Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Nat Immunol ; 20(8): 1059-1070, 2019 08.
Article in English | MEDLINE | ID: mdl-31308541

ABSTRACT

Dysfunction of virus-specific CD4+ T cells in chronic human infections is poorly understood. We performed genome-wide transcriptional analyses and functional assays of CD4+ T cells specific for human immunodeficiency virus (HIV) from HIV-infected people before and after initiation of antiretroviral therapy (ART). A follicular helper T cell (TFH cell)-like profile characterized HIV-specific CD4+ T cells in viremic infection. HIV-specific CD4+ T cells from people spontaneously controlling the virus (elite controllers) robustly expressed genes associated with the TH1, TH17 and TH22 subsets of helper T cells. Viral suppression by ART resulted in a distinct transcriptional landscape, with a reduction in the expression of genes associated with TFH cells, but persistently low expression of genes associated with TH1, TH17 and TH22 cells compared to the elite controller profile. Thus, altered differentiation is central to the impairment of HIV-specific CD4+ T cells and involves both gain of function and loss of function.


Subject(s)
Anti-HIV Agents/therapeutic use , Gene Expression/drug effects , HIV Infections/drug therapy , HIV Infections/immunology , Th1 Cells/pathology , Th17 Cells/pathology , Gene Expression Profiling , HIV Infections/virology , Humans , Receptors, CXCR5/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Viral Load/drug effects , Virus Replication/drug effects
2.
J Virol ; 91(7)2017 04 01.
Article in English | MEDLINE | ID: mdl-28100613

ABSTRACT

Although multiple restriction factors have been shown to inhibit HIV/SIV replication, little is known about their expression in vivo Expression of 45 confirmed and putative HIV/SIV restriction factors was analyzed in CD4+ T cells from peripheral blood and the jejunum in rhesus macaques, revealing distinct expression patterns in naive and memory subsets. In both peripheral blood and the jejunum, memory CD4+ T cells expressed higher levels of multiple restriction factors compared to naive cells. However, relative to their expression in peripheral blood CD4+ T cells, jejunal CCR5+ CD4+ T cells exhibited significantly lower expression of multiple restriction factors, including APOBEC3G, MX2, and TRIM25, which may contribute to the exquisite susceptibility of these cells to SIV infection. In vitro stimulation with anti-CD3/CD28 antibodies or type I interferon resulted in upregulation of distinct subsets of multiple restriction factors. After infection of rhesus macaques with SIVmac239, the expression of most confirmed and putative restriction factors substantially increased in all CD4+ T cell memory subsets at the peak of acute infection. Jejunal CCR5+ CD4+ T cells exhibited the highest levels of SIV RNA, corresponding to the lower restriction factor expression in this subset relative to peripheral blood prior to infection. These results illustrate the dynamic modulation of confirmed and putative restriction factor expression by memory differentiation, stimulation, tissue microenvironment and SIV infection and suggest that differential expression of restriction factors may play a key role in modulating the susceptibility of different populations of CD4+ T cells to lentiviral infection.IMPORTANCE Restriction factors are genes that have evolved to provide intrinsic defense against viruses. HIV and simian immunodeficiency virus (SIV) target CD4+ T cells. The baseline level of expression in vivo and degree to which expression of restriction factors is modulated by conditions such as CD4+ T cell differentiation, stimulation, tissue location, or SIV infection are currently poorly understood. We measured the expression of 45 confirmed and putative restriction factors in primary CD4+ T cells from rhesus macaques under various conditions, finding dynamic changes in each state. Most dramatically, in acute SIV infection, the expression of almost all target genes analyzed increased. These are the first measurements of many of these confirmed and putative restriction factors in primary cells or during the early events after SIV infection and suggest that the level of expression of restriction factors may contribute to the differential susceptibility of CD4+ T cells to SIV infection.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Gene Expression Regulation/immunology , Immunity, Innate , Jejunum/immunology , Jejunum/virology , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/metabolism , Transcriptome/immunology
3.
PLoS Pathog ; 11(3): e1004740, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25768938

