Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
Immunology ; 163(4): 389-398, 2021 08.
Article En | MEDLINE | ID: mdl-33638871

Oncolytic viruses possess the ability to infect, replicate and lyse malignantly transformed tumour cells. This oncolytic activity amplifies the therapeutic advantage and induces a form of immunogenic cell death, characterized by increased CD8 + T-cell infiltration into the tumour microenvironment. This important feature of oncolytic viruses can result in the warming up of immunologically 'cold' tumour types, presenting the enticing possibility that oncolytic virus treatment combined with immunotherapies may enhance efficacy. In this review, we assess some of the most promising candidates that might be used for oncolytic virotherapy: immunotherapy combinations. We assess their potential as separate agents or as agents combined into a single therapy, where the immunotherapy is encoded within the genome of the oncolytic virus. The development of such advanced agents will require increasingly sophisticated model systems for their preclinical assessment and evaluation. In vivo rodent model systems are fraught with limitations in this regard. Oncolytic viruses replicate selectively within human cells and therefore require human xenografts in immune-deficient mice for their evaluation. However, the use of immune-deficient rodent models hinders the ability to study immune responses against any immunomodulatory transgenes engineered within the viral genome and expressed within the tumour microenvironment. There has therefore been a shift towards the use of more sophisticated ex vivo patient-derived model systems based on organoids and explant co-cultures with immune cells, which may be more predictive of efficacy than contrived and artificial animal models. We review the best of those model systems here.


CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/trends , Neoplasms/immunology , Oncolytic Virotherapy/trends , Oncolytic Viruses/physiology , Animals , CD8-Positive T-Lymphocytes/transplantation , Combined Modality Therapy , Disease Models, Animal , Humans , Immunization , Mice , Neoplasms/therapy , Rats , Tumor Microenvironment
2.
Cell Rep ; 32(2): 107885, 2020 07 14.
Article En | MEDLINE | ID: mdl-32668259

T cell recognition of peptides presented by human leukocyte antigens (HLAs) is mediated by the highly variable T cell receptor (TCR). Despite this built-in TCR variability, individuals can mount immune responses against viral epitopes by using identical or highly related TCRs expressed on CD8+ T cells. Characterization of these TCRs has extended our understanding of the molecular mechanisms that govern the recognition of peptide-HLA. However, few examples exist for CD4+ T cells. Here, we investigate CD4+ T cell responses to the internal proteins of the influenza A virus that correlate with protective immunity. We identify five internal epitopes that are commonly recognized by CD4+ T cells in five HLA-DR1+ subjects and show conservation across viral strains and zoonotic reservoirs. TCR repertoire analysis demonstrates several shared gene usage biases underpinned by complementary biochemical features evident in a structural comparison. These epitopes are attractive targets for vaccination and other T cell therapies.


CD4-Positive T-Lymphocytes/immunology , Epitopes/immunology , Immunoglobulin Variable Region/genetics , Influenza A virus/immunology , Adult , Amino Acid Motifs , Amino Acid Sequence , Animals , Birds/virology , Complementarity Determining Regions/chemistry , Conserved Sequence , Epitopes/chemistry , Female , Germ Cells/metabolism , HLA-DR1 Antigen/immunology , Humans , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/immunology , Male , Middle Aged , Peptides/chemistry , Peptides/immunology , Receptors, Antigen, T-Cell/metabolism , Swine/virology , Tissue Donors , Viral Proteins/immunology , Young Adult , Zoonoses/immunology , Zoonoses/virology
3.
Nucleic Acids Res ; 48(D1): D1057-D1062, 2020 01 08.
Article En | MEDLINE | ID: mdl-31588507

Here, we report an update of the VDJdb database with a substantial increase in the number of T-cell receptor (TCR) sequences and their cognate antigens. The update further provides a new database infrastructure featuring two additional analysis modes that facilitate database querying and real-world data analysis. The increased yield of TCR specificity identification methods and the overall increase in the number of studies in the field has allowed us to expand the database more than 5-fold. Furthermore, several new analysis methods are included. For example, batch annotation of TCR repertoire sequencing samples allows for annotating large datasets on-line. Using recently developed bioinformatic methods for TCR motif mining, we have built a reduced set of high-quality TCR motifs that can be used for both training TCR specificity predictors and matching against TCRs of interest. These additions enhance the versatility of the VDJdb in the task of exploring T-cell antigen specificities. The database is available at https://vdjdb.cdr3.net.


Computational Biology/methods , Databases, Genetic , Nucleotide Motifs , Receptors, Antigen, T-Cell/genetics , V(D)J Recombination , Amino Acid Sequence , High-Throughput Nucleotide Sequencing , Humans , Position-Specific Scoring Matrices , Receptors, Antigen, T-Cell/chemistry , Sequence Analysis, DNA , Software , Web Browser
4.
J Immunol ; 195(12): 5827-36, 2015 Dec 15.
Article En | MEDLINE | ID: mdl-26553072

Fluorochrome-conjugated peptide-MHC (pMHC) class I multimers are staple components of the immunologist's toolbox, enabling reliable quantification and analysis of Ag-specific CD8(+) T cells irrespective of functional outputs. In contrast, widespread use of the equivalent pMHC class II (pMHC-II) reagents has been hindered by intrinsically weaker TCR affinities for pMHC-II, a lack of cooperative binding between the TCR and CD4 coreceptor, and a low frequency of Ag-specific CD4(+) T cell populations in the peripheral blood. In this study, we show that peptide flanking regions, extending beyond the central nonamer core of MHC-II-bound peptides, can enhance TCR-pMHC-II binding and T cell activation without loss of specificity. Consistent with these findings, pMHC-II multimers incorporating peptide flanking residue modifications proved superior for the ex vivo detection, characterization, and manipulation of Ag-specific CD4(+) T cells, highlighting an unappreciated feature of TCR-pMHC-II interactions.


Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Separation/methods , HLA Antigens/immunology , Peptide Fragments/metabolism , Cell Line , Histocompatibility Antigens Class II/metabolism , Humans , Lymphocyte Activation , Protein Binding , Receptors, Antigen, T-Cell/immunology , T-Cell Antigen Receptor Specificity
5.
Oncotarget ; 6(28): 24649-59, 2015 Sep 22.
Article En | MEDLINE | ID: mdl-26433463

Foxp3(+) regulatory T cells (Tregs) are often highly enriched within the tumor-infiltrating T cell pool. Using a well-characterised model of carcinogen-induced fibrosarcomas we show that the enriched tumor-infiltrating Treg population comprises largely of CXCR3(+) T-bet(+) 'TH1-like' Tregs which are thymus-derived Helios(+) cells. Whilst IL-2 maintains homeostatic ratios of Tregs in lymphoid organs, we found that the perturbation in Treg frequencies in tumors is IL-2 independent. Moreover, we show that the TH1 phenotype of tumor-infiltrating Tregs is dispensable for their ability to influence tumor progression. We did however find that unlike Tconvs, the majority of intra-tumoral Tregs express the activation markers CD69, CD25, ICOS, CD103 and CTLA4 and are significantly more proliferative than Tconvs. Moreover, we have found that CD69(+) Tregs are more suppressive than their CD69- counterparts. Collectively, these data indicate superior activation of Tregs in the tumor microenvironment, promoting their suppressive ability and selective proliferation at this site.


Cell Proliferation , Fibrosarcoma/metabolism , Interleukin-2/metabolism , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/metabolism , Sarcoma, Experimental/metabolism , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Fibrosarcoma/chemically induced , Fibrosarcoma/genetics , Fibrosarcoma/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interleukin-2/antagonists & inhibitors , Interleukin-2/immunology , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Methylcholanthrene , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Sarcoma, Experimental/chemically induced , Sarcoma, Experimental/genetics , Sarcoma, Experimental/immunology , Signal Transduction , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Tumor Microenvironment
6.
Immunology ; 146(1): 11-22, 2015 Sep.
Article En | MEDLINE | ID: mdl-26076649

Analysis of antigen-specific T-cell populations by flow cytometry with peptide-MHC (pMHC) multimers is now commonplace. These reagents allow the tracking and phenotyping of T cells during infection, autoimmunity and cancer, and can be particularly revealing when used for monitoring therapeutic interventions. In 2009, we reviewed a number of 'tricks' that could be used to improve this powerful technology. More recent advances have demonstrated the potential benefits of using higher order multimers and of 'boosting' staining by inclusion of an antibody against the pMHC multimer. These developments now allow staining of T cells where the interaction between the pMHC and the T-cell receptor is over 20-fold weaker (K(D) > 1 mm) than could previously be achieved. Such improvements are particularly relevant when using pMHC multimers to stain anti-cancer or autoimmune T-cell populations, which tend to bear lower affinity T-cell receptors. Here, we update our previous work to include discussion of newer tricks that can produce substantially brighter staining even when using log-fold lower concentrations of pMHC multimer. We further provide a practical guide to using pMHC multimers that includes a description of several common pitfalls and how to circumvent them.


CD8-Positive T-Lymphocytes/immunology , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Staining and Labeling/methods , Antibodies/immunology , CD8-Positive T-Lymphocytes/cytology , Flow Cytometry/methods , Fluorescent Dyes , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Peptides/immunology , Protein Multimerization
7.
J Immunol ; 194(1): 463-74, 2015 Jan 01.
Article En | MEDLINE | ID: mdl-25452566

Fluorochrome-conjugated peptide-MHC (pMHC) multimers are commonly used in combination with flow cytometry for direct ex vivo visualization and characterization of Ag-specific T cells, but these reagents can fail to stain cells when TCR affinity and/or TCR cell-surface density are low. pMHC multimer staining of tumor-specific, autoimmune, or MHC class II-restricted T cells can be particularly challenging, as these T cells tend to express relatively low-affinity TCRs. In this study, we attempted to improve staining using anti-fluorochrome unconjugated primary Abs followed by secondary staining with anti-Ab fluorochrome-conjugated Abs to amplify fluorescence intensity. Unexpectedly, we found that the simple addition of an anti-fluorochrome unconjugated Ab during staining resulted in considerably improved fluorescence intensity with both pMHC tetramers and dextramers and with PE-, allophycocyanin-, or FITC-based reagents. Importantly, when combined with protein kinase inhibitor treatment, Ab stabilization allowed pMHC tetramer staining of T cells even when the cognate TCR-pMHC affinity was extremely low (KD >1 mM) and produced the best results that we have observed to date. We find that this inexpensive addition to pMHC multimer staining protocols also allows improved recovery of cells that have recently been exposed to Ag, improvements in the recovery of self-specific T cells from PBMCs or whole-blood samples, and the use of less reagent during staining. In summary, Ab stabilization of pMHC multimers during T cell staining extends the range of TCR affinities that can be detected, yields considerably enhanced staining intensities, and is compatible with using reduced amounts of these expensive reagents.


Flow Cytometry/methods , Fluorescent Antibody Technique/methods , Receptors, Antigen, T-Cell/immunology , Staining and Labeling/methods , T-Lymphocytes/immunology , Antibodies/chemistry , Antibodies/immunology , Cells, Cultured , Fluorescent Dyes/chemistry , Humans , Phycocyanin/chemistry , Protein Binding/immunology , Protein Kinase Inhibitors/pharmacology , T-Lymphocytes/cytology
8.
Eur J Immunol ; 45(2): 584-91, 2015 Feb.
Article En | MEDLINE | ID: mdl-25471691

MHC anchor residue-modified "heteroclitic" peptides have been used in many cancer vaccine trials and often induce greater immune responses than the wild-type peptide. The best-studied system to date is the decamer MART-1/Melan-A26-35 peptide, EAAGIGILTV, where the natural alanine at position 2 has been modified to leucine to improve human leukocyte antigen (HLA)-A*0201 anchoring. The resulting ELAGIGILTV peptide has been used in many studies. We recently showed that T cells primed with the ELAGIGILTV peptide can fail to recognize the natural tumor-expressed peptide efficiently, thereby providing a potential molecular reason for why clinical trials of this peptide have been unsuccessful. Here, we solved the structure of a TCR in complex with HLA-A*0201-EAAGIGILTV peptide and compared it with its heteroclitic counterpart , HLA-A*0201-ELAGIGILTV. The data demonstrate that a suboptimal anchor residue at position 2 enables the TCR to "pull" the peptide away from the MHC binding groove, facilitating extra contacts with both the peptide and MHC surface. These data explain how a TCR can distinguish between two epitopes that differ by only a single MHC anchor residue and demonstrate how weak MHC anchoring can enable an induced-fit interaction with the TCR. Our findings constitute a novel demonstration of the extreme sensitivity of the TCR to minor alterations in peptide conformation.


Alanine/chemistry , Epitopes, T-Lymphocyte/metabolism , HLA-A2 Antigen/chemistry , Leucine/chemistry , MART-1 Antigen/chemistry , Peptides/chemistry , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Alanine/genetics , Amino Acid Sequence , Amino Acid Substitution , Crystallography, X-Ray , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Leucine/genetics , MART-1 Antigen/genetics , MART-1 Antigen/immunology , Models, Molecular , Molecular Sequence Data , Peptides/genetics , Peptides/immunology , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
9.
J Biol Chem ; 289(2): 628-38, 2014 Jan 10.
Article En | MEDLINE | ID: mdl-24196962

αß T-cell receptors (TCRs) engage antigens using complementarity-determining region (CDR) loops that are either germ line-encoded (CDR1 and CDR2) or somatically rearranged (CDR3). TCR ligands compose a presentation platform (major histocompatibility complex (MHC)) and a variable antigenic component consisting of a short "foreign" peptide. The sequence of events when the TCR engages its peptide-MHC (pMHC) ligand remains unclear. Some studies suggest that the germ line elements of the TCR engage the MHC prior to peptide scanning, but this order of binding is difficult to reconcile with some TCR-pMHC structures. Here, we used TCRs that exhibited enhanced pMHC binding as a result of mutations in either CDR2 and/or CDR3 loops, that bound to the MHC or peptide, respectively, to dissect the roles of these loops in stabilizing TCR-pMHC interactions. Our data show that TCR-peptide interactions play a strongly dominant energetic role providing a binding mode that is both temporally and energetically complementary with a system requiring positive selection by self-pMHC in the thymus and rapid recognition of non-self-pMHC in the periphery.


Complementarity Determining Regions/metabolism , HLA Antigens/metabolism , Peptides/metabolism , Receptors, Antigen, T-Cell/metabolism , Amino Acid Sequence , Binding, Competitive , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Crystallography, X-Ray , HLA Antigens/chemistry , HLA Antigens/genetics , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Humans , Kinetics , Ligands , Models, Molecular , Molecular Sequence Data , Mutation , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , T-Cell Antigen Receptor Specificity
10.
Eur J Immunol ; 43(10): 2613-25, 2013 Oct.
Article En | MEDLINE | ID: mdl-23857287

Balancing the generation of immune responses capable of controlling virus replication with those causing immunopathology is critical for the survival of the host and resolution of influenza-induced inflammation. Based on the capacity of interleukin-6 (IL-6) to govern both optimal T-cell responses and inflammatory resolution, we hypothesised that IL-6 plays an important role in maintaining this balance. Comparison of innate and adaptive immune responses in influenza-infected wild-type control and IL-6-deficient mice revealed striking differences in virus clearance, lung immunopathology and generation of heterosubtypic immunity. Mice lacking IL-6 displayed a profound defect in their ability to mount an anti-viral T-cell response. Failure to adequately control virus was further associated with an enhanced infiltration of inflammatory monocytes into the lung and an elevated production of the pro-inflammatory cytokines, IFN-α and TNF-α. These events were associated with severe lung damage, characterised by profound vascular leakage and death. Our data highlight an essential role for IL-6 in orchestrating anti-viral immunity through an ability to limit inflammation, promote protective adaptive immune responses and prevent fatal immunopathology.


CD4-Positive T-Lymphocytes/immunology , Influenza A virus/physiology , Interleukin-6/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Viral/immunology , Animals , CD4-Positive T-Lymphocytes/virology , Cell Movement/genetics , Cells, Cultured , Cytokines/metabolism , Female , Inflammation Mediators/metabolism , Interleukin-6/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/genetics , Pneumonia, Viral/genetics , Pneumonia, Viral/pathology , Viral Load/genetics , Virus Replication/genetics
11.
Cancer Res ; 72(21): 5473-82, 2012 Nov 01.
Article En | MEDLINE | ID: mdl-22962270

The evolution of immune blockades in tumors limits successful antitumor immunity, but the mechanisms underlying this process are not fully understood. Depletion of regulatory T cells (Treg), a T-cell subset that dampens excessive inflammatory and autoreactive responses, can allow activation of tumor-specific T cells. However, cancer immunotherapy studies have shown that a persistent failure of activated lymphocytes to infiltrate tumors remains a fundamental problem. In evaluating this issue, we found that despite an increase in T-cell activation and proliferation following Treg depletion, there was no significant association with tumor growth rate. In contrast, there was a highly significant association between low tumor growth rate and the extent of T-cell infiltration. Further analyses revealed a total concordance between low tumor growth rate, high T-cell infiltration, and the presence of high endothelial venules (HEV). HEV are blood vessels normally found in secondary lymphoid tissue where they are specialized for lymphocyte recruitment. Thus, our findings suggest that Treg depletion may promote HEV neogenesis, facilitating increased lymphocyte infiltration and destruction of the tumor tissue. These findings are important as they point to a hitherto unidentified role of Tregs, the manipulation of which may refine strategies for more effective cancer immunotherapy.


Chemotaxis, Leukocyte/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Venules/immunology , Animals , Cell Proliferation , Endothelium, Vascular/immunology , Flow Cytometry , Immunohistochemistry , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/pathology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
12.
J Immunol ; 187(2): 654-63, 2011 Jul 15.
Article En | MEDLINE | ID: mdl-21677135

CD8(+) T cells recognize immunogenic peptides presented at the cell surface bound to MHCI molecules. Ag recognition involves the binding of both TCR and CD8 coreceptor to the same peptide-MHCI (pMHCI) ligand. Specificity is determined by the TCR, whereas CD8 mediates effects on Ag sensitivity. Anti-CD8 Abs have been used extensively to examine the role of CD8 in CD8(+) T cell activation. However, as previous studies have yielded conflicting results, it is unclear from the literature whether anti-CD8 Abs per se are capable of inducing effector function. In this article, we report on the ability of seven monoclonal anti-human CD8 Abs to activate six human CD8(+) T cell clones with a total of five different specificities. Six of seven anti-human CD8 Abs tested did not activate CD8(+) T cells. In contrast, one anti-human CD8 Ab, OKT8, induced effector function in all CD8(+) T cells examined. Moreover, OKT8 was found to enhance TCR/pMHCI on-rates and, as a consequence, could be used to improve pMHCI tetramer staining and the visualization of Ag-specific CD8(+) T cells. The anti-mouse CD8 Abs, CT-CD8a and CT-CD8b, also activated CD8(+) T cells despite opposing effects on pMHCI tetramer staining. The observed heterogeneity in the ability of anti-CD8 Abs to trigger T cell effector function provides an explanation for the apparent incongruity observed in previous studies and should be taken into consideration when interpreting results generated with these reagents. Furthermore, the ability of Ab-mediated CD8 engagement to deliver an activation signal underscores the importance of CD8 in CD8(+) T cell signaling.


Antibodies/physiology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytotoxicity, Immunologic , HLA-A Antigens/chemistry , Peptides/chemistry , Receptors, Antigen, T-Cell/deficiency , Antibodies/metabolism , CD8-Positive T-Lymphocytes/cytology , Clone Cells , HLA-A Antigens/immunology , HLA-A2 Antigen , Humans , Immunophenotyping , Ligands , Peptides/analysis , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Staining and Labeling , Surface Plasmon Resonance
13.
Thorax ; 66(5): 368-74, 2011 May.
Article En | MEDLINE | ID: mdl-21310755

BACKGROUND: Seasonal influenza A infection affects a significant cohort of the global population annually, resulting in considerable morbidity and mortality. Therapeutic strategies are of limited efficacy, and during a pandemic outbreak would only be available to a minority of the global population. Over-the-counter medicines are routinely taken by individuals suffering from influenza, but few studies have been conducted to determine their effectiveness in reducing pulmonary immunopathology or the influence they exert upon the generation of protective immunity. METHODS: A mouse model of influenza infection was utilised to assess the efficacy of paracetamol (acetaminophen) in reducing influenza-induced pathology and to examine whether paracetamol affects generation of protective immunity. RESULTS: Administration (intraperitoneal) of paracetamol significantly decreased the infiltration of inflammatory cells into the airway spaces, reduced pulmonary immunopathology associated with acute infection and improved the overall lung function of mice, without adversely affecting the induction of virus-specific adaptive responses. Mice treated with paracetamol exhibited an ability to resist a second infection with heterologous virus comparable with that of untreated mice. CONCLUSIONS: Our results demonstrate that paracetamol dramatically reduces the morbidity associated with influenza but does not compromise the development of adaptive immune responses. Overall, these data support the utility of paracetamol for reducing the clinical symptoms associated with influenza virus infection.


Acetaminophen/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections/drug therapy , Respiratory Tract Infections/drug therapy , Acetaminophen/pharmacology , Adaptive Immunity/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Celecoxib , Cyclooxygenase 2 Inhibitors/therapeutic use , Dinoprostone/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Female , Immunity, Innate/drug effects , Liver/drug effects , Liver/physiopathology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Pyrazoles/therapeutic use , Respiratory Tract Infections/immunology , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Sulfonamides/therapeutic use , Viral Load/drug effects , Virus Shedding/drug effects
14.
Cancer Res ; 71(3): 736-46, 2011 Feb 01.
Article En | MEDLINE | ID: mdl-21156649

A significant enrichment of CD4(+)Foxp3(+) T cells (regulatory T cells, Treg) is frequently observed in murine and human carcinomas. As Tregs can limit effective antitumor immune responses, thereby promoting tumor progression, it is important that the mechanisms underpinning intratumoral accumulation of Tregs are identified. Because of evidence gathered mostly in vitro, the conversion of conventional T cells (Tconv) into Tregs has been proposed as one such mechanism. We assessed the contribution of conversion in vivo by analyzing the TCR (T-cell receptor) repertoires of Tconvs and Tregs in carcinogen-induced tumors in mice. Our results indicate that the TCR repertoires of Tregs and Tconvs within tumor-infiltrating lymphocytes (TIL) are largely distinct. Indeed, the cell population with the greatest degree of repertoire similarity with tumor-infiltrating Tregs was the Treg population from the tumor-draining lymph node. These findings demonstrate that conversion of Tconvs does not contribute significantly to the accumulation of tumor-infiltrating Tregs; rather, Tconvs and Tregs arise from different populations with unique TCR repertoires. Enrichment of Tregs within TILs most likely, therefore, reflects differences in the way that Tregs and Tconvs are influenced by the tumor microenvironment. Elucidating the nature of these influences may indicate how the balance between tumor-infiltrating Tregs and Tconvs can be manipulated for therapeutic purposes.


Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Experimental/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Carcinogens , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Methylcholanthrene , Mice , Mice, Transgenic , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics
15.
J Immunol ; 183(5): 2915-20, 2009 Sep 01.
Article En | MEDLINE | ID: mdl-19667099

Despite its use widely as a therapeutic agent, and proposed use as vaccine adjuvant, the effect of IFNalpha on T cell function is poorly understood. As a pleiotropic innate cytokine produced rapidly in response to pathogens, it is well placed to impinge on specific immune responses. The aim of this study was to examine the impact of IFNalpha on the function of human memory CD4(+) T cells using the recall Ags purified protein derivative, tetanus toxoid, and hemagglutinin. IFNalpha administered either in vivo or added exogenously in vitro tended to enhance proliferative responses of purified protein derivative-specific T cells in marked contrast to the other cognate populations whose responses were often diminished. Purifying the memory CD4(+)CD45RO(+) T cells confirmed IFNalpha acted directly on these cells and not via an intermediate. The T cells could be divided into two broad categories depending on how IFNalpha effected their responses to cognate Ag: 1) enhanced proliferation and a striking increase in IFNgamma-production compared with smaller increases in IL-10 (increased ratio of IFNgamma:IL-10), and 2) neutral or diminished proliferation coupled with a smaller increase in IFNgamma relative to the increase in IL-10 (reduced IFNgamma:IL-10 ratio). IFNalpha has a role in modifying memory T cell responses when they are exposed to cognate Ag and may be important in vaccination strategies designed to augment particular Th memory responses.


CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon-alpha/physiology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/pathology , Humans , Immunologic Memory/drug effects , Injections, Subcutaneous , Interferon-alpha/administration & dosage , Interferon-alpha/therapeutic use , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Tetanus Toxoid/pharmacology , Tuberculin/pharmacology
17.
J Immunol ; 182(9): 5203-7, 2009 May 01.
Article En | MEDLINE | ID: mdl-19380765

CD59, a broadly expressed GPI-anchored molecule, regulates formation of the membrane attack complex of the complement cascade. We previously demonstrated that mouse CD59 also down-modulates CD4(+) T cell activity in vivo. In this study, we explored the role of CD59 on human CD4(+) T cells. Our data demonstrate that CD59 is up-regulated on activated CD4(+) T cells and serves to down-modulate their activity in response to polyclonal and Ag-specific stimulation. The therapeutic potential of this finding was explored using T cells isolated from colorectal cancer patients. The findings were striking and indicated that blockade of CD59 significantly enhanced the CD4(+) T cell response to two different tumor Ags. These data highlight the potential for manipulating CD59 expression on T cells for boosting weak immune responses, such as those found in individuals with cancer.


CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD59 Antigens/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Epitopes, T-Lymphocyte/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/pathology , CD59 Antigens/genetics , CD59 Antigens/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunotherapy, Adoptive , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , U937 Cells , Up-Regulation/genetics , Up-Regulation/immunology
18.
J Immunol Methods ; 340(1): 11-24, 2009 Jan 01.
Article En | MEDLINE | ID: mdl-18929568

Flow cytometry with fluorochrome-conjugated peptide-major histocompatibility complex (pMHC) tetramers has transformed the study of antigen-specific T-cells by enabling their visualization, enumeration, phenotypic characterization and isolation from ex vivo samples. Here, we demonstrate that the reversible protein kinase inhibitor (PKI) dasatinib improves the staining intensity of human (CD8+ and CD4+) and murine T-cells without concomitant increases in background staining. Dasatinib enhances the capture of cognate pMHC tetramers from solution and produces higher intensity staining at lower pMHC concentrations. Furthermore, dasatinib reduces pMHC tetramer-induced cell death and substantially lowers the T-cell receptor (TCR)/pMHC interaction affinity threshold required for cell staining. Accordingly, dasatinib permits the identification of T-cells with very low affinity TCR/pMHC interactions, such as those that typically predominate in tumour-specific responses and autoimmune conditions that are not amenable to detection by current technology.


CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Flow Cytometry/methods , HLA-A2 Antigen/immunology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Adult , Animals , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Dasatinib , HLA-DR Antigens/immunology , Humans , Mice , Mice, Transgenic , Models, Immunological , Staining and Labeling/methods
19.
PLoS Pathog ; 4(2): e1000006, 2008 Feb 29.
Article En | MEDLINE | ID: mdl-18389078

Currently, our understanding of mechanisms underlying cell-mediated immunity and particularly of mechanisms that promote robust T cell memory to respiratory viruses is incomplete. Interleukin (IL)-6 has recently re-emerged as an important regulator of T cell proliferation and survival. Since IL-6 is abundant following infection with influenza virus, we analyzed virus-specific T cell activity in both wild type and IL-6 deficient mice. Studies outlined herein highlight a novel role for IL-6 in the development of T cell memory to influenza virus. Specifically, we find that CD4+ but not CD8+ T cell memory is critically dependent upon IL-6. This effect of IL-6 includes its ability to suppress CD4+CD25+ regulatory T cells (Treg). We demonstrate that influenza-induced IL-6 limits the activity of virus-specific Tregs, thereby facilitating the activity of virus-specific memory CD4+ T cells. These experiments reveal a critical role for IL-6 in ensuring, within the timeframe of an acute infection with a cytopathic virus, that antigen-specific Tregs have no opportunity to down-modulate the immune response, thereby favoring pathogen clearance and survival of the host.


CD4-Positive T-Lymphocytes/immunology , Influenza A virus/immunology , Interleukin-6/physiology , Orthomyxoviridae Infections/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Female , Flow Cytometry , Gene Silencing , Immunologic Memory , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/metabolism , Spleen/drug effects , Spleen/immunology , Spleen/virology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology , Tumor Necrosis Factor-alpha/metabolism , Viral Load
20.
Eur J Immunol ; 37(3): 798-806, 2007 Mar.
Article En | MEDLINE | ID: mdl-17295390

The interaction between T cell receptors (TCR) and peptide-major histocompatibility complex (pMHC) antigens can lead to varying degrees of agonism (T cell activation), or antagonism. The P14 TCR recognises the lymphocytic choriomeningitis virus (LCMV)-derived peptide, gp33 residues 33-41 (KAVYNFATC), presented in the context of H-2D(b). The cellular responses to various related H-2D(b) peptide ligands are very well characterised, and P14 TCR-transgenic mice have been used extensively in models of virus infection, autoimmunity and tumour rejection. Here, we analyse the binding of the P14 soluble TCR to a broad panel of related H-2D(b)-peptide complexes by surface plasmon resonance, and compare this with their diverse cellular responses. P14 TCR binds H-2D(b)-gp33 with a KD of 3 microM (+/-0.5 microM), typical of an immunodominant antiviral TCR, but with unusually fast kinetics (k(off) = 1 s(-1)), corresponding to a half-life of 0.7 s at 25 degrees C, outside the range previously observed for murine agonist TCR/pMHC interactions. The most striking feature of these data is that a very short half-life does not preclude the ability of a TCR/pMHC interaction to induce antiviral immunity, autoimmune disease and tumour rejection.


H-2 Antigens/metabolism , Peptides/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigens, Viral/metabolism , Antigens, Viral/physiology , Glycoproteins/metabolism , Glycoproteins/physiology , H-2 Antigens/physiology , Histocompatibility Antigen H-2D , Ligands , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Peptide Fragments/metabolism , Peptide Fragments/physiology , Peptides/genetics , Peptides/physiology , Protein Binding/immunology , Receptors, Antigen, T-Cell, alpha-beta/physiology , Viral Proteins/metabolism , Viral Proteins/physiology
...