Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nat Methods ; 17(10): 1025-1032, 2020 10.
Article in English | MEDLINE | ID: mdl-32929269

ABSTRACT

The immune system's ability to recognize peptides on major histocompatibility molecules contributes to the eradication of cancers and pathogens. Tracking these responses in vivo could help evaluate the efficacy of immune interventions and improve mechanistic understanding of immune responses. For this purpose, we employ synTacs, which are dimeric major histocompatibility molecule scaffolds of defined composition. SynTacs, when labeled with positron-emitting isotopes, can noninvasively image antigen-specific CD8+ T cells in vivo. Using radiolabeled synTacs loaded with the appropriate peptides, we imaged human papillomavirus-specific CD8+ T cells by positron emission tomography in mice bearing human papillomavirus-positive tumors, as well as influenza A virus-specific CD8+ T cells in the lungs of influenza A virus-infected mice. It is thus possible to visualize antigen-specific CD8+ T-cell populations in vivo, which may serve prognostic and diagnostic roles.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Influenza A virus/immunology , Orthomyxoviridae Infections/virology , Papillomaviridae/immunology , Positron-Emission Tomography/methods , Animals , Antigens , Cloning, Molecular , Epitopes/genetics , Epitopes/metabolism , Female , Gene Expression Regulation/immunology , Histocompatibility Antigens Class I/classification , Histocompatibility Antigens Class I/immunology , Humans , Immunoglobulin G/classification , Immunoglobulin G/immunology , Lung/virology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology
2.
Immunology ; 144(4): 631-40, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25333865

ABSTRACT

Type 1 diabetes is characterized by T-cell-mediated destruction of the insulin-producing ß cells in pancreatic islets. A number of islet antigens recognized by CD8 T cells that contribute to disease pathogenesis in non-obese diabetic (NOD) mice have been identified; however, the antigenic specificities of the majority of the islet-infiltrating cells have yet to be determined. The primary goal of the current study was to identify candidate antigens based on the level and specificity of expression of their genes in mouse islets and in the mouse ß cell line MIN6. Peptides derived from the candidates were selected based on their predicted ability to bind H-2K(d) and were examined for recognition by islet-infiltrating T cells from NOD mice. Several proteins, including those encoded by Abcc8, Atp2a2, Pcsk2, Peg3 and Scg2, were validated as antigens in this way. Interestingly, islet-infiltrating T cells were also found to recognize peptides derived from proglucagon, whose expression in pancreatic islets is associated with α cells, which are not usually implicated in type 1 diabetes pathogenesis. However, type 1 diabetes patients have been reported to have serum autoantibodies to glucagon, and NOD mouse studies have shown a decrease in α cell mass during disease pathogenesis. Our finding of islet-infiltrating glucagon-specific T cells is consistent with these reports and suggests the possibility of α cell involvement in development and progression of disease.


Subject(s)
Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Proglucagon/immunology , Animals , Autoantigens/metabolism , Autoimmunity , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Computational Biology , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Enzyme-Linked Immunospot Assay , Epitope Mapping , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , H-2 Antigens/immunology , H-2 Antigens/metabolism , Interferon-gamma/metabolism , Interferon-gamma Release Tests , Islets of Langerhans/metabolism , Mice, Inbred NOD , Proglucagon/metabolism , Protein Binding
3.
Proc Natl Acad Sci U S A ; 108(33): 13682-7, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21825122

ABSTRACT

Peptide-MHC (pMHC) multimers, in addition to being tools for tracking and quantifying antigen-specific T cells, can mediate downstream signaling after T-cell receptor engagement. In the absence of costimulation, this can lead to anergy or apoptosis of cognate T cells, a property that could be exploited in the setting of autoimmune disease. Most studies with class I pMHC multimers used noncovalently linked peptides, which can allow unwanted CD8(+) T-cell activation as a result of peptide transfer to cellular MHC molecules. To circumvent this problem, and given the role of self-reactive CD8(+) T cells in the development of type 1 diabetes, we designed a single-chain pMHC complex (scK(d).IGRP) by using the class I MHC molecule H-2K(d) and a covalently linked peptide derived from islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP(206-214)), a well established autoantigen in NOD mice. X-ray diffraction studies revealed that the peptide is presented in the groove of the MHC molecule in canonical fashion, and it was also demonstrated that scK(d).IGRP tetramers bound specifically to cognate CD8(+) T cells. Tetramer binding induced death of naive T cells and in vitro- and in vivo-differentiated cytotoxic T lymphocytes, and tetramer-treated cytotoxic T lymphocytes showed a diminished IFN-γ response to antigen stimulation. Tetramer accessibility to disease-relevant T cells in vivo was also demonstrated. Our study suggests the potential of single-chain pMHC tetramers as possible therapeutic agents in autoimmune disease. Their ability to affect the fate of naive and activated CD8(+) T cells makes them a potential intervention strategy in early and late stages of disease.


Subject(s)
Autoimmune Diseases/drug therapy , CD8-Positive T-Lymphocytes/drug effects , Histocompatibility Antigens/pharmacology , Peptide Fragments/pharmacology , Animals , Autoantigens , CD8-Positive T-Lymphocytes/immunology , Cell Death/drug effects , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Glucose-6-Phosphatase/immunology , Histocompatibility Antigens/chemistry , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Mice, Transgenic , Peptide Fragments/chemistry , Peptide Fragments/immunology , Protein Multimerization
4.
Int Immunol ; 22(3): 191-203, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20093428

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease characterized by T cell-mediated destruction of insulin-producing pancreatic beta cells. In both humans and the non-obese diabetic (NOD) mouse model of T1D, class II MHC alleles are the primary determinant of disease susceptibility. However, class I MHC genes also influence risk. These findings are consistent with the requirement for both CD4(+) and CD8(+) T cells in the pathogenesis of T1D. Although a large body of work has permitted the identification of multiple mechanisms to explain the diabetes-protective effect of particular class II MHC alleles, studies examining the protective influence of class I alleles are lacking. Here, we explored this question by performing biochemical and structural analyses of the murine class I MHC molecule H-2K(wm7), which exerts a diabetes-protective effect in NOD mice. We have found that H-2K(wm7) molecules are predominantly occupied by the single self-peptide VNDIFERI, derived from the ubiquitous protein histone H2B. This unexpected finding suggests that the inability of H-2K(wm7) to support T1D development could be due, at least in part, to the failure of peptides from critical beta-cell antigens to adequately compete for binding and be presented to T cells. Predominant presentation of a single peptide would also be expected to influence T-cell selection, potentially leading to a reduced ability to select a diabetogenic CD8(+) T-cell repertoire. The report that one of the predominant peptides bound by T1D-protective HLA-A*31 is histone derived suggests the potential translation of our findings to human diabetes-protective class I MHC molecules.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , H-2 Antigens/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Separation , Crystallography , Female , Flow Cytometry , H-2 Antigens/chemistry , H-2 Antigens/genetics , Histones/chemistry , Histones/genetics , Histones/metabolism , Humans , Mass Spectrometry , Mice , Mice, Inbred NOD , Molecular Sequence Data , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Phylogeny , Protein Structure, Quaternary
5.
J Clin Invest ; 131(23)2021 12 01.
Article in English | MEDLINE | ID: mdl-34673568

ABSTRACT

To delineate the in vivo role of different costimulatory signals in activating and expanding highly functional virus-specific cytotoxic CD8+ T cells, we designed synTacs, infusible biologics that recapitulate antigen-specific T cell activation signals delivered by antigen-presenting cells. We constructed synTacs consisting of dimeric Fc-domain scaffolds linking CD28- or 4-1BB-specific ligands to HLA-A2 MHC molecules covalently tethered to HIV- or CMV-derived peptides. Treatment of HIV-infected donor PBMCs with synTacs bearing HIV- or CMV-derived peptides induced vigorous and selective ex vivo expansion of highly functional HIV- and/or CMV-specific CD8+ T cells, respectively, with potent antiviral activities. Intravenous injection of HIV- or CMV-specific synTacs into immunodeficient mice intrasplenically engrafted with donor PBMCs markedly and selectively expanded HIV-specific (32-fold) or CMV-specific (46-fold) human CD8+ T cells populating their spleens. Notably, these expanded HIV- or CMV-specific CD8+ T cells directed potent in vivo suppression of HIV or CMV infections in the humanized mice, providing strong rationale for consideration of synTac-based approaches as a therapeutic strategy to cure HIV and treat CMV and other viral infections. The synTac platform flexibility supports facile delineation of in vivo effects of different costimulatory signals on patient-derived virus-specific CD8+ T cells, enabling optimization of individualized therapies, including HIV cure strategies.


Subject(s)
Cytomegalovirus Infections/metabolism , HIV Infections/metabolism , Immunotherapy/methods , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/virology , Animals , Antigen-Presenting Cells/immunology , Biological Products , CD8-Positive T-Lymphocytes/cytology , Cytomegalovirus , HEK293 Cells , HLA-A2 Antigen/metabolism , Humans , In Vitro Techniques , Jurkat Cells , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Ligands , Mice , Mice, SCID , Peptides , Spleen/metabolism , T-Lymphocytes, Cytotoxic/immunology
6.
Sci Rep ; 11(1): 19220, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34584159

ABSTRACT

Targeted pharmacologic activation of antigen-specific (AgS) T cells may bypass limitations inherent in current T cell-based cancer therapies. We describe two immunotherapeutics platforms for selective delivery of costimulatory ligands and peptide-HLA (pHLA) to AgS T cells. We engineered and deployed on these platforms an affinity-attenuated variant of interleukin-2, which selectively expands oligoclonal and polyfunctional AgS T cells in vitro and synergizes with CD80 signals for superior proliferation versus peptide stimulation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Neoplasms/therapy , Recombinant Fusion Proteins/immunology , Animals , B7-1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , HLA-A Antigens/genetics , HLA-A Antigens/immunology , Humans , Lymphocyte Activation , Mice , Mice, Transgenic , Mutation , Neoplasms/immunology , Peptides/genetics , Peptides/immunology , Primary Cell Culture , Protein Engineering , Recombinant Fusion Proteins/genetics
7.
Clin Cancer Res ; 26(8): 1953-1964, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31964784

ABSTRACT

PURPOSE: To assess the potential for CUE-101, a novel therapeutic fusion protein, to selectively activate and expand HPV16 E711-20-specific CD8+ T cells as an off-the shelf therapy for the treatment of HPV16-driven tumors, including head and neck squamous cell carcinoma (HNSCC), cervical, and anal cancers. EXPERIMENTAL DESIGN: CUE-101 is an Fc fusion protein composed of a human leukocyte antigen (HLA) complex, an HPV16 E7 peptide epitope, reduced affinity human IL2 molecules, and an effector attenuated human IgG1 Fc domain. Human E7-specific T cells and human peripheral blood mononuclear cells (PBMC) were tested to demonstrate cellular activity and specificity of CUE-101, whereas in vivo activity of CUE-101 was assessed in HLA-A2 transgenic mice. Antitumor efficacy with a murine surrogate (mCUE-101) was tested in the TC-1 syngeneic tumor model. RESULTS: CUE-101 demonstrates selective binding, activation, and expansion of HPV16 E711-20-specific CD8+ T cells from PBMCs relative to nontarget cells. Intravenous administration of CUE-101 induced selective expansion of HPV16 E711-20-specific CD8+ T cells in HLA-A2 (AAD) transgenic mice, and anticancer efficacy and immunologic memory was demonstrated in TC-1 tumor-bearing mice treated with mCUE-101. Combination therapy with anti-PD-1 checkpoint blockade further enhanced the observed efficacy. CONCLUSIONS: Consistent with its design, CUE-101 demonstrates selective expansion of an HPV16 E711-20-specific population of cytotoxic CD8+ T cells, a favorable safety profile, and in vitro and in vivo evidence supporting its potential for clinical efficacy in an ongoing phase I trial (NCT03978689).


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HLA-A2 Antigen/immunology , Immunoglobulin Fc Fragments/immunology , Interleukin-2/immunology , Neoplasms/therapy , Papillomavirus E7 Proteins/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Animals , Cells, Cultured , Disease Models, Animal , Female , Healthy Volunteers , Humans , Leukocytes, Mononuclear , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Neoplasms/virology
8.
J Immunol Methods ; 335(1-2): 106-15, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18439618

ABSTRACT

The method described herein provides a novel strategy for the rapid identification of CD8(+) T cell epitopes relevant to type 1 diabetes in the context of the nonobese diabetic (NOD) mouse model of disease. Obtaining the large number of antigen-sensitive monospecific T cells required for conventional antigen discovery methods has historically been problematic due to (1) difficulties in culturing autoreactive CD8(+) T cells from NOD mice and (2) the large time and resource investments required for the generation of transgenic NOD mice. We circumvented these problems by exploiting the rapid generation time of retrogenic (Rg) mice, relative to transgenic mice, as a novel source of sensitive monospecific CD8(+) T cells, using the diabetogenic AI4 T cell receptor on NOD.SCID and NOD.Rag1(-/-) backgrounds as a model. Rg AI4 T cells are diabetogenic in vivo, demonstrating for the first time that Rg mice are a means for assessing the pathogenic potential of CD8(+) T cell receptor specificities. In order to obtain a sufficient number of Rg CD8(+) T cells for antigen screens, we optimized a method for their in vitro culture that resulted in a approximately 500 fold expansion. We demonstrate the high sensitivity and specificity of expanded Rg AI4 T cells in the contexts of (1) specific peptide challenge, (2) islet cytotoxicity, and (3) their ability to resolve previously defined mimotope candidates from a positional scanning peptide library. Our method is the first to combine the speed of Rg technology with an optimized in vitro Rg T cell expansion protocol to enable the rapid discovery of T cell antigens.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Histocompatibility Antigens Class I/analysis , Immunologic Techniques , Islets of Langerhans/immunology , Animals , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Cytotoxicity, Immunologic , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Flow Cytometry , Hematopoietic Stem Cell Transplantation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Peptide Library , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes, Cytotoxic/immunology , Time Factors , Transfection
9.
Expert Rev Clin Immunol ; 6(6): 939-55, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20979558

ABSTRACT

The widely used nonobese diabetic (NOD) mouse model of autoimmune (Type 1) diabetes mellitus shares multiple characteristics with the human disease, and studies employing this model continue to yield clinically relevant and important information. Here, we review some of the recent key findings obtained from NOD mouse investigations that have both advanced our understanding of disease pathogenesis and suggested new therapeutic targets and approaches. Areas discussed include antigen discovery, identification of genes and pathways contributing to disease susceptibility, development of strategies to image islet inflammation and the testing of therapeutics. We also review recent technical advances that, combined with an improved understanding of the NOD mouse model's limitations, should work to ensure its popularity, utility and relevance in the years ahead.


Subject(s)
Autoantigens/immunology , Diabetes Mellitus, Type 1/immunology , Mice, Inbred NOD , Animals , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Disease Models, Animal , Drug Discovery , Genetic Predisposition to Disease , Humans , Immunologic Techniques/trends , Mice
10.
Proc Natl Acad Sci U S A ; 103(33): 12475-80, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16895987

ABSTRACT

Before the onset of autoimmune destruction, type 1 diabetic patients and an animal model, the nonobese diabetic (NOD) mouse, show morphological and functional abnormalities in target organs, which may act as inciting events for leukocyte infiltration. To better understand these abnormalities, but without the complications associated with lymphocytic infiltrates, we examined genes expressed in autoimmune target tissues of NOD/severe combined immunodeficient (scid) mice and of autoimmune-resistant C57BL/6/scid mice. Our results suggest that the NOD genetic background may predispose them to diabetic complications, including insulin resistance in the absence of high circulating glucose levels and without autoimmune destruction of their beta cells. Several of these genes lie within known type 1 and 2 diabetes loci. These data suggest that the NOD mouse may be a good candidate to study an interface between type 1 and type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Genetic Predisposition to Disease , Mice, Inbred NOD , Animals , Blood Glucose/metabolism , Blood Vessels/abnormalities , Diabetes Complications/genetics , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Endoplasmic Reticulum/metabolism , Extracellular Matrix/metabolism , Female , Glucose Tolerance Test , Humans , Insulin/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, SCID , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Protein Folding
SELECTION OF CITATIONS
SEARCH DETAIL