Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 12(1): 9403, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672343

RESUMEN

Human rabies remains a globally significant public health problem. Replacement of polyclonal anti-rabies immunoglobulin (RIG), a passive component of rabies post-exposure prophylaxis (PEP), with a monoclonal antibody (MAb), would eliminate the cost and availability constraints associated with RIG. Our team has developed and licensed a human monoclonal antibody RAB1 (Rabishield©), as the replacement for RIG where canine rabies is enzootic. However, for the highly diverse rabies viruses of North America, a cocktail containing two or more MAbs targeting different antigenic sites of the rabies glycoprotein should be included to ensure neutralization of all variants of the virus. In this study, two MAb cocktails, R172 (RAB1-RAB2) and R173 (RAB1-CR57), were identified and evaluated against a broad range of rabies variants from North America. R173 was found to be the most potent cocktail, as it neutralized all the tested North American RABV isolates and demonstrated broad coverage of isolates from both terrestrial and bat species. R173 could be a promising candidate as an alternative or replacement for RIG PEP in North America.


Asunto(s)
Antineoplásicos Inmunológicos , Vacunas Antirrábicas , Virus de la Rabia , Rabia , Animales , Anticuerpos Monoclonales , Anticuerpos Antivirales , Perros , Humanos , Profilaxis Posexposición
2.
Virology ; 499: 298-312, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27723488

RESUMEN

For studies on vaccines and therapies for HIV disease, SIV-HIV chimeric viruses harboring the HIV-1 env gene (SHIVenv) remain the best virus in non-human primate models. However, there are still very few SHIVenv viruses that can cause AIDS in non-CD8-depleted animals. In the present study, a recently created CCR5-using SHIVenv_B3 virus with env gene derived from acute/early HIV-1 infections (AHI) successfully established pathogenic infection in macaques. Through a series of investigations on the evolution, mutational profile, and phenotype of the virus and the resultant humoral immune response in infected rhesus macaques, we found that the E32K mutation in the Env C1 domain was associated with macaque pathogenesis, and that the electrostatic interactions in Env may favor E32K at the gp120 N terminus and "lock" the binding to heptad repeat 1 of gp41 in the trimer and produce a SHIVenv with increased fitness and pathogenesis during macaque infections.


Asunto(s)
Productos del Gen env/genética , Infecciones por VIH/virología , VIH-1/genética , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Evolución Molecular , Productos del Gen env/química , Productos del Gen env/inmunología , Proteína gp120 de Envoltorio del VIH/química , Proteína gp120 de Envoltorio del VIH/genética , Proteína gp120 de Envoltorio del VIH/inmunología , Infecciones por VIH/genética , Infecciones por VIH/inmunología , VIH-1/clasificación , VIH-1/inmunología , VIH-1/patogenicidad , Humanos , Inmunidad Humoral , Macaca mulatta , Datos de Secuencia Molecular , Mutación Missense , Filogenia , Receptores CCR5/genética , Receptores CCR5/inmunología , Receptores Virales/genética , Receptores Virales/inmunología , Alineación de Secuencia , Virus de la Inmunodeficiencia de los Simios/clasificación , Virus de la Inmunodeficiencia de los Simios/inmunología , Virulencia
3.
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
4.
Virology ; 428(2): 121-7, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22521913

RESUMEN

To create an HIV-1 vaccine that generates sufficient breadth of immune recognition to protect against the genetically diverse forms of the circulating virus, we have been exploring vaccines based on consensus and mosaic protein designs. Increasing the valency of a mosaic immunogen cocktail increases epitope coverage but with diminishing returns, as increasingly rare epitopes are incorporated into the mosaic proteins. In this study we compared the immunogenicity of 2-valent and 3-valent HIV-1 envelope mosaic immunogens in rhesus monkeys. Immunizations with the 3-valent mosaic immunogens resulted in a modest increase in the breadth of vaccine-elicited T lymphocyte responses compared to the 2-valent mosaic immunogens. However, the 3-valent mosaic immunogens elicited significantly higher neutralizing responses to Tier 1 viruses than the 2-valent mosaic immunogens. These findings underscore the potential utility of polyvalent mosaic immunogens for eliciting both cellular and humoral immune responses to HIV-1.


