Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Genome Res ; 25(12): 1921-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26377836

ABSTRACT

We describe a genome reference of the African green monkey or vervet (Chlorocebus aethiops). This member of the Old World monkey (OWM) superfamily is uniquely valuable for genetic investigations of simian immunodeficiency virus (SIV), for which it is the most abundant natural host species, and of a wide range of health-related phenotypes assessed in Caribbean vervets (C. a. sabaeus), whose numbers have expanded dramatically since Europeans introduced small numbers of their ancestors from West Africa during the colonial era. We use the reference to characterize the genomic relationship between vervets and other primates, the intra-generic phylogeny of vervet subspecies, and genome-wide structural variations of a pedigreed C. a. sabaeus population. Through comparative analyses with human and rhesus macaque, we characterize at high resolution the unique chromosomal fission events that differentiate the vervets and their close relatives from most other catarrhine primates, in whom karyotype is highly conserved. We also provide a summary of transposable elements and contrast these with the rhesus macaque and human. Analysis of sequenced genomes representing each of the main vervet subspecies supports previously hypothesized relationships between these populations, which range across most of sub-Saharan Africa, while uncovering high levels of genetic diversity within each. Sequence-based analyses of major histocompatibility complex (MHC) polymorphisms reveal extremely low diversity in Caribbean C. a. sabaeus vervets, compared to vervets from putatively ancestral West African regions. In the C. a. sabaeus research population, we discover the first structural variations that are, in some cases, predicted to have a deleterious effect; future studies will determine the phenotypic impact of these variations.


Subject(s)
Chlorocebus aethiops/genetics , Genome , Genomics , Animals , Chlorocebus aethiops/classification , Chromosome Painting , Computational Biology/methods , Evolution, Molecular , Gene Rearrangement , Genetic Variation , Genomics/methods , Karyotype , Major Histocompatibility Complex/genetics , Molecular Sequence Annotation , Phylogeny , Phylogeography
2.
J Virol ; 87(9): 5305-10, 2013 May.
Article in English | MEDLINE | ID: mdl-23449795

ABSTRACT

Here, we demonstrate that KIR2DL4 copy number variation (CNV) is associated with CD4(+) T-cell decline and functionality of cytokine-producing NK cells during primary simian immunodeficiency virus (SIV) infection in Mamu-A*01(-) Indian-origin rhesus macaques, with higher KIR2DL4 copy numbers being associated with a better preservation of CD4(+) T cells and an increased gamma interferon (IFN-γ) production from stimulated cytokine-producing NK cell subsets during acute SIVmac251 infection. These findings underscore the crucial role of activating killer-cell immunoglobulin-like receptors (KIRs) in NK cell-mediated SIV responses during early SIV infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Receptors, KIR2DL4/genetics , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/physiology , Animals , DNA Copy Number Variations , Disease Models, Animal , HIV Infections , HIV-1/immunology , HIV-1/physiology , Humans , Lymphocyte Depletion , Macaca mulatta , Receptors, KIR2DL4/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology
3.
Antimicrob Agents Chemother ; 57(2): 679-88, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23147734

ABSTRACT

Peptides derived from the C-terminal heptad repeat 2 (HR2) region of the HIV-1 gp41 envelope glycoprotein, so-called C peptides, are very efficient HIV-1 fusion inhibitors. We previously developed innovative gene therapeutic approaches aiming at the direct in vivo production of C peptides from genetically modified host cells and found that T cells expressing membrane-anchored or secreted C peptides are protected from HIV-1 infection. However, an unwanted immune response against such antiviral peptides may significantly impair clinical efficacy and pose safety risks to patients. To overcome this problem, we engineered a novel C peptide, V2o, with greatly reduced immunogenicity and excellent antiviral activity. V2o is based on the chimeric C peptide C46-EHO, which is derived from the HR2 regions of HIV-2(EHO) and HIV-1(HxB2) and has broad anti-HIV and anti-simian immunodeficiency virus activity. Antibody and major histocompatibility complex class I epitopes within the C46-EHO peptide sequence were identified by in silico and in vitro analyses. Using rational design, we removed these epitopes by amino acid substitutions and thus minimized antigenicity and immunogenicity considerably. At the same time, the antiviral activity of the deimmunized peptide V2o was preserved or even enhanced compared to that of the parental C46-EHO peptide. Thus, V2o is an ideal candidate, especially for those novel therapeutic approaches for HIV infection that involve direct in vivo production of antiviral C peptides.


