CHIKV and its associated neutralizing antibodies will aid in the development of effective treatments.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Virus Chikungunya/inmunología , Virus Chikungunya/ultraestructura , Virosomas/inmunología , Virosomas/ultraestructura , Virus Chikungunya/química , Virus Chikungunya/fisiología , Microscopía por Crioelectrón , Cristalografía por Rayos X , Humanos , Fragmentos Fab de Inmunoglobulinas/metabolismo , Modelos Moleculares , Unión Proteica , Virosomas/química , Acoplamiento Viral
12.
Proc Natl Acad Sci U S A ; 112(45): 13898-903, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26504196

RESUMEN

Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes severe acute and chronic disease in humans. Although highly inhibitory murine and human monoclonal antibodies (mAbs) have been generated, the structural basis of their neutralizing activity remains poorly characterized. Here, we determined the cryo-EM structures of chikungunya virus-like particles complexed with antibody fragments (Fab) of two highly protective human mAbs, 4J21 and 5M16, that block virus fusion with host membranes. Both mAbs bind primarily to sites within the A and B domains, as well as to the B domain's ß-ribbon connector of the viral glycoprotein E2. The footprints of these antibodies on the viral surface were consistent with results from loss-of-binding studies using an alanine scanning mutagenesis-based epitope mapping approach. The Fab fragments stabilized the position of the B domain relative to the virus, particularly for the complex with 5M16. This finding is consistent with a mechanism of neutralization in which anti-CHIKV mAbs that bridge the A and B domains impede movement of the B domain away from the underlying fusion loop on the E1 glycoprotein and therefore block the requisite pH-dependent fusion of viral and host membranes.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Fiebre Chikungunya/terapia , Virus Chikungunya/inmunología , Microscopía por Crioelectrón/métodos , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/uso terapéutico , Humanos , Conformación Proteica
13.
Lancet ; 384(9959): 2046-52, 2014 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-25132507

RESUMEN

BACKGROUND: Chikungunya virus--a mosquito-borne alphavirus--is endemic in Africa and south and southeast Asia and has recently emerged in the Caribbean. No drugs or vaccines are available for treatment or prevention. We aimed to assess the safety, tolerability, and immunogenicity of a new candidate vaccine. METHODS: VRC 311 was a phase 1, dose-escalation, open-label clinical trial of a virus-like particle (VLP) chikungunya virus vaccine, VRC-CHKVLP059-00-VP, in healthy adults aged 18-50 years who were enrolled at the National Institutes of Health Clinical Center (Bethesda, MD, USA). Participants were assigned to sequential dose level groups to receive vaccinations at 10 µg, 20 µg, or 40 µg on weeks 0, 4, and 20, with follow-up for 44 weeks after enrolment. The primary endpoints were safety and tolerability of the vaccine. Secondary endpoints were chikungunya virus-specific immune responses assessed by ELISA and neutralising antibody assays. This trial is registered with ClinicalTrials.gov, NCT01489358. FINDINGS: 25 participants were enrolled from Dec 12, 2011, to March 22, 2012, into the three dosage groups: 10 µg (n=5), 20 µg (n=10), and 40 µg (n=10). The protocol was completed by all five participants at the 10 µg dose, all ten participants at the 20 µg dose, and eight of ten participants at the 40 µg dose; non-completions were for personal circumstances unrelated to adverse events. 73 vaccinations were administered. All injections were well tolerated, with no serious adverse events reported. Neutralising antibodies were detected in all dose groups after the second vaccination (geometric mean titres of the half maximum inhibitory concentration: 2688 in the 10 µg group, 1775 in the 20 µg group, and 7246 in the 40 µg group), and a significant boost occurred after the third vaccination in all dose groups (10 µg group p=0·0197, 20 µg group p<0·0001, and 40 µg group p<0·0001). 4 weeks after the third vaccination, the geometric mean titres of the half maximum inhibitory concentration were 8745 for the 10 µg group, 4525 for the 20 µg group, and 5390 for the 40 µg group. INTERPRETATION: The chikungunya VLP vaccine was immunogenic, safe, and well tolerated. This study represents an important step in vaccine development to combat this rapidly emerging pathogen. Further studies should be done in a larger number of participants and in more diverse populations. FUNDING: Intramural Research Program of the Vaccine Research Center, National Institute of Allergy and Infectious Diseases, and National Institutes of Health.


Asunto(s)
Virus Chikungunya/inmunología , Vacunas Virales/administración & dosificación , Adolescente , Adulto , Anticuerpos Neutralizantes/análisis , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Tolerancia Inmunológica , Masculino , Persona de Mediana Edad , Vacunación
14.
PLoS Pathog ; 9(4): e1003312, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23637602

RESUMEN

Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes global epidemics of a debilitating polyarthritis in humans. As there is a pressing need for the development of therapeutic agents, we screened 230 new mouse anti-CHIKV monoclonal antibodies (MAbs) for their ability to inhibit infection of all three CHIKV genotypes. Four of 36 neutralizing MAbs (CHK-102, CHK-152, CHK-166, and CHK-263) provided complete protection against lethality as prophylaxis in highly susceptible immunocompromised mice lacking the type I IFN receptor (Ifnar(-/-) ) and mapped to distinct epitopes on the E1 and E2 structural proteins. CHK-152, the most protective MAb, was humanized, shown to block viral fusion, and require Fc effector function for optimal activity in vivo. In post-exposure therapeutic trials, administration of a single dose of a combination of two neutralizing MAbs (CHK-102+CHK-152 or CHK-166+CHK-152) limited the development of resistance and protected immunocompromised mice against disease when given 24 to 36 hours before CHIKV-induced death. Selected pairs of highly neutralizing MAbs may be a promising treatment option for CHIKV in humans.


Asunto(s)
Infecciones por Alphavirus/prevención & control , Infecciones por Alphavirus/terapia , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Receptor de Interferón alfa y beta/genética , Proteínas Estructurales Virales/inmunología , Células 3T3 , Aedes , Infecciones por Alphavirus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Línea Celular , Fiebre Chikungunya , Virus Chikungunya/inmunología , Chlorocebus aethiops , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Vero , Proteínas del Envoltorio Viral/inmunología
15.
Elife ; 2: e00435, 2013 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-23577234

RESUMEN

A 5.3 Å resolution, cryo-electron microscopy (cryoEM) map of Chikungunya virus-like particles (VLPs) has been interpreted using the previously published crystal structure of the Chikungunya E1-E2 glycoprotein heterodimer. The heterodimer structure was divided into domains to obtain a good fit to the cryoEM density. Differences in the T = 4 quasi-equivalent heterodimer components show their adaptation to different environments. The spikes on the icosahedral 3-fold axes and those in general positions are significantly different, possibly representing different phases during initial generation of fusogenic E1 trimers. CryoEM maps of neutralizing Fab fragments complexed with VLPs have been interpreted using the crystal structures of the Fab fragments and the VLP structure. Based on these analyses the CHK-152 antibody was shown to stabilize the viral surface, hindering the exposure of the fusion-loop, likely neutralizing infection by blocking fusion. The CHK-9, m10 and m242 antibodies surround the receptor-attachment site, probably inhibiting infection by blocking cell attachment. DOI:http://dx.doi.org/10.7554/eLife.00435.001.


Asunto(s)
Anticuerpos Neutralizantes/ultraestructura , Anticuerpos Antivirales/ultraestructura , Virus Chikungunya/ultraestructura , Vacunas de Partículas Similares a Virus/ultraestructura , Proteínas del Envoltorio Viral/ultraestructura , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Sitios de Unión , Virus Chikungunya/inmunología , Virus Chikungunya/metabolismo , Virus Chikungunya/patogenicidad , Microscopía por Crioelectrón , Cristalografía por Rayos X , Fragmentos Fab de Inmunoglobulinas/metabolismo , Fragmentos Fab de Inmunoglobulinas/ultraestructura , Modelos Moleculares , Unión Proteica , Conformación Proteica , Vacunas de Partículas Similares a Virus/metabolismo , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/metabolismo , Virión/inmunología , Virión/metabolismo , Virión/ultraestructura , Internalización del Virus
16.
J Virol ; 86(16): 8879-83, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22647698

RESUMEN

Virus-like particles (VLPs) can be generated from Chikungunya virus (CHIKV), but different strains yield variable quantities of particles. Here, we define the genetic basis for these differences and show that amino acid 234 in E2 substantially affects VLP production. This site is located within the acid-sensitive region (ASR) known to initiate a major conformational change in E1/E2. Selected other mutations in the ASR, or changes in pH, also increased VLP yield. These results demonstrate that the ASR of E2 plays an important role in regulating particle generation.


Asunto(s)
Virus Chikungunya/fisiología , Multimerización de Proteína , Proteínas Virales/metabolismo , Ensamble de Virus , Virus Chikungunya/inmunología , Humanos , Vacunas de Virosoma/inmunología , Vacunas Virales/inmunología
17.
Nat Med ; 16(3): 334-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20111039

RESUMEN

Chikungunya virus (CHIKV) has infected millions of people in Africa, Europe and Asia since this alphavirus reemerged from Kenya in 2004. The severity of the disease and the spread of this epidemic virus present a serious public health threat in the absence of vaccines or antiviral therapies. Here, we describe a new vaccine that protects against CHIKV infection of nonhuman primates. We show that selective expression of viral structural proteins gives rise to virus-like particles (VLPs) in vitro that resemble replication-competent alphaviruses. Immunization with these VLPs elicited neutralizing antibodies against envelope proteins from alternative CHIKV strains. Monkeys immunized with VLPs produced high-titer neutralizing antibodies that protected against viremia after high-dose challenge. We transferred these antibodies into immunodeficient mice, where they protected against subsequent lethal CHIKV challenge, indicating a humoral mechanism of protection. Immunization with alphavirus VLP vaccines represents a strategy to contain the spread of CHIKV and related pathogenic viruses in humans.


Asunto(s)
Infecciones por Alphavirus/prevención & control , Virus Chikungunya/inmunología , Vacunas Virales/uso terapéutico , Infecciones por Alphavirus/inmunología , Animales , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , Células Cultivadas , Femenino , Humanos , Inmunoglobulina G/inmunología , Macaca mulatta/inmunología , Macaca mulatta/virología , Ratones , Ratones Endogámicos BALB C , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Viremia/inmunología , Viremia/prevención & control
18.
J Immunol ; 183(4): 2425-34, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19620307

RESUMEN

Prime-boost immunization with gene-based vectors has been developed to generate more effective vaccines for AIDS, malaria, and tuberculosis. Although these vectors elicit potent T cell responses, the mechanisms by which they stimulate immunity are not well understood. In this study, we show that immunization by a single gene product, HIV-1 envelope, with alternative vector combinations elicits CD8(+) cells with different fine specificities and kinetics of mobilization. Vaccine-induced CD8(+) T cells recognized overlapping third V region loop peptides. Unexpectedly, two anchor variants bound H-2D(d) better than the native sequences, and clones with distinct specificities were elicited by alternative vectors. X-ray crystallography revealed major differences in solvent exposure of MHC-bound peptide epitopes, suggesting that processed HIV-1 envelope gave rise to MHC-I/peptide conformations recognized by distinct CD8(+) T cell populations. These findings suggest that different gene-based vectors generate peptides with alternative conformations within MHC-I that elicit distinct T cell responses after vaccination.


Asunto(s)
Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Antígenos H-2/genética , Antígenos H-2/inmunología , Proteína gp120 de Envoltorio del VIH/genética , Proteína gp120 de Envoltorio del VIH/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/metabolismo , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Línea Celular , Células Cultivadas , Células Clonales , Epítopos de Linfocito T/administración & dosificación , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/metabolismo , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Vectores Genéticos/metabolismo , Antígenos H-2/administración & dosificación , Antígenos H-2/metabolismo , Proteína gp120 de Envoltorio del VIH/administración & dosificación , Proteína gp120 de Envoltorio del VIH/metabolismo , Antígeno de Histocompatibilidad H-2D , Inmunización Secundaria , Ratones , Ratones Endogámicos BALB C , Unión Proteica/genética , Unión Proteica/inmunología
19.
Mol Cell Biol ; 26(10): 3864-74, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16648481

RESUMEN

The protein arginine methyltransferases (PRMTs) include a family of proteins with related putative methyltransferase domains that modify chromatin and regulate cellular transcription. Although some family members, PRMT1 and PRMT4, have been implicated in transcriptional modulation or intracellular signaling, the roles of other PRMTs in diverse cellular processes have not been fully established. Here, we report that PRMT2 inhibits NF-kappaB-dependent transcription and promotes apoptosis. PRMT2 exerted this effect by blocking nuclear export of IkappaB-alpha through a leptomycin-sensitive pathway, increasing nuclear IkappaB-alpha and decreasing NF-kappaB DNA binding. The highly conserved S-adenosylmethionine-binding domain of PRMT2 mediated this effect. PRMT2 also rendered cells susceptible to apoptosis by cytokines or cytotoxic drugs, likely due to its effects on NF-kappaB. Mouse embryo fibroblasts from PRMT2 genetic knockouts showed elevated NF-kappaB activity and decreased susceptibility to apoptosis compared to wild-type or complemented cells. Taken together, these data suggest that PRMT2 inhibits cell activation and promotes programmed cell death through this NF-kappaB-dependent mechanism.


Asunto(s)
Apoptosis/fisiología , Proteínas de Unión al ADN/metabolismo , FN-kappa B/antagonistas & inhibidores , Proteína O-Metiltransferasa/metabolismo , Transcripción Genética/efectos de los fármacos , Alanina/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/farmacología , Relación Dosis-Respuesta a Droga , Ensayo de Cambio de Movilidad Electroforética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fluoresceína-5-Isotiocianato , Técnica del Anticuerpo Fluorescente , Colorantes Fluorescentes , Eliminación de Gen , Genes Reporteros , Glutatión Transferasa/metabolismo , Humanos , Inmunohistoquímica , Luciferasas/metabolismo , Ratones , Microscopía Confocal , Células 3T3 NIH , Plásmidos/genética , Pruebas de Precipitina , Proteína O-Metiltransferasa/química , Proteína O-Metiltransferasa/genética , Proteína O-Metiltransferasa/farmacología , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
20.
Vaccine ; 24(17): 3677-85, 2006 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-16085341

RESUMEN

We previously reported that a mutant full-sized plasmid DNA vaccine regime in macaques was effective against a homologous challenge [Akahata W, Ido E, Shimada T, Katsuyama K, Yamamoto H, Uesaka H, et al. DNA vaccination of macaques by a full genome HIV-1 plasmid which produces non-infectious virus particles. Virology 2000;275:116-24; Akahata W, Ido E, Akiyama H, Uesaka H, Enose Y, Horiuchi R, et al. DNA vaccination of macaques by a full genome SHIV-1 plasmid that produces non-infectious virus particles. J Gen Virol 2003;84:2237-44]. In this study, to evaluate the DNA vaccination regime against a heterologous challenge, a novel plasmid named pSHIV-ZF1*IL-2 was constructed. Four monkeys were intramuscularly and intradermally injected four times with the pSHIV-ZF1*IL-2. Vaccinated monkeys were intravenously challenged with a highly pathogenic, heterologous SHIV at 11 weeks post vaccination. All the vaccinated monkeys suppressed the challenge virus rapidly under the detectable level by 16 weeks post challenge. One vaccinated monkey was protected from a loss of CD4+ T cells. These results suggest pSHIV-ZF1*IL-2 alone seems partially effective even against a challenge with a heterologous, pathogenic virus.


Asunto(s)
Vacunas contra el SIDA/inmunología , Genoma Viral , VIH-1/inmunología , Interleucina-2/genética , Plásmidos/genética , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas de ADN/inmunología , Virión/fisiología , Animales , Recuento de Linfocito CD4 , Células COS , Chlorocebus aethiops , Femenino , Anticuerpos Anti-VIH/sangre , Proteínas gp160 de Envoltorio del VIH/inmunología , Macaca mulatta , Ratones , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...