Asunto(s)
Vacunas contra el SIDA/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Inmunidad Celular , Inmunidad Humoral , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Vacunas contra el SIDA/administración & dosificación , Animales , Modelos Animales de Enfermedad , Epítopos/administración & dosificación , Epítopos/genética , Epítopos/inmunología , Infecciones por VIH/virología , VIH-1/genética , Macaca mulatta , Productos del Gen env del Virus de la Inmunodeficiencia Humana/administración & dosificación , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética
5.
J Virol ; 85(19): 10389-98, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21775457

RESUMEN

There is considerable variability in host susceptibility to human immunodeficiency virus type 1 (HIV-1) infection, but the host genetic determinants of that variability are not well understood. In addition to serving as a block for cross-species retroviral infection, TRIM5 was recently shown to play a central role in limiting primate immunodeficiency virus replication. We hypothesized that TRIM5 may also contribute to susceptibility to mucosal acquisition of simian immunodeficiency virus (SIV) in rhesus monkeys. We explored this hypothesis by establishing 3 cohorts of Indian-origin rhesus monkeys with different TRIM5 genotypes: homozygous restrictive, heterozygous permissive, and homozygous permissive. We then evaluated the effect of TRIM5 genotype on the penile transmission of SIVsmE660. We observed a significant effect of TRIM5 genotype on mucosal SIVsmE660 acquisition in that no SIV transmission occurred in monkeys with only restrictive TRIM5 alleles. In contrast, systemic SIV infections were initiated after preputial pocket exposures in monkeys that had at least one permissive TRIM5 allele. These data demonstrate that host genetic factors can play a critical role in restricting mucosal transmission of a primate immunodeficiency virus. In addition, we used our understanding of TRIM5 to establish a novel nonhuman primate penile transmission model for AIDS mucosal pathogenesis and vaccine research.


Asunto(s)
Proteínas Portadoras/inmunología , Predisposición Genética a la Enfermedad , Membrana Mucosa/inmunología , Membrana Mucosa/virología , Pene/inmunología , Pene/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Proteínas Portadoras/genética , Genotipo , Macaca mulatta , Masculino
6.
Sci Transl Med ; 3(81): 81ra36, 2011 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-21543722

RESUMEN

The RV144 vaccine trial in Thailand demonstrated that an HIV vaccine could prevent infection in humans and highlights the importance of understanding protective immunity against HIV. We used a nonhuman primate model to define immune and genetic mechanisms of protection against mucosal infection by the simian immunodeficiency virus (SIV). A plasmid DNA prime/recombinant adenovirus serotype 5 (rAd5) boost vaccine regimen was evaluated for its ability to protect monkeys from infection by SIVmac251 or SIVsmE660 isolates after repeat intrarectal challenges. Although this prime-boost vaccine regimen failed to protect against SIVmac251 infection, 50% of vaccinated monkeys were protected from infection with SIVsmE660. Among SIVsmE660-infected animals, there was about a one-log reduction in peak plasma virus RNA in monkeys expressing the major histocompatibility complex class I allele Mamu-A*01, implicating cytotoxic T lymphocytes in the control of SIV replication once infection is established. Among Mamu-A*01-negative monkeys challenged with SIVsmE660, no CD8(+) T cell response or innate immune response was associated with protection against virus acquisition. However, low levels of neutralizing antibodies and an envelope-specific CD4(+) T cell response were associated with vaccine protection in these monkeys. Moreover, monkeys that expressed two TRIM5 alleles that restrict SIV replication were more likely to be protected from infection than monkeys that expressed at least one permissive TRIM5 allele. This study begins to elucidate the mechanisms of vaccine protection against immunodeficiency viruses and highlights the need to analyze these immune and genetic correlates of protection in future trials of HIV vaccine strategies.


Asunto(s)
Membrana Mucosa/virología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Alelos , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Proteínas Portadoras/genética , Haplorrinos , Humanos
7.
Virology ; 406(1): 48-55, 2010 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-20667574

RESUMEN

The recently reported modest success of the RV144 Thai trial vaccine regimen in preventing HIV-1 acquisition has focused interest on the potential contribution to that protection of vaccine-elicited CD4(+) T cell responses. We evaluated the induction of virus-specific CD4(+) T cell responses in rhesus monkeys using a series of diverse vaccine vectors. We assessed both the magnitudes and functional profiles of the antigen-specific CD4(+) T cells by measuring cytokine production, memory differentiation, and the expression of mucosal homing molecules. We found that DNA prime/recombinant MVA boost immunizations induced particularly high-frequency virus-specific CD4(+) T cell responses with polyfunctional repertoires, and these responses were partially preserved following SHIV-89.6P challenge. The majority of the vaccine-elicited CD4(+) T cells were CD28(+) memory T cells that expressed low levels of beta7. Neither the magnitudes nor the functional profiles of the virus-specific CD4(+) T cells generated by vaccination were associated with a preservation of CD4(+) T cells or control of viral replication following SHIV-89.6P challenge. Interestingly, monkeys primed with recombinant Ad5 immunogens showed a dramatic expansion of both the magnitude and polyfunctionality of the vaccine-elicited CD4(+) T cell responses following envelope protein boost. These results demonstrate that vaccine strategies that include recombinant MVA or recombinant Ad5 vectors can elicit robust CD4(+) T cell responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , VIH-1/genética , VIH-1/inmunología , Macaca mulatta/inmunología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Citocinas/biosíntesis , Vectores Genéticos , VIH-1/fisiología , Humanos , Inmunidad Mucosa , Inmunización Secundaria , Memoria Inmunológica , Virus de la Inmunodeficiencia de los Simios/fisiología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Replicación Viral
8.
Nat Med ; 16(3): 324-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20173754

RESUMEN

An effective HIV vaccine must elicit immune responses that recognize genetically diverse viruses. It must generate CD8+ T lymphocytes that control HIV replication and CD4+ T lymphocytes that provide help for the generation and maintenance of both cellular and humoral immune responses against the virus. Creating immunogens that can elicit cellular immune responses against the genetically varied circulating isolates of HIV presents a key challenge for creating an HIV vaccine. Polyvalent mosaic immunogens derived by in silico recombination of natural strains of HIV are designed to induce cellular immune responses that recognize genetically diverse circulating virus isolates. Here we immunized rhesus monkeys by plasmid DNA prime and recombinant vaccinia virus boost with vaccine constructs expressing either consensus or polyvalent mosaic proteins. As compared to consensus immunogens, the mosaic immunogens elicited CD8+ T lymphocyte responses to more epitopes of each viral protein than did the consensus immunogens and to more variant sequences of CD8+ T lymphocyte epitopes. This increased breadth and depth of epitope recognition may contribute both to protection against infection by genetically diverse viruses and to the control of variant viruses that emerge as they mutate away from recognition by cytotoxic T lymphocytes.


Asunto(s)
Vacunas contra el SIDA/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/inmunología , VIH/inmunología , Activación de Linfocitos/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Epítopos de Linfocito T/inmunología , Antígenos VIH/inmunología , Inmunidad Celular/inmunología , Recuento de Linfocitos , Macaca mulatta/inmunología , Vacunas Sintéticas/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología
9.
J Virol ; 83(11): 5505-13, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19297477

RESUMEN

While mycobacteria have been proposed as vaccine vectors because of their persistence and safety, little has been done systematically to optimize their immunogenicity in nonhuman primates. We successfully generated recombinant Mycobacterium bovis BCG (rBCG) expressing simian immunodeficiency virus (SIV) Gag and Pol as multigenic, nonintegrating vectors, but rBCG-expressing SIV Env was unstable. A dose and route determination study in rhesus monkeys revealed that intramuscular administration of rBCG was associated with local reactogenicity, whereas intravenous and intradermal administration of 10(6) to 10(8) CFU of rBCG was well tolerated. After single or repeat rBCG inoculations, monkeys developed high-frequency gamma interferon enzyme-linked immunospot responses against BCG purified protein derivative. However, the same animals developed only modest SIV-specific CD8(+) T-cell responses. Nevertheless, high-frequency SIV-specific cellular responses were observed in the rBCG-primed monkeys after boosting with recombinant adenovirus 5 (rAd5) expressing the SIV antigens. These cellular responses were of greater magnitude and more persistent than those generated after vaccination with rAd5 alone. The vaccine-elicited cellular responses were predominantly polyfunctional CD8(+) T cells. These findings support the further exploration of mycobacteria as priming vaccine vectors.