Subject(s)
HIV Envelope Protein gp41/chemistry , HIV Fusion Inhibitors/immunology , Peptide Fragments/immunology , Amino Acid Substitution , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line , Computer Simulation , Epitope Mapping , Epitopes/immunology , HEK293 Cells , HIV Antibodies/blood , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/immunology , HIV Fusion Inhibitors/chemistry , HIV Fusion Inhibitors/pharmacology , HIV Infections/immunology , HIV-1/genetics , Histocompatibility Antigens Class I/immunology , Humans , Interferon-gamma , Macaca mulatta , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Protein Engineering , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus , T-Lymphocytes, Cytotoxic/immunology
4.
J Virol ; 86(23): 13103-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23015705

ABSTRACT

Here we show that the number of activating killer cell immunoglobulin-like receptor (KIR) copies in rhesus monkeys is associated with the extent of release of cytotoxic granules by cytolytic NK cells during primary simian immunodeficiency virus SIVmac251 infection. These findings suggest that NK cells expressing high levels of activating KIRs efficiently kill SIVmac251-infected cells, and this efficient killing contributes to the NK cell-mediated control of replication of this virus during early infection.


Subject(s)
Gene Dosage/genetics , Granzymes/immunology , Killer Cells, Natural/immunology , Macaca mulatta , Receptors, KIR/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Animals , Granzymes/metabolism , Killer Cells, Natural/metabolism , Simian Acquired Immunodeficiency Syndrome/genetics , Virus Replication/immunology
5.
J Virol ; 86(11): 6189-96, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22457527

ABSTRACT

Eliciting neutralizing antibodies is thought to be a key activity of a vaccine against human immunodeficiency virus (HIV). However, a number of studies have suggested that in addition to neutralization, interaction of IgG with Fc gamma receptors (FcγR) may play an important role in antibody-mediated protection. We have previously obtained evidence that the protective activity of the broadly neutralizing human IgG1 anti-HIV monoclonal antibody (MAb) b12 in macaques is diminished in the absence of FcγR binding capacity. To investigate antibody-dependent cellular cytotoxicity (ADCC) as a contributor to FcγR-associated protection, we developed a nonfucosylated variant of b12 (NFb12). We showed that, compared to fully fucosylated (referred to as wild-type in the text) b12, NFb12 had higher affinity for human and rhesus macaque FcγRIIIa and was more efficient in inhibiting viral replication and more effective in killing HIV-infected cells in an ADCC assay. Despite these more potent in vitro antiviral activities, NFb12 did not enhance protection in vivo against repeated low-dose vaginal challenge in the simian-human immunodeficiency virus (SHIV)/macaque model compared to wild-type b12. No difference in protection, viral load, or infection susceptibility was observed between animals given NFb12 and those given fully fucosylated b12, indicating that FcγR-mediated activities distinct from FcγRIIIa-mediated ADCC may be important in the observed protection against SHIV challenge.


Subject(s)
Antibodies, Monoclonal/administration & dosage , HIV Antibodies/administration & dosage , HIV-1/immunology , Receptors, IgG/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/metabolism , Cells, Cultured , Female , HIV Antibodies/immunology , HIV Antibodies/isolation & purification , HIV Antibodies/metabolism , Humans , Macaca mulatta , Receptors, IgG/metabolism , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity , Viral Load
6.
Clin Vaccine Immunol ; 18(11): 1969-78, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21900534

ABSTRACT

Vaccination for eliciting antigen-specific memory CD8(+) T cells may be facilitated by manipulating the pleiotropic effects of gamma interferon (IFN-γ). We assessed strategies for modulating the contribution of IFN-γ during the development of antigen-specific cytotoxic T lymphocyte (CTL) populations. We first showed that recombinant IFN-γ suppressed antigen expression in vitro from a recombinant adenovirus (rAd) vector in a dose-dependent manner and that addition of an anti-IFN-γ antibody (Ab) eliminated this suppression. Consistent with these in vitro findings, we found that HIV-1 envelope (Env)-specific CTL responses were higher in IFN-γ-knockout (GKO) mice than in wild-type mice following immunization with rAd. Since these observations suggested that IFN-γ might suppress rAd-induced CTL development, we assessed the ability of anti-IFN-γ Ab administration to augment rAd-elicited CTL in vivo. In fact, blockage of IFN-γ activity by monoclonal Ab administration was associated with elevated levels of interleukin 7 receptor alpha chain-positive (IL-7Rα(+)) Env-specific CTL populations postboost. These observations illustrate the utility of an anti-IFN-γ Ab for potentiating rAd immunizations to effect quantitative and qualitative changes in the effector and memory CTL populations.


