Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Immunity ; 38(3): 425-36, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23521884

ABSTRACT

The capacity of the immune system to adapt to rapidly evolving viruses is a primary feature of effective immunity, yet its molecular basis is unclear. Here, we investigated protective HIV-1-specific CD8+ T cell responses directed against the immunodominant p24 Gag-derived epitope KK10 (KRWIILGLNK263-272) presented by human leukocyte antigen (HLA)-B∗2705. We found that cross-reactive CD8+ T cell clonotypes were mobilized to counter the rapid emergence of HIV-1 variants that can directly affect T cell receptor (TCR) recognition. These newly recruited clonotypes expressed TCRs that engaged wild-type and mutant KK10 antigens with similar affinities and almost identical docking modes, thereby accounting for their antiviral efficacy in HLA-B∗2705+ individuals. A protective CD8+ T cell repertoire therefore encompasses the capacity to control TCR-accessible mutations, ultimately driving the development of more complex viral escape variants that disrupt antigen presentation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , HLA-B27 Antigen/immunology , Amino Acid Sequence , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Clone Cells/immunology , Clone Cells/metabolism , Clone Cells/virology , Crystallography, X-Ray , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , HIV Core Protein p24/genetics , HIV Core Protein p24/immunology , HIV Core Protein p24/metabolism , HIV Infections/virology , HIV-1/genetics , HIV-1/metabolism , HLA-B27 Antigen/chemistry , HLA-B27 Antigen/metabolism , Humans , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/immunology , Immunodominant Epitopes/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism
2.
J Immunol ; 204(1): 112-121, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31818981

ABSTRACT

CMV is an obligate and persistent intracellular pathogen that continually drives the production of highly differentiated virus-specific CD8+ T cells in an Ag-dependent manner, a phenomenon known as memory inflation. Extensive proliferation is required to generate and maintain inflationary CD8+ T cell populations, which are counterintuitively short-lived and typically exposed to limited amounts of Ag during the chronic phase of infection. An apparent discrepancy therefore exists between the magnitude of expansion and the requirement for ongoing immunogenic stimulation. To address this issue, we explored the clonal dynamics of memory inflation. First, we tracked congenically marked OT-I cell populations in recipient mice infected with murine CMV (MCMV) expressing the cognate Ag OVA. Irrespective of numerical dominance, stochastic expansions were observed in each population, such that dominant and subdominant OT-I cells were maintained at stable frequencies over time. Second, we characterized endogenous CD8+ T cell populations specific for two classic inflationary epitopes, M38 and IE3. Multiple clonotypes simultaneously underwent Ag-driven proliferation during latent infection with MCMV. In addition, the corresponding CD8+ T cell repertoires were stable over time and dominated by persistent clonotypes, many of which also occurred in more than one mouse. Collectively, these data suggest that stochastic encounters with Ag occur frequently enough to maintain oligoclonal populations of inflationary CD8+ T cells, despite intrinsic constraints on epitope display at individual sites of infection with MCMV.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Clonal Selection, Antigen-Mediated/immunology , Immunologic Memory/immunology , Muromegalovirus/immunology , Animals , Cell Proliferation , Epitopes/immunology , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology
3.
Proc Natl Acad Sci U S A ; 116(10): 3974-3981, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30765525

ABSTRACT

Accumulating evidence indicates that the immune system does not develop in a linear fashion, but rather as distinct developmental layers formed from sequential waves of hematopoietic stem cells, each giving rise to unique populations of immune cells at different stages of development. Although recent studies have indicated that conventional CD8+ T cells produced in early life persist into adulthood and exhibit distinct roles during infection, the developmental architecture of the peripheral T cell compartment remains undefined. In this study, we used a mouse model to permanently label CD8+ T cells produced during distinct windows of development and traced their history to generate fate maps of CD8+ T cells produced during different stages of life. We then used mathematical modeling to understand the age structure of the CD8+ T cell compartment across the lifespan. Interestingly, we found that survival rate of CD8+ T cells depends on both the age and developmental origin of the cells. Recently produced cells show an initial rapid decay rate, which slows with age of the animal at which the cells were produced. For cells produced at any age, the rate of decay also slows with the age of the cell. We derive a function to describe this and predict the "age distribution" of the CD8+ T cell pool for animals of any given age. These data provide a quantitative framework for understanding the ontogeny of the CD8+ T cell compartment and help to contextualize age-related changes in the CD8+ T cell response to infection.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/immunology , Models, Immunological , Aging/genetics , Animals , CD8-Positive T-Lymphocytes/cytology , Mice , Mice, Transgenic
4.
Virol J ; 18(1): 21, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33451356