ABSTRACT

The onset of protective immunity against pathogenic SIV challenge in SIVΔnef-vaccinated macaques is delayed for 15-20 weeks, a process that is related to qualitative changes in CD8+ T cell responses induced by SIVΔnef. As a novel approach to characterize cell differentiation following vaccination, we used multi-target qPCR to measure transcription factor expression in naĆÆve and memory subsets of CD8++ T cells, and in SIV-specific CD8+ T cells obtained from SIVΔnef-vaccinated or wild type SIVmac239-infected macaques. Unsupervised clustering of expression profiles organized naĆÆve and memory CD8+ T cells into groups concordant with cell surface phenotype. Transcription factor expression patterns in SIV-specific CD8+ T cells in SIVΔnef-vaccinated animals were distinct from those observed in purified CD8+ T cell subsets obtained from naĆÆve animals, and were intermediate to expression profiles of purified central memory and effector memory T cells. Expression of transcription factors elicited by SIVΔnef vaccination also varied over time: cells obtained at later time points, temporally associated with greater protection, appeared more central-memory like than cells obtained at earlier time points, which appeared more effector memory-like. Expression of transcription factors associated with effector differentiation, such as ID2 and RUNX3, were decreased over time, while expression of transcription factors associated with quiescence or memory differentiation, such as TCF7, BCOR and EOMES, increased. CD8+ T cells specific for a more conserved epitope expressed higher levels of TBX21 and BATF, and appeared more effector-like than cells specific for an escaped epitope, consistent with continued activation by replicating vaccine virus. These data suggest transcription factor expression profiling is a novel method that can provide additional data complementary to the analysis of memory cell differentiation based on classical phenotypic markers. Additionally, these data support the hypothesis that ongoing stimulation by SIVΔnef promotes a distinct protective balance of CD8+ T cell differentiation and activation states.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , Transcription Factors/immunology , Viral Regulatory and Accessory Proteins , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/immunology , Female , Gene Expression Profiling , Macaca mulatta , SAIDS Vaccines/genetics , Simian Immunodeficiency Virus/genetics , Transcription Factors/genetics
4.
Blood ; 118(12): 3321-30, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21791421

ABSTRACT

Natural killer (NK) cells are classically viewed as effector cells that kill virus-infected and neoplastic cells, but recent studies have identified a rare mucosal NK- cell subpopulation secreting the TH17 cytokine IL-22. Here, we report identification of 2 distinct lineages of mucosal NK cells characterized as NKG2A(+)NFIL3(+)RORC(-) and NKp44(+)NFIL3(+)RORC(+). NKG2A(+) NK cells were systemically distributed, cytotoxic, and secreted IFN-ƎĀ³, whereas NKp44(+) NK cells were mucosae-restricted, noncytotoxic, and produced IL-22 and IL-17. During SIV infection, NKp44(+) NK cells became apoptotic, were depleted, and had an altered functional profile characterized by decreased IL-17 secretion; increased IFN-ƎĀ³ secretion; and, surprisingly, increased potential for cytotoxicity. NKp44(+) NK cells showed no evidence of direct SIV infection; rather, depletion and altered function were associated with SIV-induced up-regulation of inflammatory mediators in the gut, including indoleamine 2,3-dioxygenase 1. Furthermore, treatment of NKp44(+) NK cells with indoleamine 2,3-dioxygenase 1 catabolites in vitro ablated IL-17 production in a dose-dependent manner, whereas other NK-cell functions were unaffected. Thus lentiviral infection both depletes and modifies the functional repertoire of mucosal NK cells involved in the maintenance of gut integrity, a finding that highlights the plasticity of this rare mucosal NK-cell population.


Subject(s)
Cytotoxicity, Immunologic/drug effects , Gastric Mucosa/drug effects , Immunity, Mucosal/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology , Inflammation/immunology , Interleukin-17/antagonists & inhibitors , Killer Cells, Natural/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , T-Lymphocyte Subsets/immunology , Animals , Cell Lineage/immunology , Cytotoxicity, Immunologic/immunology , Flow Cytometry , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Immunity, Mucosal/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Interleukins/biosynthesis , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Macaca mulatta , Natural Cytotoxicity Triggering Receptor 2/immunology , Primary Cell Culture , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/growth & development , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Viral Load/drug effects , Virus Replication/drug effects , Interleukin-22
5.
Am J Pathol ; 173(1): 130-43, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18556781