Subject(s)
AIDS Vaccines/immunology , Adenoviridae/genetics , Antibodies/immunology , CD8-Positive T-Lymphocytes/immunology , HIV-1/immunology , Interferon-gamma/antagonists & inhibitors , env Gene Products, Human Immunodeficiency Virus/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/genetics , Animals , Antibodies/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/genetics , Female , Genetic Vectors , HIV-1/genetics , Mice , Mice, Inbred BALB C , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , env Gene Products, Human Immunodeficiency Virus/genetics
7.
PLoS One ; 5(8): e12357, 2010 Aug 23.
Article in English | MEDLINE | ID: mdl-20808813

ABSTRACT

Although a variety of genetic strategies have been developed to inhibit HIV replication, few direct comparisons of the efficacy of these inhibitors have been carried out. Moreover, most studies have not examined whether genetic inhibitors are able to induce a survival advantage that results in an expansion of genetically-modified cells following HIV infection. We evaluated the efficacy of three leading genetic strategies to inhibit HIV replication: 1) an HIV-1 tat/rev-specific small hairpin (sh) RNA; 2) an RNA antisense gene specific for the HIV-1 envelope; and 3) a viral entry inhibitor, maC46. In stably transduced cell lines selected such that >95% of cells expressed the genetic inhibitor, the RNA antisense envelope and viral entry inhibitor maC46 provided the strongest inhibition of HIV-1 replication. However, when mixed populations of transduced and untransduced cells were challenged with HIV-1, the maC46 fusion inhibitor resulted in highly efficient positive selection of transduced cells, an effect that was evident even in mixed populations containing as few as 1% maC46-expressing cells. The selective advantage of the maC46 fusion inhibitor was also observed in HIV-1-infected cultures of primary T lymphocytes as well as in HIV-1-infected humanized mice. These results demonstrate robust inhibition of HIV replication with the fusion inhibitor maC46 and the antisense Env inhibitor, and importantly, a survival advantage of cells expressing the maC46 fusion inhibitor both in vitro and in vivo. Evaluation of the ability of genetic inhibitors of HIV-1 replication to confer a survival advantage on genetically-modified cells provides unique information not provided by standard techniques that may be important in the in vivo efficacy of these genes.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Membrane/metabolism , HIV Fusion Inhibitors/metabolism , HIV Infections , HIV-1/physiology , Recombinant Fusion Proteins/genetics , Animals , CD4-Positive T-Lymphocytes/virology , Cell Line , Cell Survival , Gene Expression , Genetic Therapy , HIV Fusion Inhibitors/pharmacology , HIV Infections/genetics , HIV Infections/therapy , HIV-1/drug effects , Humans , Mice , RNA, Antisense/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Species Specificity , Transduction, Genetic , Virus Internalization/drug effects , Virus Replication/drug effects , Virus Replication/genetics
8.
Blood ; 115(15): 3070-8, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20147699

ABSTRACT

One of the most puzzling observations in HIV research is the lack of pathogenicity in most nonhuman primate species that are natural hosts of simian immunodeficiency virus (SIV) infection. Despite this, natural hosts experience a level of viremia similar to humans infected with HIV or macaques infected with SIV. To determine the role of adaptive immune responses in viral containment and lack of disease, we delayed the generation of cellular and humoral immune responses by administering anti-CD8- and anti-CD20 lymphocyte-depleting antibodies to sabaeus African green monkeys (Chlorocebus sabaeus) before challenge with SIV(sab9315BR). In vivo lymphocyte depletion during primary infection resulted in a brief elevation of viremia but not in disease. Based on the magnitude and timing of SIV-specific CD8(+) T-cell responses in the lymphocyte-depleted animals, CD8(+) T-cell responses appear to contribute to viral containment in natural hosts. We found no evidence for a contribution of humoral immune responses in viral containment. These studies indicate that natural hosts have developed mechanisms in addition to classic adaptive immune responses to cope with this lentiviral infection. Thus, adaptive immune responses in natural hosts appear to be less critical for viral containment than in HIV infection.


Subject(s)
Adaptive Immunity/immunology , Chlorocebus aethiops/immunology , Chlorocebus aethiops/virology , Immunosuppression Therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Viremia/prevention & control , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Murine-Derived , Antibodies, Neutralizing/biosynthesis , Antigens, CD20/metabolism , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Immunologic Memory/immunology , Interferon-gamma/immunology , Ki-67 Antigen/metabolism , Lymphocyte Depletion , Lymphoid Tissue/immunology , Rituximab , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus , Time Factors , Viremia/immunology , Viremia/virology
9.
PLoS Pathog ; 5(12): e1000691, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20011508

ABSTRACT

African green monkeys (AGM) and other natural hosts for simian immunodeficiency virus (SIV) do not develop an AIDS-like disease following SIV infection. To evaluate differences in the role of SIV-specific adaptive immune responses between natural and nonnatural hosts, we used SIV(agmVer90) to infect vervet AGM and pigtailed macaques (PTM). This infection results in robust viral replication in both vervet AGM and pigtailed macaques (PTM) but only induces AIDS in the latter species. We delayed the development of adaptive immune responses through combined administration of anti-CD8 and anti-CD20 lymphocyte-depleting antibodies during primary infection of PTM (n = 4) and AGM (n = 4), and compared these animals to historical controls infected with the same virus. Lymphocyte depletion resulted in a 1-log increase in primary viremia and a 4-log increase in post-acute viremia in PTM. Three of the four PTM had to be euthanized within 6 weeks of inoculation due to massive CMV reactivation and disease. In contrast, all four lymphocyte-depleted AGM remained healthy. The lymphocyte-depleted AGM showed only a trend toward a prolongation in peak viremia but the groups were indistinguishable during chronic infection. These data show that adaptive immune responses are critical for controlling disease progression in pathogenic SIV infection in PTM. However, the maintenance of a disease-free course of SIV infection in AGM likely depends on a number of mechanisms including non-adaptive immune mechanisms.


Subject(s)
Adaptive Immunity/immunology , Chlorocebus aethiops/immunology , Macaca nemestrina/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , Antigens, CD20/immunology , B-Lymphocyte Subsets/immunology , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , Chlorocebus aethiops/virology , Immunohistochemistry , Immunophenotyping , In Situ Hybridization , Lymphocyte Depletion , Macaca nemestrina/virology , Reverse Transcriptase Polymerase Chain Reaction , Simian Acquired Immunodeficiency Syndrome/pathology , Viremia
10.
Nat Med ; 15(8): 861-5, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19661993

ABSTRACT

The design of an effective AIDS vaccine has eluded the efforts of the scientific community to the point that alternative approaches to classic vaccine formulations have to be considered. We propose here that HIV vaccine research could greatly benefit from the study of natural simian immunodeficiency virus (SIV) infections of African nonhuman primates. Natural SIV hosts (for example, sooty mangabeys, African green monkeys and mandrills) share many features of HIV infection of humans; however, they usually do not develop immunodeficiency. These natural, nonprogressive SIV infections represent an evolutionary adaptation that allows a peaceful coexistence of primate lentiviruses and the host immune system. This adaptation does not result in reduced viral replication but, rather, involves phenotypic changes to CD4(+) T cell subsets, limited immune activation and preserved mucosal immunity, all of which contribute to the avoidance of disease progression and, possibly, to the reduction of vertical SIV transmission. Here we summarize the current understanding of SIV infection of African nonhuman primates and discuss how unraveling these evolutionary adaptations may provide clues for new vaccine designs that might induce effective immune responses without the harmful consequences of excessive immune activation.


Subject(s)
AIDS Vaccines/therapeutic use , Acquired Immunodeficiency Syndrome/therapy , Monkey Diseases/therapy , Simian Acquired Immunodeficiency Syndrome/immunology , AIDS Vaccines/biosynthesis , Animals , Cercocebus atys/immunology , Chlorocebus aethiops/immunology , Host-Pathogen Interactions/immunology , Humans , Immunity, Active/immunology , Mandrillus/immunology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology
11.
PLoS Med ; 6(7): e1000107, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19582166

ABSTRACT

BACKGROUND: The antibody response to HIV-1 does not appear in the plasma until approximately 2-5 weeks after transmission, and neutralizing antibodies to autologous HIV-1 generally do not become detectable until 12 weeks or more after transmission. Moreover, levels of HIV-1-specific antibodies decline on antiretroviral treatment. The mechanisms of this delay in the appearance of anti-HIV-1 antibodies and of their subsequent rapid decline are not known. While the effect of HIV-1 on depletion of gut CD4(+) T cells in acute HIV-1 infection is well described, we studied blood and tissue B cells soon after infection to determine the effect of early HIV-1 on these cells. METHODS AND FINDINGS: In human participants, we analyzed B cells in blood as early as 17 days after HIV-1 infection, and in terminal ileum inductive and effector microenvironments beginning at 47 days after infection. We found that HIV-1 infection rapidly induced polyclonal activation and terminal differentiation of B cells in blood and in gut-associated lymphoid tissue (GALT) B cells. The specificities of antibodies produced by GALT memory B cells in acute HIV-1 infection (AHI) included not only HIV-1-specific antibodies, but also influenza-specific and autoreactive antibodies, indicating very early onset of HIV-1-induced polyclonal B cell activation. Follicular damage or germinal center loss in terminal ileum Peyer's patches was seen with 88% of follicles exhibiting B or T cell apoptosis and follicular lysis. CONCLUSIONS: Early induction of polyclonal B cell differentiation, coupled with follicular damage and germinal center loss soon after HIV-1 infection, may explain both the high rate of decline in HIV-1-induced antibody responses and the delay in plasma antibody responses to HIV-1. Please see later in the article for Editors' Summary.


Subject(s)
B-Lymphocytes/metabolism , Germinal Center/pathology , HIV Antibodies/blood , HIV Infections/immunology , HIV-1/immunology , Ileum/immunology , Lymphocyte Activation , Adolescent , Adult , Antibodies, Viral/blood , Apoptosis/immunology , Autoimmunity , Cell Differentiation/immunology , Female , Germinal Center/virology , Humans , Ileum/pathology , Ileum/virology , Influenza, Human/immunology , Male , Middle Aged , Peyer's Patches/pathology , Peyer's Patches/virology , Time Factors , Young Adult
12.
J Virol ; 82(23): 11577-88, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18829748

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Animals , Apoptosis Regulatory Proteins/blood , Chlorocebus aethiops , Chronic Disease , Granzymes/blood , Interferon-gamma/biosynthesis , Ki-67 Antigen/blood , Lymphocyte Activation , Macaca mulatta , RNA, Viral/blood , Tumor Necrosis Factor-alpha/biosynthesis
13.
J Virol ; 82(17): 8812-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18579590

ABSTRACT

While a diversity of immunogens that elicit qualitatively different cellular immune responses are being assessed in clinical human immunodeficiency virus vaccine trials, the consequences of those varied responses for viral control remain poorly understood. In the present study, we evaluated the induction of virus-specific T-cell responses in rhesus monkeys using a series of diverse vaccine vectors. We assessed both the magnitude and the functional profile of the virus-specific CD8(+) T cells by measuring gamma interferon, interleukin-2, and tumor necrosis factor alpha production. We found that the different vectors generated virus-specific T-cell responses of different magnitudes and with different functional profiles. Heterologous prime-boost vaccine regimens induced particularly high-frequency virus-specific T-cell responses with polyfunctional repertoires. Yet, immediately after a pathogenic simian-human immunodeficiency virus (SHIV) challenge, no significant differences were observed between these cohorts of vaccinated monkeys in the magnitudes or the functional profiles of their virus-specific CD8(+) T cells. This finding suggests that the high viral load shapes the functional repertoire of the cellular immune response during primary infection. Nevertheless, in all vaccination regimens, higher frequency and more polyfunctional vaccine-elicited virus-specific CD8(+) T-cell responses were associated with better viral control after SHIV challenge. These observations highlight the contributions of both the quality and the magnitude of vaccine-elicited cellular immune responses in the control of immunodeficiency virus replication.


Subject(s)
SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , T-Lymphocytes/immunology , Virus Replication , Adenoviridae/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Count , Cohort Studies , Cytokines/analysis , Genetic Vectors/immunology , Immunization, Secondary , Interferon-gamma/analysis , Interferon-gamma/immunology , Macaca mulatta , RNA, Viral/blood , Tumor Necrosis Factor-alpha/biosynthesis , Viral Load
14.
J Virol ; 82(11): 5631-5, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18367519

ABSTRACT

Here, we investigated the containment of virus replication in simian immunodeficiency virus (SIV) infection by CD8(+) lymphocytes. Escape mutations in Mamu-A*01 epitopes appeared first in SIV Tat TL8 and then in SIV Gag p11C. The appearance of escape mutations in SIV Gag p11C was coincident with compensatory changes outside of the epitope. Eliminating CD8(+) lymphocytes from rhesus monkeys during primary infection resulted in more rapid disease progression that was associated with preservation of canonical epitopes. These results confirm the importance of cytotoxic T cells in controlling viremia and the constraint on epitope sequences that require compensatory changes to go to fixation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Macaca mulatta/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/physiology , Virus Replication , Animals , Gene Products, gag/blood , Gene Products, tat/blood , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Macaca mulatta/virology , Mutation/genetics , Simian Acquired Immunodeficiency Syndrome/virology
15.
J Virol ; 82(11): 5618-30, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18367534

ABSTRACT

Previously we have shown that CD8(+) T cells are critical for containment of simian immunodeficiency virus (SIV) viremia and that rapid and profound depletion of CD4(+) T cells occurs in the intestinal tract of acutely infected macaques. To determine the impact of SIV-specific CD8(+) T-cell responses on the magnitude of the CD4(+) T-cell depletion, we investigated the effect of CD8(+) lymphocyte depletion during primary SIV infection on CD4(+) T-cell subsets and function in peripheral blood, lymph nodes, and intestinal tissues. In peripheral blood, CD8(+) lymphocyte-depletion changed the dynamics of CD4(+) T-cell loss, resulting in a more pronounced loss 2 weeks after infection, followed by a temporal rebound approximately 2 months after infection, when absolute numbers of CD4(+) T cells were restored to baseline levels. These CD4(+) T cells showed a markedly skewed phenotype, however, as there were decreased levels of memory cells in CD8(+) lymphocyte-depleted macaques compared to controls. In intestinal tissues and lymph nodes, we observed a significantly higher loss of CCR5(+) CD45RA(-) CD4(+) T cells in CD8(+) lymphocyte-depleted macaques than in controls, suggesting that these SIV-targeted CD4(+) T cells were eliminated more efficiently in CD8(+) lymphocyte-depleted animals. Also, CD8(+) lymphocyte depletion significantly affected the ability to generate SIV Gag-specific CD4(+) T-cell responses and neutralizing antibodies. These results reemphasize that SIV-specific CD8(+) T-cell responses are absolutely critical to initiate at least partial control of SIV infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Leukocyte Common Antigens/immunology , Macaca mulatta/immunology , Receptors, CCR5/immunology , Simian Immunodeficiency Virus/immunology , Animals , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/cytology , Cell Movement/immunology , Disease Progression , Gene Products, gag/immunology , Intestines/cytology , Intestines/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/virology , Macaca mulatta/genetics , Macaca mulatta/metabolism , Macaca mulatta/virology , Male , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism , Survival Rate , Virus Replication
16.
J Virol ; 80(24): 12032-40, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17035327

ABSTRACT

Dominant epitope-specific CD8(+) T-lymphocyte responses play a central role in controlling viral spread. We explored the basis for the development of this focused immune response in simian immunodeficiency virus (SIV)- and simian-human immunodeficiency virus (SHIV)-infected rhesus monkeys through the use of two dominant (p11C and p199RY) and two subdominant (p68A and p56A) epitopes. Using real-time PCR to quantitate T-cell receptor (TCR) variable region beta (Vbeta) family usage, we show that CD8(+) T-lymphocyte populations specific for dominant epitopes are characterized by a diverse Vbeta repertoire, whereas those specific for subdominant epitopes employ a dramatically more focused Vbeta repertoire. We also demonstrate that dominant epitope-specific CD8(+) T lymphocytes employ TCRs with multiple CDR3 lengths, whereas subdominant epitope-specific cells employ TCRs with a more restricted CDR3 length. Thus, the relative dominance of an epitope-specific CD8(+) T-lymphocyte response reflects the clonal diversity of that response. These findings suggest that the limited clonal repertoire of subdominant epitope-specific CD8(+) T-lymphocyte populations may limit the ability of these epitope-specific T-lymphocyte populations to expand and therefore limit the ability of these cell populations to contribute to the control of viral replication.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Immunity, Cellular/immunology , Receptors, Antigen, T-Cell/immunology , Simian Immunodeficiency Virus/immunology , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/metabolism , Chromatography, High Pressure Liquid , DNA Primers , DNA, Complementary/genetics , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Flow Cytometry , Genes, T-Cell Receptor beta/genetics , Macaca mulatta , Molecular Sequence Data , Peptides/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Sequence Analysis, DNA
17.
J Virol ; 80(22): 10950-6, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16943292

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Subsets/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Animals , Immunity, Cellular , Immunologic Memory , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/mortality , Simian Immunodeficiency Virus/immunology , Statistics as Topic , Tumor Necrosis Factor-alpha/biosynthesis
18.
Science ; 312(5779): 1530-3, 2006 Jun 09.
Article in English | MEDLINE | ID: mdl-16763152

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Vaccines, DNA/immunology , Amino Acid Sequence , Animals , Humans , Macaca mulatta , Molecular Sequence Data , Plasmids , Simian Acquired Immunodeficiency Syndrome/prevention & control , Survival Analysis , Vaccines, Synthetic/immunology , Virus Replication
19.
AIDS Res Hum Retroviruses ; 22(5): 445-52, 2006 May.
Article in English | MEDLINE | ID: mdl-16706622

ABSTRACT

Because of the importance of developing HIV vaccine strategies that generate cytotoxic T lymphocyte (CTL) responses with a maximal breadth of epitope recognition, we have explored a variety of novel strategies designed to overcome the usual propensity of CTLs to focus recognition on a limited number of dominant epitopes. In studies of rhesus monkeys expressing the Mamu-A*01 MHC class I allele, we show that variously configured multiepitope plasmid DNA vaccine constructs elicit CTL populations that do not evidence skewing of recognition to dominant epitopes. Nevertheless, repeated boosting of these vaccinated monkeys with different live recombinant vaccine vectors uncovers and amplifies the usual CTL epitope dominance hierarchy. Importantly, in vitro peptide stimulation of peripheral blood mononuclear cells from monkeys that have received only a multiepitope plasmid DNA priming immunization uncovers this dominance hierarchy. Therefore, the dominance hierarchy of the vaccine-elicited epitope-specific CTL populations is inherent in the T lymphocytes of the monkeys after initial exposure to epitope peptides, and the ultimate breadth of epitope recognition cannot be modified thereafter. This finding underscores the enormous challenge associated with increasing the breadth of CTL recognition through vaccination.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Immunodominant Epitopes/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/immunology , Alleles , Animals , In Vitro Techniques , Macaca mulatta , Peptides/immunology , Plasmids/genetics
20.
J Immunol ; 176(9): 5338-45, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16622001

ABSTRACT

Functional impairment of virus-specific memory CD8(+) T lymphocytes has been associated with clinical disease progression following HIV, SIV, and simian human immunodeficiency virus infection. These lymphocytes have a reduced capacity to produce antiviral cytokines and mediators involved in the lysis of virally infected cells. In the present study, we used polychromatic flow cytometry to assess the frequency and functional capacity of central memory (CD28(+)CD95(+)) and effector memory (CD28(-)CD95(+)) subpopulations of Gag-specific CD8(+) T cells in SIV/simian human immunodeficiency virus-infected rhesus monkeys. The aim of this study was to determine whether Ag-specific, memory CD8(+) T cell function could be preserved in infected monkeys that had been immunized before infection with a vaccine regimen consisting of a plasmid DNA prime followed by a recombinant viral vector boost. We observed that vaccination was associated with the preservation of Gag-specific central memory CD8(+) T cells that were functionally capable of producing IFN-gamma, and effector memory CD8(+) T cells that were capable of producing granzyme B following viral Ag exposure.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Macaca mulatta/immunology , Macaca mulatta/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Viral Vaccines/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Communication/immunology , Cells, Cultured , Disease Progression , Epitopes, T-Lymphocyte/immunology , Gene Expression Regulation , Gene Products, gag/immunology , Granzymes , Interferon-gamma/biosynthesis , Macaca mulatta/metabolism , Peptides/immunology , Serine Endopeptidases/metabolism , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...