Asunto(s)
Adenoviridae/genética , Vacuna BCG/inmunología , Inmunización Secundaria/métodos , Macaca mulatta/inmunología , Mycobacterium bovis/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Linfocitos T/inmunología , Animales , Vacuna BCG/genética , Vacuna BCG/metabolismo , Células Cultivadas , Productos del Gen env/genética , Productos del Gen env/inmunología , Productos del Gen env/metabolismo , Productos del Gen gag/genética , Productos del Gen gag/inmunología , Productos del Gen gag/metabolismo , Productos del Gen pol/genética , Productos del Gen pol/inmunología , Productos del Gen pol/metabolismo , Vectores Genéticos/genética , Mycobacterium bovis/genética , Mycobacterium bovis/metabolismo , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/metabolismo , Volumetría , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/metabolismo
10.
J Virol ; 83(6): 2686-96, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19129440

RESUMEN

Although there is increasing evidence that individuals already infected with human immunodeficiency virus type 1 (HIV-1) can be infected with a heterologous strain of the virus, the extent of protection against superinfection conferred by the first infection and the biologic consequences of superinfection are not well understood. We explored these questions in the simian immunodeficiency virus (SIV)/rhesus monkey model of HIV-1/AIDS. We infected cohorts of rhesus monkeys with either SIVmac251 or SIVsmE660 and then exposed animals to the reciprocal virus through intrarectal inoculations. Employing a quantitative real-time PCR assay, we determined the replication kinetics of the two strains of virus for 20 weeks. We found that primary infection with a replication-competent virus did not protect against acquisition of infection by a heterologous virus but did confer relative control of the superinfecting virus. In animals that became superinfected, there was a reduction in peak replication and rapid control of the second virus. The relative susceptibility to superinfection was not correlated with CD4(+) T-cell count, CD4(+) memory T-cell subsets, cytokine production by virus-specific CD8(+) or CD4(+) cells, or neutralizing antibodies at the time of exposure to the second virus. Although there were transient increases in viral loads of the primary virus and a modest decline in CD4(+) T-cell counts after superinfection, there was no evidence of disease acceleration. These findings indicate that an immunodeficiency virus infection confers partial protection against a second immunodeficiency virus infection, but this protection may be mediated by mechanisms other than classical adaptive immune responses.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/fisiología , Interferencia Viral , Animales , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Citocinas/biosíntesis , Anticuerpos Anti-VIH/sangre , Memoria Inmunológica , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/crecimiento & desarrollo , Virus de la Inmunodeficiencia de los Simios/inmunología , Carga Viral
11.
J Virol ; 82(23): 11577-88, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18829748

RESUMEN

African green monkeys (AGM) do not develop overt signs of disease following simian immunodeficiency virus (SIV) infection. While it is still unknown how natural hosts like AGM can cope with this lentivirus infection, a large number of investigations have shown that CD8(+) T-cell responses are critical for the containment of AIDS viruses in humans and Asian nonhuman primates. Here we have compared the phenotypes of T-cell subsets and magnitudes of SIV-specific CD8(+) T-cell responses in vervet AGM chronically infected with SIVagm and rhesus monkeys (RM) infected with SIVmac. In comparison to RM, vervet AGM exhibited weaker signs of immune activation and associated proliferation of CD8(+) T cells as detected by granzyme B, Ki-67, and programmed death 1 staining. By gamma interferon enzyme-linked immunospot assay and intracellular cytokine staining, SIV Gag- and Env-specific immune responses were detectable at variable but lower levels in vervet AGM than in RM. These observations demonstrate that natural hosts like SIV-infected vervet AGM develop SIV-specific T-cell responses, but the disease-free course of infection does not depend on the generation of robust CD8(+) T-cell responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/sangre , Chlorocebus aethiops , Enfermedad Crónica , Granzimas/sangre , Interferón gamma/biosíntesis , Antígeno Ki-67/sangre , Activación de Linfocitos , Macaca mulatta , ARN Viral/sangre , Factor de Necrosis Tumoral alfa/biosíntesis
12.
J Virol ; 81(22): 12368-74, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17686853

RESUMEN

Defining the immune correlates of the protection against human immunodeficiency virus type 1 (HIV-1) acquisition in individuals who are exposed to HIV-1 but do not become infected may provide important direction for the creation of an HIV-1 vaccine. We have employed the simian immunodeficiency virus (SIV)/rhesus monkey model to determine whether monkeys can be repeatedly exposed to a primate lentivirus by a mucosal route and escape infection and whether virus-specific immune correlates of protection from infection can be identified in uninfected monkeys. Five of 18 rhesus monkeys exposed 18 times by intrarectal inoculation to SIVmac251 or SIVsmE660 were resistant to infection, indicating that the exposed/uninfected phenotype can be reproduced in a nonhuman primate AIDS model. However, routine peripheral blood lymphocyte gamma interferon enzyme-linked immunospot (ELISPOT), tetramer, and intracellular cytokine staining assays, as well as cytokine-augmented ELISPOT and peptide-stimulated tetramer assays, failed to define a systemic antigen-specific cellular immune correlate to this protection. Further, local cell-mediated immunity could not be demonstrated by tetramer assays of these protected monkeys, and local humoral immunity was not associated with protection against acquisition of virus in another cohort of mucosally exposed monkeys. Therefore, resistance to mucosal infection in these monkeys may not be mediated by adaptive virus-specific immune mechanisms. Rather, innate immune mechanisms or an intact epithelial barrier may be responsible for protection against mucosal infection in this population of monkeys.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/inmunología , Mucosa Intestinal/inmunología , Recto/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Anticuerpos Antivirales/análisis , Modelos Animales de Enfermedad , Humanos , Mucosa Intestinal/virología , Macaca mulatta , Recto/virología , Linfocitos T/inmunología
13.
J Virol ; 81(15): 8009-15, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17522197

RESUMEN

It has long been appreciated that CD4+ T lymphocytes are dysfunctional in human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV)-infected individuals, and it has recently been shown that HIV/SIV infections are associated with a dramatic early destruction of memory CD4+ T lymphocytes. However, the relative contributions of CD4+ T-lymphocyte dysfunction and loss to immune dysregulation during primary HIV/SIV infection have not been fully elucidated. In the current study, we evaluated CD4+ T lymphocytes and their functional repertoire during primary SIVmac251 infection in rhesus monkeys. We show that the extent of loss of memory CD4+ T lymphocytes and staphylococcal enterotoxin B-stimulated cytokine production by total CD4+ T lymphocytes during primary SIVmac251 infection is tightly linked in a cohort of six rhesus monkeys to set point plasma viral RNA levels, with greater loss and dysfunction being associated with higher steady-state viral replication. Moreover, in exploring the mechanism underlying this phenomenon, we demonstrate that the loss of functional CD4+ T lymphocytes during primary SIVmac251 infection is associated with both a selective depletion of memory CD4+ T cells and a loss of the functional capacity of the memory CD4+ T lymphocytes that escape viral destruction.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Memoria Inmunológica/fisiología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Recuento de Linfocito CD4 , Linfocitos T CD8-positivos/metabolismo , Productos del Gen gag/inmunología , VIH/inmunología , Humanos , Macaca mulatta , ARN Viral/sangre , Subgrupos de Linfocitos T/metabolismo
14.
J Virol ; 81(7): 3414-27, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17229693

RESUMEN

We previously demonstrated that replication-competent adenovirus (Ad)-simian immunodeficiency virus (SIV) recombinant prime/protein boost regimens elicit potent immunogenicity and strong, durable protection of rhesus macaques against SIV(mac251). Additionally, native Tat vaccines have conferred strong protection against simian/human immunodeficiency virus SHIV(89.6P) challenge of cynomolgus monkeys, while native, inactivated, or vectored Tat vaccines have failed to elicit similar protective efficacy in rhesus macaques. Here we asked if priming rhesus macaques with replicating Ad-human immunodeficiency virus (HIV) tat and boosting with the Tat protein would elicit protection against SHIV(89.6P). We also evaluated a Tat/Env regimen, adding an Ad-HIV env recombinant and envelope protein boost to test whether envelope antibodies would augment acute-phase protection. Further, expecting cellular immunity to enhance chronic viremia control, we tested a multigenic group: Ad-HIV tat, -HIV env, -SIV gag, and -SIV nef recombinants and Tat, Env, and Nef proteins. All regimens were immunogenic. A hierarchy was observed in enzyme-linked immunospot responses (with the strongest response for Env, followed by Gag, followed by Nef, followed by Tat) and antibody titers (with the highest titer for Env, followed by Tat, followed by Nef, followed by Gag). Following intravenous SHIV(89.6P) challenge, all macaques became infected. Compared to controls, no protection was seen in the Tat-only group, confirming previous reports for rhesus macaques. However, the multigenic group blunted acute viremia by approximately 1 log (P = 0.017), and both the multigenic and Tat/Env groups reduced chronic viremia by 3 and 4 logs, respectively, compared to controls (multigenic, P = 0.0003; Tat/Env, P < 0.0001). The strikingly greater reduction in the Tat/Env group than in the multigenic group (P = 0.014) was correlated with Tat and Env binding antibodies. Since prechallenge anti-Env antibodies lacked SHIV(89.6P)-neutralizing activity, other functional anti-Env and anti-Tat activities are under investigation, as is a possible synergy between the Tat and Env immunogens.


Asunto(s)
Adenoviridae/genética , Productos del Gen env/metabolismo , Productos del Gen tat/metabolismo , VIH/inmunología , VIH/metabolismo , Virus de la Inmunodeficiencia de los Simios/fisiología , Replicación Viral , Animales , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/farmacología , Productos del Gen env/genética , Productos del Gen env/inmunología , Productos del Gen tat/genética , Productos del Gen tat/inmunología , VIH/genética , Memoria Inmunológica/inmunología , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Macaca mulatta , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
15.
J Infect Dis ; 196(12): 1784-93, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18190259

RESUMEN

Understanding the impact of human immunodeficiency virus (HIV) infection on the clinical manifestations and kinetics of measles virus (MV) replication in MV-vaccinated and unvaccinated individuals is important for developing successful vaccine strategies for measles eradication. To model the pathogenesis of MV infection in MV-vaccinated and unvaccinated individuals infected with HIV, previously vaccinated and unvaccinated rhesus monkeys infected with simian immunodeficiency virus (SIV) were challenged with MV and monitored for clinical, virologic, and immunologic sequelae of infection. The magnitude and duration of MV viremia were unchanged by SIV infection. Nevertheless, clinical manifestations of MV infection were altered in animals with significant CD4(+) T lymphocyte loss. Importantly, 2 of the 3 SIV-infected monkeys with high titers of vaccine-induced MV-neutralizing antibody developed clinical evidence of MV infection. Thus, in this experimental animal model, a high-titer vaccine-induced MV-neutralizing antibody response does not protect against clinical manifestations of measles in the setting of a chronic acquired immunodeficiency syndrome virus infection.


Asunto(s)
Vacuna Antisarampión/inmunología , Virus del Sarampión/inmunología , Sarampión/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Linfocitos T CD4-Positivos/inmunología , Macaca mulatta , Sarampión/virología , Virus del Sarampión/fisiología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/fisiología , Carga Viral , Replicación Viral
16.
J Virol ; 80(22): 10950-6, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16943292

RESUMEN

Understanding the characteristics of the virus-specific T-lymphocyte response that will confer optimal protection against the clinical progression of AIDS will inform the development of an effective cellular immunity-based human immunodeficiency virus vaccine. We have recently shown that survival in plasmid DNA-primed/recombinant adenovirus-boosted rhesus monkeys that are challenged with the simian immunodeficiency virus SIVmac251 is associated with the preservation postchallenge of central memory CD4(+) T lymphocytes and robust gamma interferon (IFN-gamma)-producing SIV-specific CD8(+) and CD4(+) T-lymphocyte responses. The present studies were initiated to extend these observations to determine which virus-specific T-lymphocyte subpopulations play a primary role in controlling disease progression and to characterize the functional repertoire of these cells. We show that the preservation of the SIV-specific central memory CD8(+) T-lymphocyte population and a linked SIV-specific CD4(+) T-lymphocyte response are associated with prolonged survival in vaccinated monkeys following challenge. Furthermore, we demonstrate that SIV-specific IFN-gamma-, tumor necrosis factor alpha-, and interleukin-2-producing T lymphocytes are all comparably associated with protection against disease progression. These findings underscore the contribution of virus-specific central memory T lymphocytes to controlling clinical progression in vaccinated individuals following a primate immunodeficiency virus infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Subgrupos Linfocitarios/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Animales , Inmunidad Celular , Memoria Inmunológica , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/mortalidad , Virus de la Inmunodeficiencia de los Simios/inmunología , Estadística como Asunto , Factor de Necrosis Tumoral alfa/biosíntesis
17.
Science ; 312(5779): 1530-3, 2006 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-16763152

RESUMEN

Vaccine-induced cellular immunity controls virus replication in simian immunodeficiency virus (SIV)-infected monkeys only transiently, leading to the question of whether such vaccines for AIDS will be effective. We immunized monkeys with plasmid DNA and replication-defective adenoviral vectors encoding SIV proteins and then challenged them with pathogenic SIV. Although these monkeys demonstrated a reduction in viremia restricted to the early phase of SIV infection, they showed a prolonged survival. This survival was associated with preserved central memory CD4+ T lymphocytes and could be predicted by the magnitude of the vaccine-induced cellular immune response. These immune correlates of vaccine efficacy should guide the evaluation of AIDS vaccines in humans.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas de ADN/inmunología , Secuencia de Aminoácidos , Animales , Humanos , Macaca mulatta , Datos de Secuencia Molecular , Plásmidos , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Análisis de Supervivencia , Vacunas Sintéticas/inmunología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...