ABSTRACT

BACKGROUND: The generation of accurate and reproducible viral sequence data is necessary to understand the diversity present in populations of RNA viruses isolated from clinical samples. While various sequencing methods are available, they often require high quality templates and high viral titer to ensure reliable data. METHODS: We modified a multiplex PCR and sequencing approach to characterize populations of simian immunodeficiency virus (SIV) isolated from nonhuman primates. We chose this approach with the aim of reducing the number of required input templates while maintaining fidelity and sensitivity. We conducted replicate sequencing experiments using different numbers of quantified viral RNA (vRNA) or viral cDNA as input material. We performed assays with clonal SIVmac239 to detect false positives, and we mixed SIVmac239 and a variant with 24 point mutations (SIVmac239-24X) to measure variant detection sensitivity. RESULTS: We found that utilizing a starting material of quantified viral cDNA templates had a lower rate of false positives and increased reproducibility when compared to that of quantified vRNA templates. This study identifies the importance of rigorously validating deep sequencing methods and including replicate samples when using a new method to characterize low frequency variants in a population with a small number of templates. CONCLUSIONS: Because the need to generate reproducible and accurate sequencing data from diverse viruses from low titer samples, we modified a multiplex PCR and sequencing approach to characterize SIV from populations from non-human primates. We found that increasing starting template numbers increased the reproducibility and decreased the number of false positives identified, and this was further seen when cDNA was used as a starting material. Ultimately, we highlight the importance of vigorously validating methods to prevent overinterpretation of low frequency variants in a sample.


Subject(s)
DNA, Viral/genetics , High-Throughput Nucleotide Sequencing/methods , Multiplex Polymerase Chain Reaction/methods , Multiplex Polymerase Chain Reaction/standards , RNA, Viral/genetics , Simian Immunodeficiency Virus/genetics , Animals , Genome, Viral , Humans , Macaca mulatta , Reproducibility of Results , Sensitivity and Specificity , Simian Acquired Immunodeficiency Syndrome/virology
5.
Proc Natl Acad Sci U S A ; 115(29): E6817-E6825, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29967140

ABSTRACT

Lifelong interactions between host and the ubiquitous and persistent cytomegalovirus (CMV) have been proposed to contribute to the age-related decline in immunity. Prior work from us and others found some support for that idea, yet evidence that this led to increased vulnerability to other infections was not obtained. Moreover, evidence has accumulated that CMV infection can be beneficial to immune defense in young/adult mice and humans, dominantly via enhanced innate immunity. Here, we describe an unexpected impact of murine CMV (MCMV) upon the T cell response of old mice to Listeria monocytogenes expressing the model antigen, OVA (Lm-OVA). Single-cell sequencing of the OVA-specific CD8 T cell receptor ß (TCRß) repertoire of old mice demonstrated that old MCMV-infected mice recruited many diverse clonotypes that afforded broad and often more efficient recognition of antigenic peptide variants. This stood in contrast to old control mice, which exhibited strong narrowing and homogenization of the elicited repertoire. High-throughput sequencing of the total naïve CD8 TCRß repertoire showed that many of these diverse OVA-specific clonotypes were present in the naïve CD8 repertoire of mice in all groups (adult, old control, and old MCMV+) yet were only recruited into the Lm-OVA response in MCMV+ old mice. These results have profound implications for our understanding of T cell immunity over a life span and suggest that our coevolution with CMV may include surprising, potentially positive impacts on adaptive heterologous immunity in late life.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Immunity, Cellular , Listeria monocytogenes/immunology , Listeriosis/immunology , Muromegalovirus/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cytomegalovirus Infections/pathology , Listeriosis/pathology , Male , Mice
6.
PLoS Pathog ; 14(3): e1006964, 2018 03.
Article in English | MEDLINE | ID: mdl-29590202

ABSTRACT

Defining the complex dynamics of Zika virus (ZIKV) infection in pregnancy and during transmission between vertebrate hosts and mosquito vectors is critical for a thorough understanding of viral transmission, pathogenesis, immune evasion, and potential reservoir establishment. Within-host viral diversity in ZIKV infection is low, which makes it difficult to evaluate infection dynamics. To overcome this biological hurdle, we constructed a molecularly barcoded ZIKV. This virus stock consists of a "synthetic swarm" whose members are genetically identical except for a run of eight consecutive degenerate codons, which creates approximately 64,000 theoretical nucleotide combinations that all encode the same amino acids. Deep sequencing this region of the ZIKV genome enables counting of individual barcodes to quantify the number and relative proportions of viral lineages present within a host. Here we used these molecularly barcoded ZIKV variants to study the dynamics of ZIKV infection in pregnant and non-pregnant macaques as well as during mosquito infection/transmission. The barcoded virus had no discernible fitness defects in vivo, and the proportions of individual barcoded virus templates remained stable throughout the duration of acute plasma viremia. ZIKV RNA also was detected in maternal plasma from a pregnant animal infected with barcoded virus for 67 days. The complexity of the virus population declined precipitously 8 days following infection of the dam, consistent with the timing of typical resolution of ZIKV in non-pregnant macaques and remained low for the subsequent duration of viremia. Our approach showed that synthetic swarm viruses can be used to probe the composition of ZIKV populations over time in vivo to understand vertical transmission, persistent reservoirs, bottlenecks, and evolutionary dynamics.


Subject(s)
Biological Evolution , Gene Library , Infectious Disease Transmission, Vertical , Macaca mulatta/genetics , Mosquito Vectors , Zika Virus Infection/complications , Zika Virus/classification , Animals , Female , High-Throughput Nucleotide Sequencing , Macaca mulatta/virology , Male , Viremia , Zika Virus/genetics , Zika Virus/pathogenicity , Zika Virus Infection/transmission , Zika Virus Infection/virology
7.
Int J Mol Sci ; 21(22)2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33198297

ABSTRACT

T follicular helper (Tfh) cells are a specialised subset of CD4+ T cells that play a significant role in the adaptive immune response, providing critical help to B cells within the germinal centres (GC) of secondary lymphoid organs. The B cell receptors of GC B cells undergo multiple rounds of somatic hypermutation and affinity maturation within the GC response, a process dependent on cognate interactions with Tfh cells. B cells that receive sufficient help from Tfh cells form antibody-producing long-lived plasma and memory B cells that provide the basis of decades of effective and efficient protection and are considered the gold standard in correlates of protection post-vaccination. However, the T cell response to vaccination has been understudied, and over the last 10 years, exponential improvements in the technological underpinnings of sampling techniques, experimental and analytical tools have allowed multidisciplinary characterisation of the role of T cells and the immune system as a whole. Of particular interest to the field of vaccinology are GCs and Tfh cells, representing a unique target for improving immunisation strategies. Here, we discuss recent insights into the unique journey of Tfh cells from thymus to lymph node during differentiation and their role in the production of high-quality antibody responses as well as their journey back to the periphery as a population of memory cells. Further, we explore their function in health and disease and the power of next-generation sequencing techniques to uncover their potential as modulators of vaccine-induced immunity.


Subject(s)
Germinal Center/immunology , Systems Biology , T Follicular Helper Cells/immunology , Vaccines , Animals , B-Lymphocytes/immunology , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Humans , Immune System , Lymph Nodes/immunology , RNA, Small Cytoplasmic/metabolism , RNA-Seq , Receptors, Antigen, T-Cell/immunology , Signal Transduction , T-Lymphocytes/immunology , Treatment Outcome , Vaccination
8.
Immunol Cell Biol ; 97(6): 586-596, 2019 07.
Article in English | MEDLINE | ID: mdl-30875134

ABSTRACT

The peripheral maturation of human CD1d-restricted natural killer T (NKT) cells has not been well described. In this study, we identified four major subsets of NKT cells in adults, distinguished by the expression of CD4, CD8 and CCR5. Phenotypic analysis suggested a hierarchical pattern of differentiation, whereby immature CD4+ CD8- CCR5- cells progressed to an intermediate CD4+ CD8- CCR5+ stage, which remained less differentiated than the CD4- CD8- and CD4- CD8+ subsets, both of which expressed CCR5. This interpretation was supported by functional data, including clonogenic potential and cytokine secretion profiles, as well as T-cell receptor (TCR) excision circle analysis. Moreover, conventional and high-throughput sequencing of the corresponding TCR repertoires demonstrated significant clonotypic overlap within individuals, especially between the more differentiated CD4- CD8- and CD4- CD8+ subsets. Collectively, these results mapped a linear differentiation pathway across the post-thymic landscape of human CD1d-restricted NKT cells.


Subject(s)
Lymphocyte Subsets/immunology , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/genetics , Antigens, CD1d/metabolism , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Differentiation , Cells, Cultured , Clone Cells , Cytokines/metabolism , Flow Cytometry , High-Throughput Nucleotide Sequencing , Humans , Immunophenotyping , Receptors, CCR5/metabolism
9.
PLoS Pathog ; 13(5): e1006359, 2017 May.
Article in English | MEDLINE | ID: mdl-28472156

ABSTRACT

HIV and SIV infection dynamics are commonly investigated by measuring plasma viral loads. However, this total viral load value represents the sum of many individual infection events, which are difficult to independently track using conventional sequencing approaches. To overcome this challenge, we generated a genetically tagged virus stock (SIVmac239M) with a 34-base genetic barcode inserted between the vpx and vpr accessory genes of the infectious molecular clone SIVmac239. Next-generation sequencing of the virus stock identified at least 9,336 individual barcodes, or clonotypes, with an average genetic distance of 7 bases between any two barcodes. In vitro infection of rhesus CD4+ T cells and in vivo infection of rhesus macaques revealed levels of viral replication of SIVmac239M comparable to parental SIVmac239. After intravenous inoculation of 2.2x105 infectious units of SIVmac239M, an average of 1,247 barcodes were identified during acute infection in 26 infected rhesus macaques. Of the barcodes identified in the stock, at least 85.6% actively replicated in at least one animal, and on average each barcode was found in 5 monkeys. Four infected animals were treated with combination antiretroviral therapy (cART) for 82 days starting on day 6 post-infection (study 1). Plasma viremia was reduced from >106 to <15 vRNA copies/mL by the time treatment was interrupted. Virus rapidly rebounded following treatment interruption and between 87 and 136 distinct clonotypes were detected in plasma at peak rebound viremia. This study confirmed that SIVmac239M viremia could be successfully curtailed with cART, and that upon cART discontinuation, rebounding viral variants could be identified and quantified. An additional 6 animals infected with SIVmac239M were treated with cART beginning on day 4 post-infection for 305, 374, or 482 days (study 2). Upon treatment interruption, between 4 and 8 distinct viral clonotypes were detected in each animal at peak rebound viremia. The relative proportions of the rebounding viral clonotypes, spanning a range of 5 logs, were largely preserved over time for each animal. The viral growth rate during recrudescence and the relative abundance of each rebounding clonotype were used to estimate the average frequency of reactivation per animal. Using these parameters, reactivation frequencies were calculated and ranged from 0.33-0.70 events per day, likely representing reactivation from long-lived latently infected cells. The use of SIVmac239M therefore provides a powerful tool to investigate SIV latency and the frequency of viral reactivation after treatment interruption.


Subject(s)
Genetic Variation , Genome, Viral/genetics , Models, Theoretical , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Virus Replication , Animals , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/virology , Genetic Markers/genetics , Macaca mulatta , Male , Sequence Analysis, DNA , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/genetics , Viral Load , Viremia
10.
J Immunol ; 196(4): 1604-16, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26764033

ABSTRACT

CMV is the most common congenital infection in the United States. The major target of congenital CMV is the brain, with clinical manifestations including mental retardation, vision impairment, and sensorineural hearing loss. Previous reports have shown that CD8(+) T cells are required to control viral replication and significant numbers of CMV-specific CD8(+) T cells persist in the brain even after the initial infection has been cleared. However, the dynamics of CD8(+) T cells in the brain during latency remain largely undefined. In this report, we used TCR sequencing to track the development and maintenance of neonatal clonotypes in the brain and spleen of mice during chronic infection. Given the discontinuous nature of tissue-resident memory CD8(+) T cells, we hypothesized that neonatal TCR clonotypes would be locked in the brain and persist into adulthood. Surprisingly, we found that the Ag-specific T cell repertoire in neonatal-infected mice diversified during persistent infection in both the brain and spleen, while maintaining substantial similarity between the CD8(+) T cell populations in the brain and spleen in both early and late infection. However, despite the diversification of, and potential interchange between, the spleen and brain Ag-specific T cell repertoires, we observed that germline-encoded TCR clonotypes, characteristic of neonatal infection, persisted in the brain, albeit sometimes in low abundance. These results provide valuable insights into the evolution of CD8(+) T cell repertoires following neonatal CMV infection and thus have important implications for the development of therapeutic strategies to control CMV in early life.


Subject(s)
Brain/immunology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Immunologic Memory , Animals , Animals, Newborn , Cytomegalovirus/immunology , Genes, T-Cell Receptor , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/immunology , Spleen/cytology , Spleen/immunology
11.
Immunol Cell Biol ; 94(9): 838-848, 2016 10.
Article in English | MEDLINE | ID: mdl-27142943

ABSTRACT

Neonates are particularly susceptible to a number of infections, and the neonatal CD8+ T-cell response demonstrates differences in both the phenotype and magnitude of responses to infection compared with adults. However, the underlying basis for these differences is unclear. We have used a mathematical modeling approach to analyze the dynamics of neonatal and adult CD8+ T-cell responses following in vitro stimulation and in vivo infection, which allows us to dissect key cell-intrinsic differences in expansion, differentiation and memory formation. We found that neonatal cells started dividing 8 h earlier and proliferated at a faster rate (0.077 vs 0.105 per day) than adult cells in vitro. In addition, neonatal cells also differentiated more rapidly, as measured by the loss in CD62L and Ly6C expression. We extended our mathematical modeling to analysis of neonatal and adult CD8+ T cells responding in vivo and demonstrated that neonatal cells divide more slowly than adult cells after day 4 post infection. However, neonatal cells differentiate more rapidly, upregulating more KLRG1 per division than adult cells (20% vs 5%). The dynamics of memory formation were also found to be different, with neonatal effector cells showing increased death (1.0 vs 2.45 per day). Comparison of the division of human cord blood and adult naive cells stimulated in vitro showed more division in cord blood-derived cells, consistent with the observations in mice. This work highlights differences of the cell-intrinsic division and differentiation program in neonatal CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Models, Immunological , Adolescent , Adoptive Transfer , Adult , Animals , Cell Differentiation , Cell Division , Cell Proliferation , Fetal Blood/cytology , Humans , Immunologic Memory , Infant, Newborn , Kinetics , Male , Mice , Middle Aged , Phenotype , Young Adult
12.
Immunol Cell Biol ; 94(6): 604-11, 2016 07.
Article in English | MEDLINE | ID: mdl-26860370

ABSTRACT

Heterogeneity of T cells is a hallmark of a successful adaptive immune response, harnessing the vast diversity of antigen-specific T cells into a coordinated evolution of effector and memory outcomes. The T cell receptor (TCR) repertoire is highly diverse to account for the highly heterogeneous antigenic world. During the response to a virus multiple individual clones of antigen specific CD8+ (Ag-specific) T cells can be identified against a single epitope and multiple epitopes are recognised. Advances in single-cell technologies have provided the potential to study Ag-specific T cell heterogeneity at both surface phenotype and transcriptome levels, thereby allowing investigation of the diversity within the same apparent sub-population. We propose a new method (VDJPuzzle) to reconstruct the native TCRαß from single cell RNA-seq data of Ag-specific T cells and then to link these with the gene expression profile of individual cells. We applied this method using rare Ag-specific T cells isolated from peripheral blood of a subject who cleared hepatitis C virus infection. We successfully reconstructed productive TCRαß in 56 of a total of 63 cells (89%), with double α and double ß in 18, and 7% respectively, and double TCRαß in 2 cells. The method was validated via standard single cell PCR sequencing of the TCR. We demonstrate that single-cell transcriptome analysis can successfully distinguish Ag-specific T cell populations sorted directly from resting memory cells in peripheral blood and sorted after ex vivo stimulation. This approach allows a detailed analysis of the TCR diversity and its relationship with the transcriptional profile of different clones.


Subject(s)
Epitopes/genetics , Receptors, Antigen, T-Cell/metabolism , Single-Cell Analysis/methods , T-Lymphocytes/metabolism , Transcriptome/genetics , Epitopes, T-Lymphocyte/immunology , Gene Expression Profiling , Gene Expression Regulation , Humans , Receptors, Antigen, T-Cell, alpha-beta/genetics , Sequence Analysis, RNA , V(D)J Recombination/genetics
13.
J Immunol ; 193(11): 5626-36, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25348625

ABSTRACT

Despite progress toward understanding the correlates of protective T cell immunity in HIV infection, the optimal approach to Ag delivery by vaccination remains uncertain. We characterized two immunodominant CD8 T cell populations generated in response to immunization of BALB/c mice with a replication-deficient adenovirus serotype 5 vector expressing the HIV-derived Gag and Pol proteins at equivalent levels. The Gag-AI9/H-2K(d) epitope elicited high-avidity CD8 T cell populations with architecturally diverse clonotypic repertoires that displayed potent lytic activity in vivo. In contrast, the Pol-LI9/H-2D(d) epitope elicited motif-constrained CD8 T cell repertoires that displayed lower levels of physical avidity and lytic activity despite equivalent measures of overall clonality. Although low-dose vaccination enhanced the functional profiles of both epitope-specific CD8 T cell populations, greater polyfunctionality was apparent within the Pol-LI9/H-2D(d) specificity. Higher proportions of central memory-like cells were present after low-dose vaccination and at later time points. However, there were no noteworthy phenotypic differences between epitope-specific CD8 T cell populations across vaccine doses or time points. Collectively, these data indicate that the functional and phenotypic properties of vaccine-induced CD8 T cell populations are sensitive to dose manipulation, yet constrained by epitope specificity in a clonotype-dependent manner.


Subject(s)
AIDS Vaccines , CD8-Positive T-Lymphocytes/immunology , Immunodominant Epitopes/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , pol Gene Products, Human Immunodeficiency Virus/metabolism , Adenoviridae/genetics , Animals , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Cytotoxicity, Immunologic , Female , Genetic Vectors , H-2 Antigens/metabolism , Histocompatibility Antigen H-2D/metabolism , Humans , Immunodominant Epitopes/genetics , Immunologic Memory , Mice , Mice, Inbred BALB C , Vaccination , gag Gene Products, Human Immunodeficiency Virus/genetics , pol Gene Products, Human Immunodeficiency Virus/genetics
14.
Immunol Cell Biol ; 93(7): 625-33, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25801351

ABSTRACT

Basic parameters of the naive antigen (Ag)-specific T-cell repertoire in humans remain poorly defined. Systematic characterization of this 'ground state' immunity in comparison with memory will allow a better understanding of clonal selection during immune challenge. Here, we used high-definition cell isolation from umbilical cord blood samples to establish the baseline frequency, phenotype and T-cell antigen receptor (TCR) repertoire of CD8(+) T-cell precursor populations specific for a range of viral and self-derived Ags. Across the board, these precursor populations were phenotypically naive and occurred with hierarchical frequencies clustered by Ag specificity. The corresponding patterns of TCR architecture were highly ordered and displayed partial overlap with adult memory, indicating biased structuring of the T-cell repertoire during Ag-driven selection. Collectively, these results provide new insights into the complex nature and dynamics of the naive T-cell compartment.


Subject(s)
Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Clonal Selection, Antigen-Mediated , Fetal Blood/immunology , Hematopoietic Stem Cells/immunology , Phosphoproteins/immunology , Receptors, Antigen, T-Cell/immunology , T-Cell Antigen Receptor Specificity , Viral Matrix Proteins/immunology , Adult , Aging/immunology , Dasatinib/pharmacology , Fetal Blood/cytology , Flow Cytometry , HLA Antigens/immunology , Humans , Immunologic Memory , Immunomagnetic Separation , Immunophenotyping , Infant, Newborn , Peptide Fragments/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics
15.
J Virol ; 88(7): 3837-49, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24453357

ABSTRACT

HIV undergoes high rates of mutation and recombination during reverse transcription, but it is not known whether these events occur independently or are linked mechanistically. Here we used a system of silent marker mutations in HIV and a single round of infection in primary T lymphocytes combined with a high-throughput sequencing and mathematical modeling approach to directly estimate the viral recombination and mutation rates. From >7 million nucleotides (nt) of sequences from HIV infection, we observed 4,801 recombination events and 859 substitution mutations (≈1.51 and 0.12 events per 1,000 nt, respectively). We used experimental controls to account for PCR-induced and transfection-induced recombination and sequencing error. We found that the single-cycle virus-induced mutation rate is 4.6 × 10(-5) mutations per nt after correction. By sorting of our data into recombined and nonrecombined sequences, we found a significantly higher mutation rate in recombined regions (P = 0.003 by Fisher's exact test). We used a permutation approach to eliminate a number of potential confounding factors and confirm that mutation occurs around the site of recombination and is not simply colocated in the genome. By comparing mutation rates in recombined and nonrecombined regions, we found that recombination-associated mutations account for 15 to 20% of all mutations occurring during reverse transcription.


Subject(s)
Amino Acid Substitution , HIV/growth & development , HIV/genetics , Mutation, Missense , Recombination, Genetic , T-Lymphocytes/virology , Cells, Cultured , High-Throughput Nucleotide Sequencing , Humans , Models, Theoretical
16.
PLoS Pathog ; 9(9): e1003572, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24068921

ABSTRACT

Microbial infection during various stages of human development produces widely different clinical outcomes, yet the links between age-related changes in the immune compartment and functional immunity remain unclear. The ability of the immune system to respond to specific antigens and mediate protection in early life is closely correlated with the level of diversification of lymphocyte antigen receptors. We have previously shown that the neonatal primary CD8+ T cell response to replication competent virus is significantly constricted compared to the adult response. In the present study, we have analyzed the subsequent formation of neonatal memory CD8+ T cells and their response to secondary infectious challenge. In particular, we asked whether the less diverse CD8+ T cell clonotypes that are elicited by neonatal vaccination with replication competent virus are 'locked-in' to the adult memory T cell, and thus may compromise the strength of adult immunity. Here we report that neonatal memory CD8+ T cells mediate poor recall responses compared to adults and are comprised of a repertoire of lower avidity T cells. During a later infectious challenge the neonatal memory CD8+ T cells compete poorly with the fully diverse repertoire of naïve adult CD8+ T cells and are outgrown by the adult primary response. This has important implications for the timing of vaccination in early life.


Subject(s)
Aging , CD8-Positive T-Lymphocytes/immunology , Immunologic Deficiency Syndromes/etiology , Immunologic Memory , Listeriosis/physiopathology , Models, Immunological , Vaccines, Attenuated/immunology , Animals , Animals, Newborn , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , DNA, Recombinant/metabolism , Herpes Simplex/immunology , Herpes Simplex/prevention & control , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/metabolism , Immune System/growth & development , Immune System/immunology , Immune System/pathology , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Immunologic Deficiency Syndromes/pathology , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Listeriosis/immunology , Listeriosis/microbiology , Listeriosis/prevention & control , Mice, Inbred Strains , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Specific Pathogen-Free Organisms , Vaccines, Attenuated/adverse effects , Vaccinia/immunology , Vaccinia/prevention & control , Vaccinia/virology , Vaccinia virus/genetics , Vaccinia virus/immunology , Vaccinia virus/metabolism , Virulence
17.
J Immunol ; 190(3): 931-9, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23267020

ABSTRACT

CD8(+) T cells recognize infected or dysregulated cells via the clonotypically expressed αß TCR, which engages Ag in the form of peptide bound to MHC class I (MHC I) on the target cell surface. Previous studies have indicated that a diverse Ag-specific TCR repertoire can be beneficial to the host, yet the determinants of clonotypic diversity are poorly defined. To better understand the factors that govern TCR repertoire formation, we conducted a comprehensive clonotypic analysis of CD8(+) T cell populations directed against epitopes derived from EBV and CMV. Neither pathogen source nor the restricting MHC I molecule were linked with TCR diversity; indeed, both HLA-A and HLA-B molecules were observed to interact with an overlapping repertoire of expressed TRBV genes. Peptide specificity, however, markedly impacted TCR diversity. In addition, distinct peptides sharing HLA restriction and viral origin mobilized TCR repertoires with distinct patterns of TRBV gene usage. Notably, no relationship was observed between immunodominance and TCR diversity. These findings provide new insights into the forces that shape the Ag-specific TCR repertoire in vivo and highlight a determinative role for the peptide component of the peptide-MHC I complex on the molecular frontline of CD8(+) T cell-mediated immune surveillance.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Gene Rearrangement, T-Lymphocyte/immunology , HLA-A Antigens/immunology , HLA-B Antigens/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Cell Antigen Receptor Specificity/immunology , Amino Acid Sequence , CD8-Positive T-Lymphocytes/metabolism , Clonal Selection, Antigen-Mediated , Cytomegalovirus/immunology , Herpesvirus 4, Human/immunology , Humans , Immunodominant Epitopes/immunology , Immunologic Surveillance , Interferon-gamma/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology
18.
PLoS Pathog ; 8(2): e1002544, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22383879

ABSTRACT

The elderly are particularly susceptible to influenza A virus infections, with increased occurrence, disease severity and reduced vaccine efficacy attributed to declining immunity. Experimentally, the age-dependent decline in influenza-specific CD8(+) T cell responsiveness reflects both functional compromise and the emergence of 'repertoire holes' arising from the loss of low frequency clonotypes. In this study, we asked whether early priming limits the time-related attrition of immune competence. Though primary responses in aged mice were compromised, animals vaccinated at 6 weeks then challenged >20 months later had T-cell responses that were normal in magnitude. Both functional quality and the persistence of 'preferred' TCR clonotypes that expand in a characteristic immunodominance hierarchy were maintained following early priming. Similar to the early priming, vaccination at 22 months followed by challenge retained a response magnitude equivalent to young mice. However, late priming resulted in reduced TCRß diversity in comparison with vaccination earlier in life. Thus, early priming was critical to maintaining individual and population-wide TCRß diversity. In summary, early exposure leads to the long-term maintenance of memory T cells and thus preserves optimal, influenza-specific CD8(+) T-cell responsiveness and protects against the age-related attrition of naïve T-cell precursors. Our study supports development of vaccines that prime CD8(+) T-cells early in life to elicit the broadest possible spectrum of CD8(+) T-cell memory and preserve the magnitude, functionality and TCR usage of responding populations. In addition, our study provides the most comprehensive analysis of the aged (primary, secondary primed-early and secondary primed-late) TCR repertoires published to date.


Subject(s)
Aging/immunology , Antigen Presentation/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Immunologic Memory/physiology , Age Factors , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Epitopes, T-Lymphocyte/immunology , Female , Mice , Mice, Inbred C57BL , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/physiology , Receptors, Antigen, T-Cell/immunology , Validation Studies as Topic
19.
J Immunol ; 189(11): 5356-66, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23087407

ABSTRACT

Persistent CMV infection has been associated with immune senescence. To address the causal impact of lifelong persistent viral infection on immune homeostasis and defense, we infected young mice systemically with HSV-1, murine CMV, or both viruses and studied their T cell homeostasis and function. Herpesvirus(+) mice exhibited increased all-cause mortality compared with controls. Upon Listeria-OVA infection, 23-mo-old animals that had experienced lifelong herpesvirus infections showed impaired bacterial control and CD8 T cell function, along with distinct alterations in the T cell repertoire both before and after Listeria challenge, compared with age-matched, herpesvirus-free controls. Herpesvirus infection was associated with reduced naive CD8 T cell precursors above the loss attributable to aging. Moreover, the OVA-specific CD8 T cell repertoire recruited after Listeria challenge was entirely nonoverlapping between control and herpesvirus(+) mice. To our knowledge, this study for the first time causally links lifelong herpesvirus infection to all-cause mortality in mice and to disturbances in the T cell repertoire, which themselves correspond to impaired immunity to a new infection in aging.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/immunology , Herpesviridae Infections/immunology , Listeriosis/immunology , Precursor Cells, T-Lymphoid/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Coinfection , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Herpesvirus 1, Human/immunology , Immunologic Memory , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Listeriosis/microbiology , Listeriosis/pathology , Mice , Muromegalovirus/immunology , Ovalbumin/immunology , Precursor Cells, T-Lymphoid/pathology
20.
Proc Natl Acad Sci U S A ; 108(33): 13694-9, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21813761

ABSTRACT

Immunity against new infections declines in the last quartile of life, as do numbers of naive T cells. Peripheral maintenance of naive T cells over the lifespan is necessary because their production drastically declines by puberty, a result of thymic involution. We report that this maintenance is not random in advanced aging. As numbers and diversity of naive CD8(+) T cells declined with aging, surviving cells underwent faster rates of homeostatic proliferation, were selected for high T-cell receptor:pMHC avidity, and preferentially acquired "memory-like" phenotype. These high-avidity precursors preferentially responded to infection and exhibited strong antimicrobial function. Thus, T-cell receptor avidity for self-pMHC provides a proofreading mechanism to maintain some of the fittest T cells in the otherwise crumbling naive repertoire, providing a degree of compensation for numerical and diversity defects in old T cells.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/cytology , HLA Antigens/immunology , Receptors, Antigen, T-Cell/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cellular Senescence/immunology , Homeostasis/immunology , Immunity/immunology , Immunologic Memory , Lymphocyte Count , Mice
SELECTION OF CITATIONS
SEARCH DETAIL