ABSTRACT

Human immunodeficiency virus encephalitis causes dementia in acquired immune deficiency syndrome patients. Using proteomic analysis of postmortem cerebrospinal fluid (CSF) and brain tissue from the simian immunodeficiency virus primate model, we demonstrate here a specific increase in YKL-40 that was tightly associated with lentiviral encephalitis. Longitudinal analysis of CSF from simian immunodeficiency virus-infected pigtailed macaques showed an increase in YKL-40 concentration 2 to 8 weeks before death from encephalitis. This increase in YKL-40 correlated with an increase in CSF viral load; it may therefore represent a biomarker for the development of encephalitis. Analysis of banked human CSF from human immunodeficiency virus-infected patients also demonstrated a correlation between YKL-40 concentration and CSF viral load. In vitro studies demonstrated increased YKL-40 expression and secretion by macrophages and microglia but not by neurons or astrocytes. We found that YKL40 displaced extracellular matrix-bound basic fibroblast growth factor (bFGF) as well as inhibited the mitogenic activity of both fibroblast growth factor receptor 1-expressing BaF3 cells and bFGF-induced axonal branching in hippocampal cultures. Taken together, these findings demonstrate that during lentiviral encephalitis, YKL-40 may interfere with the biological activity of bFGF and potentially of other heparin-binding growth factors and chemokines that can affect neuronal function or survival.


Subject(s)
Biomarkers/cerebrospinal fluid , Encephalitis, Viral/cerebrospinal fluid , Fibroblast Growth Factor 2/metabolism , Simian Acquired Immunodeficiency Syndrome/cerebrospinal fluid , Adipokines , Animals , Blotting, Western , Chitinase-3-Like Protein 1 , Encephalitis, Viral/metabolism , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix , Glycoproteins , HIV Infections/complications , HIV Infections/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Lectins , Macaca nemestrina , Mass Spectrometry , Microglia/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Simian Acquired Immunodeficiency Syndrome/complications , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Immunodeficiency Virus , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
6.
Front Immunol ; 4: 32, 2013.
Article in English | MEDLINE | ID: mdl-23423644

ABSTRACT

Human NK (hNK) cells play a key role in mediating host immune responses against various infectious diseases. For practical reasons, the majority of the data on hNK cells has been generated using peripheral blood lymphocytes. In contrast, our knowledge of NK cells in human tissues is limited, and not much is known about developmental pathways of hNK cell subpopulations in vivo. Although research in mice has elucidated a number of fundamental features of NK cell biology, mouse, and hNK cells significantly differ in their subpopulations, functions, and receptor repertoires. Thus, there is a need for a model that is more closely related to humans and yet allows experimental manipulations. Non-human primate models offer numerous opportunities for the study of NK cells, including the study of the role of NK cells after solid organ and stem cell transplantation, as well as in acute viral infection. Macaque NK cells can be depleted in vivo or adoptively transferred in an autologous system. All of these studies are either difficult or unethical to carry out in humans. Here we highlight recent advances in rhesus NK cell research and their parallels in humans. Using high-throughput transcriptional profiling, we demonstrate that the human CD56(bright) and CD56(dim) NK cell subsets have phenotypically and functionally analogous counterparts in rhesus macaques. Thus, the use of non-human primate models offers the potential to substantially advance hNK cell research.

7.
Virology ; 354(1): 116-31, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-16884757

ABSTRACT

Determining the impact of antiretroviral therapy on virus evolution could advance the development of improved therapeutics/vaccines against HIV. Toward this goal, we analyzed virus burden, quasispecies complexity, and T cell responses in SIV/DeltaB670-infected rhesus macaques+/-treatment for 7 months with PMPA (2-30 weeks postinfection). Treatment divided the animals into two groups: poor responders (a reduction of < or =1 log) and responders (> or =2 log reduction) in virus burden. Virus evolution in poor responders and untreated controls was characterized by expression of a complex quasispecies that evolved as the disease progressed. This included the universal loss of a viral genotype selected against by in vitro passage in monkey cells and selected for by propagation in human cells. In contrast, a good response to PMPA was characterized by infection with a less complex quasispecies that evolved more slowly. Interestingly, in 2 of the best responders, the human-preferred genotype persisted until the study was discontinued (89 weeks p.i.). Neither virus burden nor the magnitude of the T cell response at 2 weeks postinfection predicted PMPA responsiveness. However, responders expressed a less complex quasispecies than nonresponders prior to treatment. These data suggest a role for intrinsic host factors in treatment responsiveness, and lend support for therapeutic vaccination as an adjunct to effective therapy.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/therapeutic use , Organophosphonates/therapeutic use , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/genetics , Adenine/pharmacology , Adenine/therapeutic use , Amino Acid Sequence , Animals , Antiviral Agents/pharmacology , Drug Resistance, Viral , Evolution, Molecular , Genome, Viral , Genotype , Glycosylation , Heteroduplex Analysis , Humans , Macaca mulatta , Molecular Sequence Data , Organophosphonates/pharmacology , Sequence Homology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/classification , Simian Immunodeficiency Virus/physiology , T-Lymphocytes/immunology , Tenofovir , Viral Load , Virus Replication
8.
Virology ; 348(1): 200-15, 2006 Apr 25.
Article in English | MEDLINE | ID: mdl-16439000

ABSTRACT

DNA immunization in conjunction with antiretroviral therapy was evaluated in SIV-infected rhesus macaques treated with [R]-9-[2-phosphonylmethoxypropyl]adenine (PMPA). Macaques were immunized monthly with DNA vaccines expressing either SIV gag/tat or SIV gag/tat and 19 CD8+ T cell epitopes during 7 months of therapy. Half the animals from each group were additionally immunized before infection. Only 60% of the animals (4 controls, 20 vaccinated) responded to PMPA (ART responders). All 4 ART responder controls demonstrated viral rebound or CD4 decline after PMPA was withdrawn. In contrast, 17 of 20 vaccinated ART responders contained viral rebound for over 7 months after PMPA was withdrawn. Viral control correlated with stable CD4 counts, higher lymphoproliferation and an increase in the magnitude and breadth of the CD8+ T cell response. Immunizing before infection or with multi-epitopes enhanced these effects. These results demonstrate that DNA immunization during antiretroviral therapy may be an effective strategy to treat HIV infection.


Subject(s)
Adenine/analogs & derivatives , Organophosphonates/administration & dosage , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Vaccines, DNA/administration & dosage , Viremia/prevention & control , Adenine/administration & dosage , Adenine/therapeutic use , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Disease Progression , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Gene Products, gag/genetics , Gene Products, gag/immunology , Gene Products, tat/genetics , Gene Products, tat/immunology , Immunotherapy, Active/methods , Macaca mulatta , Organophosphonates/therapeutic use , RNA, Viral/blood , Simian Immunodeficiency Virus/drug effects , Statistics as Topic , Tenofovir , Vaccines, DNA/immunology , Withholding Treatment
9.
J Gen Virol ; 87(Pt 1): 139-149, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16361426

ABSTRACT

Adenovirus serotype 35 (Ad35) is a promising vaccine platform for human immunodeficiency virus (HIV) infection and emerging infectious diseases as it is uncommon in humans worldwide and is distinct from Ad5, the major vaccine serotype for which many individuals have pre-existing immunity. The immunogenicity of a first-generation, replication-competent Ad35-based vaccine was tested in the simian immunodeficiency virus (SIV) rhesus macaque model by evaluating its capacity to boost immunity generated by Ad5-based vectors. A series of four immunizations with replication-defective Ad5 vectors expressing SIVmac239 gag induced high-frequency responses mediated by both CD8(+) and CD4(+) T cells directed against several epitopes. Ad5-specific neutralizing antibody responses that did not neutralize Ad35 were rapidly induced but waned over time. Subsequent immunization with Ad5-based vectors was minimally effective, whereas immunization with Ad35-based vectors generated a strong increase in the frequency of Gag-specific T cells with specificities that were unchanged. While this boosting response was relatively transient, challenge with the distinct pathogenic isolate SIV/DeltaB670 generated robust and selective recall responses to Gag with similar specificities as induced by vaccination that were elevated for 25 weeks relative to controls. Vaccination had measurable albeit minor effects on virus load. Unexpectedly, regional hypervariability within the Gag sequence of SIV/DeltaB670 was associated with mutation of the conserved CD8(+) T-cell epitope CM9 without concurrent flanking mutations and in the absence of immune pressure. These findings support the further development of Ad35 as a vaccine vector, and promote vaccine regimens that utilize serial administration of heterologous adenoviruses.


Subject(s)
Adenoviruses, Human/immunology , Gene Products, gag/genetics , Simian Immunodeficiency Virus/immunology , Viral Vaccines/immunology , Adenoviruses, Human/classification , Animals , Antibody Formation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gene Products, gag/administration & dosage , Gene Products, gag/blood , Gene Products, gag/immunology , Genetic Vectors/immunology , Immunity, Cellular , Immunization, Secondary , Macaca mulatta , Simian Immunodeficiency Virus/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage
10.
J Infect Dis ; 187(4): 576-88, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12599074

ABSTRACT

To establish experimental Pneumocystis carinii infection in simian immunodeficiency virus (SIV)-infected macaques as a model of acquired immunodeficiency syndrome (AIDS)-associated P. carinii pneumonia (PCP), SIV-infected macaques were inoculated intrabronchially with macaque-derived P. carinii, and P. carinii-specific polymerase chain reaction (PCR) and flow cytometric analysis of bronchoalveolar lavage fluid were done biweekly for up to 44 weeks after inoculation. All inoculated animals had a P. carinii-specific PCR product after infection. CD8(+) T cells in lung lavage samples from SIV- and P. carinii-coinfected animals increased to >90% of total CD3(+) cells, a pattern associated with naturally acquired P. carinii infection. Progression of disease also was correlated with increased neutrophil infiltration to the lungs. The animals had a protracted period of asymptomatic colonization with P. carinii before progression to PCP. The development of a model of PCP in SIV-infected rhesus macaques provides the means to study AIDS-associated PCP.


Subject(s)
AIDS-Related Opportunistic Infections/complications , Pneumocystis , Pneumonia, Pneumocystis/complications , Simian Acquired Immunodeficiency Syndrome/complications , Simian Immunodeficiency Virus , AIDS-Related Opportunistic Infections/diagnosis , AIDS-Related Opportunistic Infections/etiology , Animals , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/virology , CD3 Complex/analysis , CD8-Positive T-Lymphocytes/immunology , DNA, Fungal/analysis , Disease Models, Animal , Female , Lung/microbiology , Lung/pathology , Macaca mulatta , Male , Neutrophil Infiltration , Pneumocystis/genetics , Pneumocystis/isolation & purification , Pneumonia, Pneumocystis/diagnosis , Pneumonia, Pneumocystis/etiology , Polymerase Chain Reaction , Time Factors
11.
J Virol ; 76(7): 3309-17, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11884556

ABSTRACT

An effective vaccine against human immunodeficiency virus (HIV) should protect against mucosal transmission of genetically divergent isolates. As a safe alternative to live attenuated vaccines, the immunogenicity and protective efficacy of a DNA vaccine containing simian immunodeficiency virus (SIV) strain 17E-Fr (SIV/17E-Fr) gag-pol-env was analyzed in rhesus macaques. Significant levels of cytotoxic T lymphocytes (CTL), but low to undetectable serum antibody responses, were observed following multiple immunizations. SIV-specific mucosal antibodies and CTL were also detected in rectal washes and gut-associated lymphoid tissues, respectively. Vaccinated and naive control monkeys were challenged intrarectally with SIV strain DeltaB670 (SIV/DeltaB670), a primary isolate whose env is 15% dissimilar to that of the vaccine strain. Four of seven vaccinees were protected from infection as determined by the inability to identify viral RNA or DNA sequences in the peripheral blood and the absence of anamnestic antibody responses postchallenge. This is the first report of mucosal protection against a primary pathogenic, heterologous isolate of SIV by using a commercially viable vaccine approach. These results support further development of a DNA vaccine for protection against HIV.


Subject(s)
Immunity, Mucosal , Membrane Glycoproteins , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Vaccination , Viral Envelope Proteins , Amino Acid Sequence , Animals , Antibodies, Viral/analysis , Gene Products, env/genetics , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , Immunoglobulin A/analysis , Immunoglobulin G/analysis , Intestines/immunology , Lymphoid Tissue/immunology , Macaca mulatta , Molecular Sequence Data , Rectum/immunology , Sequence Alignment , Simian Immunodeficiency Virus/genetics , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL