Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Publication year range
1.
Cell ; 176(5): 967-981.e19, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30739797

ABSTRACT

Tissue-resident lymphocytes play a key role in immune surveillance, but it remains unclear how these inherently stable cell populations respond to chronic inflammation. In the setting of celiac disease (CeD), where exposure to dietary antigen can be controlled, gluten-induced inflammation triggered a profound depletion of naturally occurring Vγ4+/Vδ1+ intraepithelial lymphocytes (IELs) with innate cytolytic properties and specificity for the butyrophilin-like (BTNL) molecules BTNL3/BTNL8. Creation of a new niche with reduced expression of BTNL8 and loss of Vγ4+/Vδ1+ IELs was accompanied by the expansion of gluten-sensitive, interferon-γ-producing Vδ1+ IELs bearing T cell receptors (TCRs) with a shared non-germline-encoded motif that failed to recognize BTNL3/BTNL8. Exclusion of dietary gluten restored BTNL8 expression but was insufficient to reconstitute the physiological Vγ4+/Vδ1+ subset among TCRγδ+ IELs. Collectively, these data show that chronic inflammation permanently reconfigures the tissue-resident TCRγδ+ IEL compartment in CeD. VIDEO ABSTRACT.


Subject(s)
Celiac Disease/immunology , Inflammation/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Antigens , Butyrophilins/metabolism , Celiac Disease/physiopathology , Chronic Disease , Diet, Gluten-Free , Glutens/metabolism , HEK293 Cells , Humans , Inflammation/metabolism , Intestinal Mucosa/immunology , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism
2.
Nat Immunol ; 21(12): 1552-1562, 2020 12.
Article in English | MEDLINE | ID: mdl-33046887

ABSTRACT

T cell memory relies on the generation of antigen-specific progenitors with stem-like properties. However, the identity of these progenitors has remained unclear, precluding a full understanding of the differentiation trajectories that underpin the heterogeneity of antigen-experienced T cells. We used a systematic approach guided by single-cell RNA-sequencing data to map the organizational structure of the human CD8+ memory T cell pool under physiological conditions. We identified two previously unrecognized subsets of clonally, epigenetically, functionally, phenotypically and transcriptionally distinct stem-like CD8+ memory T cells. Progenitors lacking the inhibitory receptors programmed death-1 (PD-1) and T cell immunoreceptor with Ig and ITIM domains (TIGIT) were committed to a functional lineage, whereas progenitors expressing PD-1 and TIGIT were committed to a dysfunctional, exhausted-like lineage. Collectively, these data reveal the existence of parallel differentiation programs in the human CD8+ memory T cell pool, with potentially broad implications for the development of immunotherapies and vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory , Lymphoid Progenitor Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Biomarkers , Cell Differentiation/immunology , Computational Biology/methods , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunophenotyping , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/immunology , Mice , Telomere Homeostasis
3.
Immunity ; 56(5): 1082-1097.e6, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37100059

ABSTRACT

CD4+ T cell-mediated immunity against Streptococcus pneumoniae (pneumococcus) can protect against recurrent bacterial colonization and invasive pneumococcal diseases (IPDs). Although such immune responses are common, the pertinent antigens have remained elusive. We identified an immunodominant CD4+ T cell epitope derived from pneumolysin (Ply), a member of the bacterial cholesterol-dependent cytolysins (CDCs). This epitope was broadly immunogenic as a consequence of presentation by the pervasive human leukocyte antigen (HLA) allotypes DPB1∗02 and DPB1∗04 and recognition via architecturally diverse T cell receptors (TCRs). Moreover, the immunogenicity of Ply427-444 was underpinned by core residues in the conserved undecapeptide region (ECTGLAWEWWR), enabling cross-recognition of heterologous bacterial pathogens expressing CDCs. Molecular studies further showed that HLA-DP4-Ply427-441 was engaged similarly by private and public TCRs. Collectively, these findings reveal the mechanistic determinants of near-global immune focusing on a trans-phyla bacterial epitope, which could inform ancillary strategies to combat various life-threatening infectious diseases, including IPDs.


Subject(s)
CD4-Positive T-Lymphocytes , Cytotoxins , Humans , Bacteria , Epitopes, T-Lymphocyte , Cholesterol
4.
Nat Immunol ; 20(3): 301-312, 2019 03.
Article in English | MEDLINE | ID: mdl-30664737

ABSTRACT

The fetus is thought to be protected from exposure to foreign antigens, yet CD45RO+ T cells reside in the fetal intestine. Here we combined functional assays with mass cytometry, single-cell RNA sequencing and high-throughput T cell antigen receptor (TCR) sequencing to characterize the CD4+ T cell compartment in the human fetal intestine. We identified 22 CD4+ T cell clusters, including naive-like, regulatory-like and memory-like subpopulations, which were confirmed and further characterized at the transcriptional level. Memory-like CD4+ T cells had high expression of Ki-67, indicative of cell division, and CD5, a surrogate marker of TCR avidity, and produced the cytokines IFN-γ and IL-2. Pathway analysis revealed a differentiation trajectory associated with cellular activation and proinflammatory effector functions, and TCR repertoire analysis indicated clonal expansions, distinct repertoire characteristics and interconnections between subpopulations of memory-like CD4+ T cells. Imaging mass cytometry indicated that memory-like CD4+ T cells colocalized with antigen-presenting cells. Collectively, these results provide evidence for the generation of memory-like CD4+ T cells in the human fetal intestine that is consistent with exposure to foreign antigens.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fetus/immunology , Immunologic Memory/immunology , Intestines/immunology , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD5 Antigens/genetics , CD5 Antigens/immunology , CD5 Antigens/metabolism , Cells, Cultured , Fetus/cytology , Fetus/metabolism , Flow Cytometry , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/immunology , High-Throughput Nucleotide Sequencing , Humans , Immunologic Memory/genetics , Immunophenotyping , Intestines/cytology , Intestines/embryology , Ki-67 Antigen/genetics , Ki-67 Antigen/immunology , Ki-67 Antigen/metabolism
5.
Nat Immunol ; 19(4): 397-406, 2018 04.
Article in English | MEDLINE | ID: mdl-29531339

ABSTRACT

The hallmark function of αß T cell antigen receptors (TCRs) involves the highly specific co-recognition of a major histocompatibility complex molecule and its carried peptide. However, the molecular basis of the interactions of TCRs with the lipid antigen-presenting molecule CD1c is unknown. We identified frequent staining of human T cells with CD1c tetramers across numerous subjects. Whereas TCRs typically show high specificity for antigen, both tetramer binding and autoreactivity occurred with CD1c in complex with numerous, chemically diverse self lipids. Such extreme polyspecificity was attributable to binding of the TCR over the closed surface of CD1c, with the TCR covering the portal where lipids normally protrude. The TCR essentially failed to contact lipids because they were fully seated within CD1c. These data demonstrate the sequestration of lipids within CD1c as a mechanism of autoreactivity and point to small lipid size as a determinant of autoreactive T cell responses.


Subject(s)
Antigens, CD1/immunology , Autoantigens/immunology , Autoimmunity/immunology , Glycoproteins/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Antigen Presentation/immunology , Humans , Lipids/immunology , Lymphocyte Activation/immunology
6.
Nat Immunol ; 18(11): 1228-1237, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28945243

ABSTRACT

Adaptive immune responses protect against infection with dengue virus (DENV), yet cross-reactivity with distinct serotypes can precipitate life-threatening clinical disease. We found that clonotypes expressing the T cell antigen receptor (TCR) ß-chain variable region 11 (TRBV11-2) were 'preferentially' activated and mobilized within immunodominant human-leukocyte-antigen-(HLA)-A*11:01-restricted CD8+ T cell populations specific for variants of the nonstructural protein epitope NS3133 that characterize the serotypes DENV1, DENV3 and DENV4. In contrast, the NS3133-DENV2-specific repertoire was largely devoid of such TCRs. Structural analysis of a representative TRBV11-2+ TCR demonstrated that cross-serotype reactivity was governed by unique interplay between the variable antigenic determinant and germline-encoded residues in the second ß-chain complementarity-determining region (CDR2ß). Extensive mutagenesis studies of three distinct TRBV11-2+ TCRs further confirmed that antigen recognition was dependent on key contacts between the serotype-defined peptide and discrete residues in the CDR2ß loop. Collectively, these data reveal an innate-like mode of epitope recognition with potential implications for the outcome of sequential exposure to heterologous DENVs.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross Reactions/immunology , Dengue Virus/immunology , Germ-Line Mutation/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Amino Acid Sequence , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Dengue/genetics , Dengue/immunology , Dengue/virology , Dengue Virus/classification , Dengue Virus/genetics , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , HLA-A Antigens/chemistry , HLA-A Antigens/genetics , HLA-A Antigens/immunology , Humans , Models, Molecular , Protein Structure, Tertiary , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Serotyping , Surface Plasmon Resonance
7.
Immunity ; 47(5): 835-847.e4, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29150238

ABSTRACT

Immune response (Ir) genes, originally proposed by Baruj Benacerraf to explain differential antigen-specific responses in animal models, have become synonymous with the major histocompatibility complex (MHC). We discovered a non-MHC-linked Ir gene in a T cell receptor (TCR) locus that was required for CD8+ T cell responses to the Plasmodium berghei GAP5040-48 epitope in mice expressing the MHC class I allele H-2Db. GAP5040-48-specific CD8+ T cell responses emerged from a very large pool of naive Vß8.1+ precursors, which dictated susceptibility to cerebral malaria and conferred protection against recombinant Listeria monocytogenes infection. Structural analysis of a prototypical Vß8.1+ TCR-H-2Db-GAP5040-48 ternary complex revealed that germline-encoded complementarity-determining region 1ß residues present exclusively in the Vß8.1 segment mediated essential interactions with the GAP5040-48 peptide. Collectively, these findings demonstrated that Vß8.1 functioned as an Ir gene that was indispensable for immune reactivity against the malaria GAP5040-48 epitope.


Subject(s)
Histocompatibility Antigen H-2D/genetics , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Receptors, Antigen, T-Cell/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Complementarity Determining Regions , Epitopes , Genes, T-Cell Receptor beta , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptide Fragments/immunology
8.
Proc Natl Acad Sci U S A ; 120(33): e2303155120, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37561786

ABSTRACT

Human cytomegalovirus (HCMV) is a major human pathogen whose life-long persistence is enabled by its remarkable capacity to systematically subvert host immune defenses. In exploring the finding that HCMV infection up-regulates tumor necrosis factor receptor 2 (TNFR2), a ligand for the pro-inflammatory antiviral cytokine TNFα, we found that the underlying mechanism was due to targeting of the protease, A Disintegrin And Metalloproteinase 17 (ADAM17). ADAM17 is the prototype 'sheddase', a family of proteases that cleaves other membrane-bound proteins to release biologically active ectodomains into the supernatant. HCMV impaired ADAM17 surface expression through the action of two virally-encoded proteins in its UL/b' region, UL148 and UL148D. Proteomic plasma membrane profiling of cells infected with an HCMV double-deletion mutant for UL148 and UL148D with restored ADAM17 expression, combined with ADAM17 functional blockade, showed that HCMV stabilized the surface expression of 114 proteins (P < 0.05) in an ADAM17-dependent fashion. These included reported substrates of ADAM17 with established immunological functions such as TNFR2 and jagged1, but also numerous unreported host and viral targets, such as nectin1, UL8, and UL144. Regulation of TNFα-induced cytokine responses and NK inhibition during HCMV infection were dependent on this impairment of ADAM17. We therefore identify a viral immunoregulatory mechanism in which targeting a single sheddase enables broad regulation of multiple critical surface receptors, revealing a paradigm for viral-encoded immunomodulation.


Subject(s)
Cytomegalovirus , Tumor Necrosis Factor-alpha , Humans , Cytomegalovirus/physiology , Tumor Necrosis Factor-alpha/metabolism , Proteome/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Proteomics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Cytokines/metabolism , Cell Membrane/metabolism , Metalloproteases/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Membrane Glycoproteins/metabolism , Viral Proteins/metabolism
9.
Eur J Immunol ; : e2451190, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39072722

ABSTRACT

Sepsis affects 25 million children per year globally, leading to 2.9 million deaths and substantial disability in survivors. Extensive characterization of interactions between the host and bacteria in children is required to design novel preventive and therapeutic strategies tailored to this age group. Vγ9Vδ2 T cells are the first T cells generated in humans. These cells are defined by the expression of Vγ9Vδ2 T-cell receptors (TCRs, using the TRGV9 and TRDV2 gene segments), which react strongly against the prototypical bacterial phosphoantigen HMBPP. We investigated this reactivity by analyzing the TCR δ (TRD) repertoire in the blood of 76 children (0-16 years) with blood culture-proven bacterial sepsis caused by HMBPP-positive Escherichia coli or by HMBPP-negative Staphylococcus aureus or by HMBPP-negative Streptococcus pneumoniae. Strikingly, we found that S. aureus, and to a lesser extent E. coli but not S. pneumoniae, shaped the TRDV2 repertoire in young children (<2 years) but not in older children or adults. This dichotomy was due to the selective expansion of a fetal TRDV2 repertoire. Thus, young children possess fetal-derived Vγ9Vδ2 T cells that are highly responsive toward specific bacterial pathogens.

10.
J Biol Chem ; 299(8): 104981, 2023 08.
Article in English | MEDLINE | ID: mdl-37390984

ABSTRACT

CD8+ T cell-mediated recognition of peptide-major histocompatibility complex class I (pMHCI) molecules involves cooperative binding of the T cell receptor (TCR), which confers antigen specificity, and the CD8 coreceptor, which stabilizes the TCR/pMHCI complex. Earlier work has shown that the sensitivity of antigen recognition can be regulated in vitro by altering the strength of the pMHCI/CD8 interaction. Here, we characterized two CD8 variants with moderately enhanced affinities for pMHCI, aiming to boost antigen sensitivity without inducing non-specific activation. Expression of these CD8 variants in model systems preferentially enhanced pMHCI antigen recognition in the context of low-affinity TCRs. A similar effect was observed using primary CD4+ T cells transduced with cancer-targeting TCRs. The introduction of high-affinity CD8 variants also enhanced the functional sensitivity of primary CD8+ T cells expressing cancer-targeting TCRs, but comparable results were obtained using exogenous wild-type CD8. Specificity was retained in every case, with no evidence of reactivity in the absence of cognate antigen. Collectively, these findings highlight a generically applicable mechanism to enhance the sensitivity of low-affinity pMHCI antigen recognition, which could augment the therapeutic efficacy of clinically relevant TCRs.


Subject(s)
CD8 Antigens , CD8-Positive T-Lymphocytes , Histocompatibility Antigens Class I , Lymphocyte Activation , Histocompatibility Antigens Class I/metabolism , Peptides/metabolism , Receptors, Antigen, T-Cell/metabolism , Humans
11.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Article in English | MEDLINE | ID: mdl-34272276

ABSTRACT

CD8+ T cells are inherently cross-reactive and recognize numerous peptide antigens in the context of a given major histocompatibility complex class I (MHCI) molecule via the clonotypically expressed T cell receptor (TCR). The lineally expressed coreceptor CD8 interacts coordinately with MHCI at a distinct and largely invariant site to slow the TCR/peptide-MHCI (pMHCI) dissociation rate and enhance antigen sensitivity. However, this biological effect is not necessarily uniform, and theoretical models suggest that antigen sensitivity can be modulated in a differential manner by CD8. We used two intrinsically controlled systems to determine how the relationship between the TCR/pMHCI interaction and the pMHCI/CD8 interaction affects the functional sensitivity of antigen recognition. Our data show that modulation of the pMHCI/CD8 interaction can reorder the agonist hierarchy of peptide ligands across a spectrum of affinities for the TCR.


Subject(s)
CD8 Antigens/immunology , Peptides/agonists , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Antigens/chemistry , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cross Reactions , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Kinetics , Ligands , Lymphocyte Activation , Models, Immunological , Mutation
12.
J Immunol ; 207(4): 1009-1017, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34321228

ABSTRACT

The human CD8+ T cell clone 6C5 has previously been shown to recognize the tert-butyl-modified Bax161-170 peptide LLSY(3-tBu)FGTPT presented by HLA-A*02:01. This nonnatural epitope was likely created as a by-product of fluorenylmethoxycarbonyl protecting group peptide synthesis and bound poorly to HLA-A*02:01. In this study, we used a systematic approach to identify and characterize natural ligands for the 6C5 TCR. Functional analyses revealed that 6C5 T cells only recognized the LLSYFGTPT peptide when tBu was added to the tyrosine residue and did not recognize the LLSYFGTPT peptide modified with larger (di-tBu) or smaller chemical groups (Me). Combinatorial peptide library screening further showed that 6C5 T cells recognized a series of self-derived peptides with dissimilar amino acid sequences to LLSY(3-tBu)FGTPT. Structural studies of LLSY(3-tBu)FGTPT and two other activating nonamers (IIGWMWIPV and LLGWVFAQV) in complex with HLA-A*02:01 demonstrated similar overall peptide conformations and highlighted the importance of the position (P) 4 residue for T cell recognition, particularly the capacity of the bulky amino acid tryptophan to substitute for the tBu-modified tyrosine residue in conjunction with other changes at P5 and P6. Collectively, these results indicated that chemical modifications directly altered the immunogenicity of a synthetic peptide via molecular mimicry, leading to the inadvertent activation of a T cell clone with unexpected and potentially autoreactive specificities.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Peptide Fragments/immunology , Peptides/immunology , Amino Acid Sequence , Antigen Presentation/immunology , Cells, Cultured , Epitopes, T-Lymphocyte/immunology , Humans , Ligands , Peptide Library
13.
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
14.
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
15.
PLoS Biol ; 16(6): e2005523, 2018 06.
Article in English | MEDLINE | ID: mdl-29933397

ABSTRACT

Adaptive immunity relies on the generation and maintenance of memory T cells to provide protection against repeated antigen exposure. It has been hypothesised that a self-renewing population of T cells, named stem cell-like memory T (TSCM) cells, are responsible for maintaining memory. However, it is not clear if the dynamics of TSCM cells in vivo are compatible with this hypothesis. To address this issue, we investigated the dynamics of TSCM cells under physiological conditions in humans in vivo using a multidisciplinary approach that combines mathematical modelling, stable isotope labelling, telomere length analysis, and cross-sectional data from vaccine recipients. We show that, unexpectedly, the average longevity of a TSCM clone is very short (half-life < 1 year, degree of self-renewal = 430 days): far too short to constitute a stem cell population. However, we also find that the TSCM population is comprised of at least 2 kinetically distinct subpopulations that turn over at different rates. Whilst one subpopulation is rapidly replaced (half-life = 5 months) and explains the rapid average turnover of the bulk TSCM population, the half-life of the other TSCM subpopulation is approximately 9 years, consistent with the longevity of the recall response. We also show that this latter population exhibited a high degree of self-renewal, with a cell residing without dying or differentiating for 15% of our lifetime. Finally, although small, the population was not subject to excessive stochasticity. We conclude that the majority of TSCM cells are not stem cell-like but that there is a subpopulation of TSCM cells whose dynamics are compatible with their putative role in the maintenance of T cell memory.


Subject(s)
Cell Self Renewal/immunology , Immunologic Memory , T-Lymphocyte Subsets/immunology , Adult , Aged, 80 and over , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Humans , Kinetics , Mathematical Concepts , Middle Aged , Models, Immunological , T-Lymphocyte Subsets/cytology , Telomere Homeostasis/immunology , Yellow fever virus/immunology
16.
J Immunol ; 203(5): 1276-1287, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31308093

ABSTRACT

CD4+ T cells are essential for immune protection against viruses, yet their multiple roles remain ill-defined at the single-cell level in humans. Using HLA class II tetramers, we studied the functional properties and clonotypic architecture of EBV-specific CD4+ T cells in patients with infectious mononucleosis, a symptomatic manifestation of primary EBV infection, and in long-term healthy carriers of EBV. We found that primary infection elicited oligoclonal expansions of TH1-like EBV-specific CD4+ T cells armed with cytotoxic proteins that responded immediately ex vivo to challenge with EBV-infected B cells. Importantly, these acutely generated cytotoxic CD4+ T cells were highly activated and transcriptionally distinct from classically described cytotoxic CD4+ memory T cells that accumulate during other persistent viral infections, including CMV and HIV. In contrast, EBV-specific memory CD4+ T cells displayed increased cytokine polyfunctionality but lacked cytotoxic activity. These findings suggested an important effector role for acutely generated cytotoxic CD4+ T cells that could potentially be harnessed to improve the efficacy of vaccines against EBV.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , T-Lymphocytes, Cytotoxic/immunology , B-Lymphocytes/immunology , CD4 Antigens/immunology , Humans , Immunologic Memory/immunology , Infectious Mononucleosis/immunology
17.
J Immunol ; 202(3): 943-955, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30635396

ABSTRACT

Candidate vaccines designed to generate T cell-based immunity are typically vectored by nonpersistent viruses, which largely fail to elicit durable effector memory T cell responses. This limitation can be overcome using recombinant strains of CMV. Proof-of-principle studies have demonstrated the potential benefits of this approach, most notably in the SIV model, but safety concerns require the development of nonreplicating alternatives with comparable immunogenicity. In this study, we show that IL-33 promotes the accumulation and recall kinetics of circulating and tissue-resident memory T cells in mice infected with murine CMV. Using a replication-deficient murine CMV vector, we further show that exogenous IL-33 boosts vaccine-induced memory T cell responses, which protect against subsequent heterologous viral challenge. These data suggest that IL-33 could serve as a useful adjuvant to improve the efficacy of vaccines based on attenuated derivatives of CMV.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Vaccines/immunology , Immunologic Memory , Interleukin-33/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Cytomegalovirus , Interleukin-33/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus , Vaccines, Attenuated/immunology
18.
Diabetologia ; 63(6): 1174-1185, 2020 06.
Article in English | MEDLINE | ID: mdl-32157332

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to characterise islet autoantibody profiles and immune cell phenotypes in slow progressors to type 1 diabetes. METHODS: Immunological variables were compared across peripheral blood samples obtained from slow progressors to type 1 diabetes, individuals with newly diagnosed or long-standing type 1 diabetes, and healthy individuals. Polychromatic flow cytometry was used to characterise the phenotypic attributes of B and T cells. Islet autoantigen-specific B cells were quantified using an enzyme-linked immunospot (ELISpot) assay and islet autoantigen-specific CD8+ T cells were quantified using peptide-HLA class I tetramers. Radioimmunoassays were used to detect islet autoantibodies. Sera were assayed for various chemokines, cytokines and soluble receptors via ELISAs. RESULTS: Islet autoantibodies were lost over time in slow progressors. Various B cell subsets expressed higher levels of CD95 in slow progressors, especially after polyclonal stimulation, compared with the corresponding B cell subsets in healthy donors (p < 0.05). The phenotypic characteristics of CD4+ and CD8+ T cells were similar in slow progressors and healthy donors. Lower frequencies of CD4+ T cells with a central memory phenotype (CD27int, CD127+, CD95int) were observed in slow progressors compared with healthy donors (mean percentage of total CD4+ T cells was 3.00% in slow progressors vs 4.67% in healthy donors, p < 0.05). Autoreactive B cell responses to proinsulin were detected at higher frequencies in slow progressors compared with healthy donors (median no. of spots was 0 in healthy donors vs 24.34 in slow progressors, p < 0.05) in an ELISpot assay. Islet autoantigen-specific CD8+ T cell responses were largely absent in slow progressors and healthy donors. Serum levels of DcR3, the decoy receptor for CD95L, were elevated in slow progressors compared with healthy donors (median was 1087 pg/ml in slow progressors vs 651 pg/ml in healthy donors, p = 0.06). CONCLUSIONS/INTERPRETATION: In this study, we found that slow progression to type 1 diabetes was associated with a loss of islet autoantibodies and a distinct B cell phenotype, consistent with enhanced apoptotic regulation of peripheral autoreactivity via CD95. These phenotypic changes warrant further studies in larger cohorts to determine their functional implications.


Subject(s)
Autoantibodies/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , fas Receptor/immunology , Autoantibodies/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/metabolism , Flow Cytometry , Humans , Proinsulin/immunology , Proinsulin/metabolism , fas Receptor/metabolism
19.
Immunol Cell Biol ; 98(9): 770-781, 2020 10.
Article in English | MEDLINE | ID: mdl-32568415

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are key players in the immune response against microbial infection. The MAIT T-cell receptor (TCR) recognizes a diverse array of microbial ligands, and recent reports have highlighted the variability in the MAIT TCR that could further contribute to discrimination of ligand. The MAIT TCR complementarity determining region (CDR)3ß sequence displays a high level of diversity across individuals, and clonotype usage appears to be dependent on antigenic exposure. To address the relationship between the MAIT TCR and microbial ligand, we utilized a previously defined panel of MAIT cell clones that demonstrated variability in responses against different microbial infections. Sequencing of these clones revealed four pairs, each with shared (identical) CDR3α and different CDR3ß sequences. These pairs demonstrated varied responses against microbially infected dendritic cells as well as against 5-(2-oxopropylideneamino)-6-d-ribitylaminouracil, a ligand abundant in Salmonella enterica serovar Typhimurium, suggesting that the CDR3ß contributes to differences in ligand discrimination. Taken together, these results highlight a key role for the MAIT CDR3ß region in distinguishing between MR1-bound antigens and ligands.


Subject(s)
Bacterial Infections/immunology , Complementarity Determining Regions/genetics , Genes, T-Cell Receptor beta , Lymphocyte Activation , Mucosal-Associated Invariant T Cells , Humans , Ligands , Mucosal-Associated Invariant T Cells/immunology , Ribitol/analogs & derivatives , Uracil/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL