Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 215
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 162(5): 1090-100, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26279189

RESUMEN

Epstein-Barr virus (EBV) represents a major global health problem. Though it is associated with infectious mononucleosis and ∼200,000 cancers annually worldwide, a vaccine is not available. The major target of immunity is EBV glycoprotein 350/220 (gp350) that mediates attachment to B cells through complement receptor 2 (CR2/CD21). Here, we created self-assembling nanoparticles that displayed different domains of gp350 in a symmetric array. By focusing presentation of the CR2-binding domain on nanoparticles, potent neutralizing antibodies were elicited in mice and non-human primates. The structurally designed nanoparticle vaccine increased neutralization 10- to 100-fold compared to soluble gp350 by targeting a functionally conserved site of vulnerability, improving vaccine-induced protection in a mouse model. This rational approach to EBV vaccine design elicited potent neutralizing antibody responses by arrayed presentation of a conserved viral entry domain, a strategy that can be applied to other viruses.


Asunto(s)
Vacunas contra Herpesvirus/química , Vacunas contra Herpesvirus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Cristalografía por Rayos X , Diseño de Fármacos , Femenino , Herpesvirus Humano 4 , Vacunas contra Herpesvirus/genética , Vacunas contra Herpesvirus/aislamiento & purificación , Macaca fascicularis , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Nanopartículas/ultraestructura , Receptores de Complemento 3d/química , Receptores de Complemento 3d/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación
2.
Plant J ; 111(5): 1308-1323, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35778946

RESUMEN

Terpene synthases (TPSs) have diverse biological functions in plants. Though the roles of TPSs in herbivore defense are well established in many plant species, their role in bacterial defense has been scarce and is emerging. Through functional genomics, here we report the in planta role of potato (Solanum tuberosum) terpene synthase (StTPS18) in bacterial defense. Expression of StTPS18 was highest in leaves and was induced in response to Pseudomonas syringae and methyl jasmonate treatments. The recombinant StTPS18 exhibited bona fide (E,E)-farnesol synthase activity forming a sesquiterpenoid, (E,E)-farnesol as the sole product, utilising (E,E)-farnesyl diphosphate (FPP). Subcellular localization of GFP fusion protein revealed that StTPS18 is localized to the cytosol. Silencing and overexpression of StTPS18 in potato resulted in reduced and enhanced tolerance, respectively, to bacterial pathogens P. syringae and Ralstonia solanacearum. Bacterial growth assay using medium containing (E,E)-farnesol significantly inhibited P. syringae growth. Moreover, StTPS18 overexpressing transgenic potato and Nicotiana tabacum leaves, and (E,E)-farnesol and P. syringae infiltrated potato leaves exhibited elevated expression of sterol pathway and members of pathogenesis-related genes with enhanced phytosterol accumulation. Interestingly, enhanced phytosterols in 13 C3 -(E,E)-farnesol infiltrated potato leaves were devoid of any noticeable 13 C labeling, indicating no direct utilization of (E,E)-farnesol in phytosterols formation. Furthermore, leaves of StTPS18 overexpressing transgenic lines had no detectable (E,E)-farnesol similar to the control plant, and emitted lower levels of sesquiterpenes than the control. These findings point towards an indirect involvement of StTPS18 and its product (E,E)-farnesol in bacterial defense through upregulation of phytosterol biosynthesis and defense genes.


Asunto(s)
Fitosteroles , Solanum tuberosum , Farnesol/metabolismo , Fitosteroles/metabolismo , Enfermedades de las Plantas/microbiología , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Solanum tuberosum/metabolismo , Nicotiana/metabolismo
3.
Physiol Plant ; 171(1): 7-21, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32880963

RESUMEN

Terpene synthases (TPSs) produce a variety of terpenoids that play numerous functional roles in primary and secondary metabolism, as well as in ecological interactions. Here, we report the functional characterization of an inducible potato TPS gene encoding bulnesol/elemol synthase (StBUS/ELS). The expression of StBUS/ELS in potato leaves was significantly induced in response to both bacterial (Pseudomonas syringae) and fungal (Alternaria solani) infection as well as methyl jasmonate treatment, indicating its role in defense. The leaves exhibited the highest StBUS/ELS expression followed by the stem with least and similar expression in tuber, sprout and root. Recombinant StBUS/ELS catalyzed the formation of different sesquiterpenes by utilizing farnesyl diphosphate as substrate, and the monoterpene geraniol from geranyl diphosphate. Among the sesquiterpenes formed by StBUS/ELS, elemol was the predominant product followed by α-bulnesene, bulnesol and ß-elemene. Further gas chromatography-mass spectrometry (GC-MS) analysis of StBUS/ELS assay products at different injection temperatures revealed elemol and bulnesol as the major products at 275 and 200/150°C, respectively, without much change in the levels of minor products. This indicated thermal rearrangement of bulnesol into elemol at higher temperatures. Transient overexpression of StBUS/ELS in potato leaves conferred tolerance against the growth of bacteria P. syringae and Ralstonia solanacearum, and the fungus A. solani. Further, expression analysis of pathogenesis-related (PR) genes in StBUS/ELS overexpressing leaves showed no significant change in comparison to control, indicating a direct involvement of StBUS/ELS enzymatic products against pathogens. Overall, our study suggested that StBUS/ELS is a pathogen-inducible TPS encoding bulnesol/elemol synthase and could provide a direct role in defense against biotic stress in potato.


Asunto(s)
Transferasas Alquil y Aril , Sesquiterpenos , Solanum tuberosum , Transferasas Alquil y Aril/genética , Alternaria , Proteínas de Plantas/genética , Solanum tuberosum/genética , Terpenos
4.
Niger J Clin Pract ; 24(9): 1410-1413, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34531358

RESUMEN

Rehabilitation of completely edentulous patients with implant-supported prosthesis (ISP) is the need of the day, but in many patients, the routine ISP is not possible due to the severe atrophic residual ridges. The present case series describes three cases with atrophic ridges rehabilitated using zygomatic implants and/or All-on-4 treatment concepts. In case 1 and case 3, in maxillary arch, there was not enough bone in Zone 3 to rehabilitate the patient with routine protocol followed for the All-on-4 treatment concept, so in these two cases, zygomatic implants were placed. Case 1 and case 2 were rehabilitated with Malo Bridge and case 3 with acrylic teeth-hybrid prosthesis using a computerized milling procedure to obtain improved fit, function, esthetics, and ease of retrievability whenever required.


Asunto(s)
Implantes Dentales , Arcada Edéntula , Prótesis Dental de Soporte Implantado , Estudios de Seguimiento , Humanos , Arcada Edéntula/cirugía , Maxilar/cirugía , Prótesis e Implantes , Resultado del Tratamiento , Cigoma/cirugía
5.
Nature ; 514(7524): 642-5, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25119033

RESUMEN

To protect against human immunodeficiency virus (HIV-1) infection, broadly neutralizing antibodies (bnAbs) must be active at the portals of viral entry in the gastrointestinal or cervicovaginal tracts. The localization and persistence of antibodies at these sites is influenced by the neonatal Fc receptor (FcRn), whose role in protecting against infection in vivo has not been defined. Here, we show that a bnAb with enhanced FcRn binding has increased gut mucosal tissue localization, which improves protection against lentiviral infection in non-human primates. A bnAb directed to the CD4-binding site of the HIV-1 envelope (Env) protein (denoted VRC01) was modified by site-directed mutagenesis to increase its binding affinity for FcRn. This enhanced FcRn-binding mutant bnAb, denoted VRC01-LS, displayed increased transcytosis across human FcRn-expressing cellular monolayers in vitro while retaining FcγRIIIa binding and function, including antibody-dependent cell-mediated cytotoxicity (ADCC) activity, at levels similar to VRC01 (the wild type). VRC01-LS had a threefold longer serum half-life than VRC01 in non-human primates and persisted in the rectal mucosa even when it was no longer detectable in the serum. Notably, VRC01-LS mediated protection superior to that afforded by VRC01 against intrarectal infection with simian-human immunodeficiency virus (SHIV). These findings suggest that modification of FcRn binding provides a mechanism not only to increase serum half-life but also to enhance mucosal localization that confers immune protection. Mutations that enhance FcRn function could therefore increase the potency and durability of passive immunization strategies to prevent HIV-1 infection.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Antígenos de Histocompatibilidad Clase I/inmunología , Receptores Fc/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Administración Rectal , Animales , Anticuerpos Neutralizantes/análisis , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/genética , Afinidad de Anticuerpos/genética , Afinidad de Anticuerpos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Sitios de Unión/genética , Antígenos CD4/metabolismo , Femenino , VIH/química , VIH/inmunología , Anticuerpos Anti-VIH/análisis , Anticuerpos Anti-VIH/sangre , Anticuerpos Anti-VIH/genética , Anticuerpos Anti-VIH/inmunología , Proteínas gp160 de Envoltorio del VIH/química , Proteínas gp160 de Envoltorio del VIH/inmunología , Semivida , Inmunidad Mucosa/inmunología , Inmunización Pasiva , Mucosa Intestinal/inmunología , Macaca mulatta , Masculino , Ratones , Mutagénesis Sitio-Dirigida , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Recto/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Transcitosis
6.
Nature ; 511(7511): 601-5, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25043006

RESUMEN

Inflammation in HIV infection is predictive of non-AIDS morbidity and death, higher set point plasma virus load and virus acquisition; thus, therapeutic agents are in development to reduce its causes and consequences. However, inflammation may simultaneously confer both detrimental and beneficial effects. This dichotomy is particularly applicable to type I interferons (IFN-I) which, while contributing to innate control of infection, also provide target cells for the virus during acute infection, impair CD4 T-cell recovery, and are associated with disease progression. Here we manipulated IFN-I signalling in rhesus macaques (Macaca mulatta) during simian immunodeficiency virus (SIV) transmission and acute infection with two complementary in vivo interventions. We show that blockade of the IFN-I receptor caused reduced antiviral gene expression, increased SIV reservoir size and accelerated CD4 T-cell depletion with progression to AIDS despite decreased T-cell activation. In contrast, IFN-α2a administration initially upregulated expression of antiviral genes and prevented systemic infection. However, continued IFN-α2a treatment induced IFN-I desensitization and decreased antiviral gene expression, enabling infection with increased SIV reservoir size and accelerated CD4 T-cell loss. Thus, the timing of IFN-induced innate responses in acute SIV infection profoundly affects overall disease course and outweighs the detrimental consequences of increased immune activation. Yet, the clinical consequences of manipulation of IFN signalling are difficult to predict in vivo and therapeutic interventions in human studies should be approached with caution.


Asunto(s)
Progresión de la Enfermedad , Interferón-alfa/uso terapéutico , Macaca mulatta/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Interferón-alfa/farmacología , Estimación de Kaplan-Meier , Transducción de Señal/efectos de los fármacos , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control
7.
Nature ; 505(7484): 502-8, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24352234

RESUMEN

A major challenge for the development of a highly effective AIDS vaccine is the identification of mechanisms of protective immunity. To address this question, we used a nonhuman primate challenge model with simian immunodeficiency virus (SIV). We show that antibodies to the SIV envelope are necessary and sufficient to prevent infection. Moreover, sequencing of viruses from breakthrough infections revealed selective pressure against neutralization-sensitive viruses; we identified a two-amino-acid signature that alters antigenicity and confers neutralization resistance. A similar signature confers resistance of human immunodeficiency virus (HIV)-1 to neutralization by monoclonal antibodies against variable regions 1 and 2 (V1V2), suggesting that SIV and HIV share a fundamental mechanism of immune escape from vaccine-elicited or naturally elicited antibodies. These analyses provide insight into the limited efficacy seen in HIV vaccine trials.


Asunto(s)
Vacunas contra el SIDA/inmunología , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , VIH-1/inmunología , Vacunas contra el SIDAS/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/inmunología , Susceptibilidad a Enfermedades/inmunología , Femenino , Efecto Fundador , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH/inmunología , VIH-1/química , Humanos , Evasión Inmune/inmunología , Macaca mulatta , Masculino , Datos de Secuencia Molecular , Filogenia , Riesgo , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/química , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/fisiología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología
8.
Sensors (Basel) ; 20(3)2020 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-32041224

RESUMEN

Warm-season legumes have been receiving increased attention as forage resources in the southern United States and other countries. However, the near infrared spectroscopy (NIRS) technique has not been widely explored for predicting the forage quality of many of these legumes. The objective of this research was to assess the performance of NIRS in predicting the forage quality parameters of five warm-season legumes-guar (Cyamopsis tetragonoloba), tepary bean (Phaseolus acutifolius), pigeon pea (Cajanus cajan), soybean (Glycine max), and mothbean (Vigna aconitifolia)-using three machine learning techniques: partial least square (PLS), support vector machine (SVM), and Gaussian processes (GP). Additionally, the efficacy of global models in predicting forage quality was investigated. A set of 70 forage samples was used to develop species-based models for concentrations of crude protein (CP), acid detergent fiber (ADF), neutral detergent fiber (NDF), and in vitro true digestibility (IVTD) of guar and tepary bean forages, and CP and IVTD in pigeon pea and soybean. All species-based models were tested through 10-fold cross-validations, followed by external validations using 20 samples of each species. The global models for CP and IVTD of warm-season legumes were developed using a set of 150 random samples, including 30 samples for each of the five species. The global models were tested through 10-fold cross-validation, and external validation using five individual sets of 20 samples each for different legume species. Among techniques, PLS consistently performed best at calibrating (R2c = 0.94-0.98) all forage quality parameters in both species-based and global models. The SVM provided the most accurate predictions for guar and soybean crops, and global models, and both SVM and PLS performed better for tepary bean and pigeon pea forages. The global modeling approach that developed a single model for all five crops yielded sufficient accuracy (R2cv/R2v = 0.92-0.99) in predicting CP of the different legumes. However, the accuracy of predictions of in vitro true digestibility (IVTD) for the different legumes was variable (R2cv/R2v = 0.42-0.98). Machine learning algorithms like SVM could help develop robust NIRS-based models for predicting forage quality with a relatively small number of samples, and thus needs further attention in different NIRS based applications.


Asunto(s)
Alimentación Animal/análisis , Fabaceae/fisiología , Aprendizaje Automático , Estaciones del Año , Espectroscopía Infrarroja Corta , Temperatura , Calibración , Reproducibilidad de los Resultados
9.
Nature ; 499(7456): 102-6, 2013 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-23698367

RESUMEN

Influenza viruses pose a significant threat to the public and are a burden on global health systems. Each year, influenza vaccines must be rapidly produced to match circulating viruses, a process constrained by dated technology and vulnerable to unexpected strains emerging from humans and animal reservoirs. Here we use knowledge of protein structure to design self-assembling nanoparticles that elicit broader and more potent immunity than traditional influenza vaccines. The viral haemagglutinin was genetically fused to ferritin, a protein that naturally forms nanoparticles composed of 24 identical polypeptides. Haemagglutinin was inserted at the interface of adjacent subunits so that it spontaneously assembled and generated eight trimeric viral spikes on its surface. Immunization with this influenza nanoparticle vaccine elicited haemagglutination inhibition antibody titres more than tenfold higher than those from the licensed inactivated vaccine. Furthermore, it elicited neutralizing antibodies to two highly conserved vulnerable haemagglutinin structures that are targets of universal vaccines: the stem and the receptor binding site on the head. Antibodies elicited by a 1999 haemagglutinin-nanoparticle vaccine neutralized H1N1 viruses from 1934 to 2007 and protected ferrets from an unmatched 2007 H1N1 virus challenge. This structure-based, self-assembling synthetic nanoparticle vaccine improves the potency and breadth of influenza virus immunity, and it provides a foundation for building broader vaccine protection against emerging influenza viruses and other pathogens.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/química , Vacunas contra la Influenza/inmunología , Nanopartículas/química , Animales , Sitios de Unión , Reacciones Cruzadas/inmunología , Femenino , Hurones/inmunología , Hurones/virología , Ferritinas/química , Pruebas de Inhibición de Hemaglutinación , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H1N1 del Virus de la Influenza A/clasificación , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Vacunas de Productos Inactivados/inmunología
10.
PLoS Pathog ; 12(3): e1005486, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26943673

RESUMEN

CD8 T cells are involved in pathogen clearance and infection-induced pathology in respiratory syncytial virus (RSV) infection. Studying bulk responses masks the contribution of individual CD8 T cell subsets to protective immunity and immunopathology. In particular, the roles of subdominant responses that are potentially beneficial to the host are rarely appreciated when the focus is on magnitude instead of quality of response. Here, by evaluating CD8 T cell responses in CB6F1 hybrid mice, in which multiple epitopes are recognized, we found that a numerically subdominant CD8 T cell response against DbM187 epitope of the virus matrix protein expressed high avidity TCR and enhanced signaling pathways associated with CD8 T cell effector functions. Each DbM187 T effector cell lysed more infected targets on a per cell basis than the numerically dominant KdM282 T cells, and controlled virus replication more efficiently with less pulmonary inflammation and illness than the previously well-characterized KdM282 T cell response. Our data suggest that the clinical outcome of viral infections is determined by the integrated functional properties of a variety of responding CD8 T cells, and that the highest magnitude response may not necessarily be the best in terms of benefit to the host. Understanding how to induce highly efficient and functional T cells would inform strategies for designing vaccines intended to provide T cell-mediated immunity.


Asunto(s)
Inmunidad Celular , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Supervivencia Celular , Femenino , Ratones , Organismos Libres de Patógenos Específicos , Subgrupos de Linfocitos T/inmunología , Carga Viral , Replicación Viral
11.
J Virol ; 90(3): 1321-32, 2016 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26581981

RESUMEN

UNLABELLED: Combination antiretroviral therapy (cART) administered shortly after human immunodeficiency virus type 1 (HIV-1) infection can suppress viremia and limit seeding of the viral reservoir, but lifelong treatment is required for the majority of patients. Highly potent broadly neutralizing HIV-1 monoclonal antibodies (MAbs) can reduce plasma viremia when administered during chronic HIV-1 infection, but the therapeutic potential of these antibodies during acute infection is unknown. We tested the ability of HIV-1 envelope glycoprotein-specific broadly neutralizing MAbs to suppress acute simian-human immunodeficiency virus (SHIV) replication in rhesus macaques. Four groups of macaques were infected with SHIV-SF162P3 and received (i) the CD4-binding-site MAb VRC01; (ii) a combination of a more potent clonal relative of VRC01 (VRC07-523) and a V3 glycan-dependent MAb (PGT121); (iii) daily cART, all on day 10, just prior to expected peak plasma viremia; or (iv) no treatment. Daily cART was initiated 11 days after MAb administration and was continued for 13 weeks in all treated animals. Over a period of 11 days after a single administration, MAb treatment significantly reduced peak viremia, accelerated the decay slope, and reduced total viral replication compared to untreated controls. Proviral DNA in lymph node CD4 T cells was also diminished after treatment with the dual MAb. These data demonstrate the virological effect of potent MAbs and support future clinical trials that investigate HIV-1-neutralizing MAbs as adjunctive therapy with cART during acute HIV-1 infection. IMPORTANCE: Treatment of chronic HIV-1 infection with potent broadly neutralizing HIV-1 MAbs has been shown to significantly reduce plasma viremia. However, the antiviral effect of MAb treatment during acute HIV-1 infection is unknown. Here, we demonstrate that MAbs targeting the HIV-1 envelope glycoprotein both suppress acute SHIV plasma viremia and limit CD4 T cell-associated viral DNA. These findings provide support for clinical trials of MAbs as adjunctive therapy with antiretroviral therapy during acute HIV-1 infection.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Anti-VIH/administración & dosificación , VIH-1/inmunología , Inmunización Pasiva , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Linfocitos T CD4-Positivos/virología , ADN Viral/análisis , Anticuerpos Anti-VIH/inmunología , Humanos , Macaca mulatta , Provirus/genética , Viremia/prevención & control
12.
J Virol ; 89(11): 5895-903, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25787288

RESUMEN

UNLABELLED: Pathogen-specific neutralizing antibodies protect against many viral infections and can potentially prevent human immunodeficiency virus (HIV) transmission in humans. However, neutralizing antibodies have so far only been shown to protect nonhuman primates (NHP) against lentiviral infection when given shortly before challenge. Thus, the clinical utility and feasibility of passive antibody transfer to confer long-term protection against HIV-1 are still debated. Here, we investigate the potential of a broadly neutralizing HIV-1 antibody to provide long-term protection in a NHP model of HIV-1 infection. A human antibody was simianized to avoid immune rejection and used to sustain therapeutic levels for ∼5 months. Two months after the final antibody administration, animals were completely protected against viral challenge. These findings demonstrate the feasibility and potential of long-term passive antibody for protection against HIV-1 in humans and provide a model to test antibody therapies for other diseases in NHP. IMPORTANCE: Antibodies against HIV are potential drugs that may be able to prevent HIV infection in humans. However, the long-term protective capacity of antibodies against HIV has not been assessed. Here, we repetitively administered a macaque version of a human anti-HIV antibody to monkeys, after which the antibody persisted in the blood for >5 months. Moreover, the antibody could be sustained at protective levels for 108 days, conferring protection 52 days after the last dose in a monkey model of HIV infection. Thus, passive antibody transfer can provide durable protection against infection by viruses that cause AIDS in primates.


Asunto(s)
Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Antivirales/administración & dosificación , VIH-1/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Inmunización Pasiva , Macaca , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Resultado del Tratamiento
13.
J Virol ; 89(16): 8334-45, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26041300

RESUMEN

Broadly neutralizing antibodies (bnAbs) can prevent lentiviral infection in nonhuman primates and may slow the spread of human immunodeficiency virus type 1 (HIV-1). Although protection by passive transfer of human bnAbs has been demonstrated in monkeys, durable expression is essential for its broader use in humans. Gene-based expression of bnAbs provides a potential solution to this problem, although immune responses to the viral vector or to the antibody may limit its durability and efficacy. Here, we delivered an adeno-associated viral vector encoding a simianized form of a CD4bs bnAb, VRC07, and evaluated its immunogenicity and protective efficacy. The expressed antibody circulated in macaques for 16 weeks at levels up to 66 g/ml, although immune suppression with cyclosporine (CsA) was needed to sustain expression. Gene-delivered simian VRC07 protected against simian-human immunodeficiency virus (SHIV) infection in monkeys 5.5 weeks after treatment. Gene transfer of an anti-HIV antibody can therefore protect against infection by viruses that cause AIDS in primates when the host immune responses are controlled.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH/prevención & control , VIH-1/inmunología , Inmunoglobulina G/genética , Modelos Moleculares , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/genética , Ciclosporina/farmacología , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Anticuerpos Anti-VIH/genética , Humanos , Inmunosupresores/farmacología , Macaca mulatta , Datos de Secuencia Molecular , Pruebas de Neutralización , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
J Immunol ; 193(4): 1799-811, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25024382

RESUMEN

Development of a vaccine against pulmonary tuberculosis may require immunization strategies that induce a high frequency of Ag-specific CD4 and CD8 T cells in the lung. The nonhuman primate model is essential for testing such approaches because it has predictive value for how vaccines elicit responses in humans. In this study, we used an aerosol vaccination strategy to administer AERAS-402, a replication-defective recombinant adenovirus (rAd) type 35 expressing Mycobacterium tuberculosis Ags Ag85A, Ag85B, and TB10.4, in bacillus Calmette-Guérin (BCG)-primed or unprimed rhesus macaques. Immunization with BCG generated low purified protein derivative-specific CD4 T cell responses in blood and bronchoalveolar lavage. In contrast, aerosolized AERAS-402 alone or following BCG induced potent and stable Ag85A/b-specific CD4 and CD8 effector T cells in bronchoalveolar lavage that largely produced IFN-γ, as well as TNF and IL-2. Such responses induced by BCG, AERAS-402, or both failed to confer overall protection following challenge with 275 CFUs M. tuberculosis Erdman, although vaccine-induced responses associated with reduced pathology were observed in some animals. Anamnestic T cell responses to Ag85A/b were not detected in blood of immunized animals after challenge. Overall, our data suggest that a high M. tuberculosis challenge dose may be a critical factor in limiting vaccine efficacy in this model. However, the ability of aerosol rAd immunization to generate potent cellular immunity in the lung suggests that using different or more immunogens, alternative rAd serotypes with enhanced immunogenicity, and a physiological challenge dose may achieve protection against M. tuberculosis.


Asunto(s)
Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis Pulmonar/prevención & control , Vacunación/métodos , Vacunas Sintéticas/inmunología , Aciltransferasas/inmunología , Administración por Inhalación , Animales , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Pulmón/inmunología , Pulmón/microbiología , Macaca mulatta , Masculino , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/virología , Vacunas contra la Tuberculosis/administración & dosificación , Tuberculosis Pulmonar/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Vacunas de ADN , Vacunas Sintéticas/administración & dosificación
15.
J Virol ; 88(21): 12669-82, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25142607

RESUMEN

UNLABELLED: Over the past 5 years, a new generation of highly potent and broadly neutralizing HIV-1 antibodies has been identified. These antibodies can protect against lentiviral infection in nonhuman primates (NHPs), suggesting that passive antibody transfer would prevent HIV-1 transmission in humans. To increase the protective efficacy of such monoclonal antibodies, we employed next-generation sequencing, computational bioinformatics, and structure-guided design to enhance the neutralization potency and breadth of VRC01, an antibody that targets the CD4 binding site of the HIV-1 envelope. One variant, VRC07-523, was 5- to 8-fold more potent than VRC01, neutralized 96% of viruses tested, and displayed minimal autoreactivity. To compare its protective efficacy to that of VRC01 in vivo, we performed a series of simian-human immunodeficiency virus (SHIV) challenge experiments in nonhuman primates and calculated the doses of VRC07-523 and VRC01 that provide 50% protection (EC50). VRC07-523 prevented infection in NHPs at a 5-fold lower concentration than VRC01. These results suggest that increased neutralization potency in vitro correlates with improved protection against infection in vivo, documenting the improved functional efficacy of VRC07-523 and its potential clinical relevance for protecting against HIV-1 infection in humans. IMPORTANCE: In the absence of an effective HIV-1 vaccine, alternative strategies are needed to block HIV-1 transmission. Direct administration of HIV-1-neutralizing antibodies may be able to prevent HIV-1 infections in humans. This approach could be especially useful in individuals at high risk for contracting HIV-1 and could be used together with antiretroviral drugs to prevent infection. To optimize the chance of success, such antibodies can be modified to improve their potency, breadth, and in vivo half-life. Here, knowledge of the structure of a potent neutralizing antibody, VRC01, that targets the CD4-binding site of the HIV-1 envelope protein was used to engineer a next-generation antibody with 5- to 8-fold increased potency in vitro. When administered to nonhuman primates, this antibody conferred protection at a 5-fold lower concentration than the original antibody. Our studies demonstrate an important correlation between in vitro assays used to evaluate the therapeutic potential of antibodies and their in vivo effectiveness.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Inmunización Pasiva/métodos , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Animales , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/genética , Anticuerpos Anti-VIH/administración & dosificación , Anticuerpos Anti-VIH/genética , VIH-1/genética , Macaca mulatta , Masculino , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
16.
J Virol ; 87(17): 9836-44, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23824823

RESUMEN

Programmed Death 1 (PD-1) expression by human/simian immunodeficiency virus (HIV/SIV)-specific CD8 T cells has been associated with defective cytokine production and reduced in vitro proliferation capacity. However, the cellular mechanisms that sustain PD-1(high) virus-specific CD8 T cell responses during chronic infection are unknown. Here, we show that the PD-1(high) phenotype is associated with accelerated in vivo CD8 T cell turnover in SIV-infected rhesus macaques, especially within the SIV-specific CD8 T cell pool. Mathematical modeling of 5-bromo-2' deoxyuridine (BrdU) labeling dynamics demonstrated a significantly increased generation rate of PD-1(high) compared to PD-1(low) CD8 T cells in all memory compartments. Simultaneous analysis of Ki67 and BrdU kinetics revealed a complex in vivo turnover profile whereby only a small fraction of PD-1(high) cells, but virtually all PD-1(low) cells, returned to rest after activation. Similar kinetics operated in both chronic and acute SIV infection. Our data suggest that the persistence of PD-1(high) SIV-specific CD8 T cells in chronic infection is maintained in vivo by a mechanism involving high production coupled with a high disappearance rate.


Asunto(s)
Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , Receptor de Muerte Celular Programada 1/metabolismo , Virus de la Inmunodeficiencia de los Simios/inmunología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Enfermedad Aguda , Animales , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Enfermedad Crónica , Citocinas/biosíntesis , Memoria Inmunológica , Activación de Linfocitos , Macaca mulatta , Fase de Descanso del Ciclo Celular , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Subgrupos de Linfocitos T/virología
17.
J Virol ; 86(13): 7052-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22532675

RESUMEN

A major goal of AIDS vaccine development is to design vaccination strategies that can elicit broad and potent protective antibodies. The initial viral targets of neutralizing antibodies (NAbs) early after human or simian immunodeficiency virus (HIV/SIV) infection are not known. The identification of early NAb epitopes that induce protective immunity or retard the progression of disease is important for AIDS vaccine development. The aim of this study was to determine the Env residues targeted by early SIV NAbs and to assess the influence of prior vaccination on neutralizing antibody kinetics and specificity during early infection. We previously described stereotypic env sequence variations in SIVmac251-infected rhesus monkeys that resulted in viral escape from NAbs. Here, we defined the early viral targets of neutralization and determined whether the ability of serum antibody from infected monkeys to neutralize SIV was altered in the setting of prior vaccination. To localize the viral determinants recognized by early NAbs, a panel of mutant pseudoviruses was assessed in a TZM-bl reporter gene neutralization assay to define the precise changes that eliminate recognition by SIV Env-specific NAbs in 16 rhesus monkeys. Changing R420 to G or R424 to Q in V4 of Env resulted in the loss of recognition by NAbs in vaccinated monkeys. In contrast, mutations in the V1 region of Env did not alter the NAb profile. These findings indicate that early NAbs are directed toward SIVmac251 Env V4 but not the V1 region, and that this env vaccination regimen did not alter the kinetics or the breadth of NAbs during early infection.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Epítopos de Linfocito B/inmunología , Productos del Gen env/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Epítopos de Linfocito B/genética , Productos del Gen env/genética , Macaca mulatta , Masculino , Proteínas Mutantes/genética , Proteínas Mutantes/inmunología , Pruebas de Neutralización , Virus de la Inmunodeficiencia de los Simios/genética
18.
J Virol ; 86(10): 5877-84, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22419810

RESUMEN

The goal of an effective AIDS vaccine is to generate immunity that will prevent human immunodeficiency virus 1 (HIV-1) acquisition. Despite limited progress toward this goal, renewed optimism has followed the recent success of the RV144 vaccine trial in Thailand. However, the lack of complete protection in this trial suggests that breakthroughs, where infection occurs despite adequate vaccination, will be a reality for many vaccine candidates. We previously reported that neutralizing antibodies elicited by DNA prime-recombinant adenovirus serotype 5 (rAd5) boost vaccination with simian immunodeficiency virus strain mac239 (SIVmac239) Gag-Pol and Env provided protection against pathogenic SIVsmE660 acquisition after repeated mucosal challenge. Here, we report that SIV-specific CD8(+) T cells elicited by that vaccine lowered both peak and set-point viral loads in macaques that became infected despite vaccination. These SIV-specific CD8(+) T cells showed strong virus-inhibitory activity (VIA) and displayed an effector memory (EM) phenotype. VIA correlated with high levels of CD107a mobilization and perforin expression in SIV-specific CD8(+) T cells. Remarkably, both the frequency and the number of Gag CM9-specific public clonotypes were strongly correlated with VIA mediated by EM CD8(+) T cells. The ability to elicit such virus-specific EM CD8(+) T cells might contribute substantially to an efficacious HIV/AIDS vaccine, even after breakthrough infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/inmunología , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/fisiología , Carga Viral , Animales , Linfocitos T CD4-Positivos/virología , Regulación hacia Abajo , Infecciones por VIH/genética , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , VIH-1/genética , VIH-1/inmunología , VIH-1/fisiología , Humanos , Macaca mulatta , Vacunas contra el SIDAS/administración & dosificación , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunación
19.
J Virol ; 86(15): 7760-70, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22593152

RESUMEN

The RV144 trial demonstrated that an experimental AIDS vaccine can prevent human immunodeficiency virus type 1 (HIV-1) infection in humans. Because of its limited efficacy, further understanding of the mechanisms of preventive AIDS vaccines remains a priority, and nonhuman primate (NHP) models of lentiviral infection provide an opportunity to define immunogens, vectors, and correlates of immunity. In this study, we show that prime-boost vaccination with a mismatched SIV envelope (Env) gene, derived from simian immunodeficiency virus SIVmac239, prevents infection by SIVsmE660 intrarectally. Analysis of different gene-based prime-boost immunization regimens revealed that recombinant adenovirus type 5 (rAd5) prime followed by replication-defective lymphocytic choriomeningitis virus (rLCMV) boost elicited robust CD4 and CD8 T-cell and humoral immune responses. This vaccine protected against infection after repetitive mucosal challenge with efficacies of 82% per exposure and 62% cumulatively. No effect was seen on viremia in infected vaccinated monkeys compared to controls. Protection correlated with the presence of neutralizing antibodies to the challenge viruses tested in peripheral blood mononuclear cells. These data indicate that a vaccine expressing a mismatched Env gene alone can prevent SIV infection in NHPs and identifies an immune correlate that may guide immunogen selection and immune monitoring for clinical efficacy trials.


Asunto(s)
Adenoviridae , Productos del Gen env/inmunología , Virus de la Coriomeningitis Linfocítica , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas de ADN/inmunología , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Animales , Femenino , Productos del Gen env/genética , VIH-1/genética , VIH-1/inmunología , Inmunización Secundaria/métodos , Macaca mulatta , Masculino , Ratones , Vacunas contra el SIDAS/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Transducción Genética , Vacunas de ADN/genética
20.
PLoS Pathog ; 7(2): e1001293, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21379569

RESUMEN

We characterized the evolution of simian immunodeficiency virus (SIV) in the male genital tract by examining blood- and semen-associated virus from experimentally and sham vaccinated rhesus monkeys during primary infection. At the time of peak virus replication, SIV sequences were intermixed between the blood and semen supporting a scenario of high-level virus "spillover" into the male genital tract. However, at the time of virus set point, compartmentalization was apparent in 4 of 7 evaluated monkeys, likely as a consequence of restricted virus gene flow between anatomic compartments after the resolution of primary viremia. These findings suggest that SIV replication in the male genital tract evolves to compartmentalization after peak viremia resolves.


Asunto(s)
Productos del Gen env/genética , Genitales Masculinos/virología , Semen/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Animales , ADN Viral/genética , Flujo Génico , Macaca mulatta , Masculino , Reacción en Cadena de la Polimerasa , Síndrome de Inmunodeficiencia Adquirida del Simio/sangre , Virus de la Inmunodeficiencia de los Simios/genética , Vacunación , Carga Viral , Viremia/prevención & control , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA