Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nano Lett ; 22(11): 4376-4382, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35616515

ABSTRACT

Autoimmune diseases and in particular type 1 diabetes rely heavily on treatments that target the symptoms rather than prevent the underlying disease. One of the barriers to better therapeutic strategies is the inability to detect and efficiently target rare autoreactive T-cell populations that are major drivers of these conditions. Here, we develop a unique artificial antigen-presenting cell (aAPC) system from biocompatible polymer particles that allows specific encapsulation of bioactive ingredients. Using our aAPC, we demonstrate that we are able to detect rare autoreactive CD4 populations in human patients, and using mouse models, we demonstrate that our particles are able to induce desensitization in the autoreactive population. This system provides a promising tool that can be used in the prevention of autoimmunity before disease onset.


Subject(s)
Diabetes Mellitus, Type 1 , T-Lymphocytes , Animals , Antigen-Presenting Cells , Autoimmunity , CD4-Positive T-Lymphocytes , Diabetes Mellitus, Type 1/therapy , Humans , Mice
2.
Immunity ; 29(3): 497-510, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18789730

ABSTRACT

Little is known about the functional differences between the human skin myeloid dendritic cell (DC) subsets, epidermal CD207(+) Langerhans cells (LCs) and dermal CD14(+) DCs. We showed that CD14(+) DCs primed CD4(+) T cells into cells that induce naive B cells to switch isotype and become plasma cells. In contrast, LCs preferentially induced the differentiation of CD4(+) T cells secreting T helper 2 (Th2) cell cytokines and were efficient at priming and crosspriming naive CD8(+) T cells. A third DC population, CD14(-)CD207(-)CD1a(+) DC, which resides in the dermis, could activate CD8(+) T cells better than CD14(+) DCs but less efficiently than LCs. Thus, the human skin displays three DC subsets, two of which, i.e., CD14(+) DCs and LCs, display functional specializations, the preferential activation of humoral and cellular immunity, respectively.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Langerhans Cells/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cytokines/immunology , Epidermis/immunology , Granzymes/metabolism , Humans , Immunologic Memory , Langerhans Cells/metabolism , Lipopolysaccharide Receptors/immunology , Lymphocyte Activation , Skin/immunology , T-Lymphocytes, Helper-Inducer/immunology
3.
Immunity ; 28(6): 787-98, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18549799

ABSTRACT

The nature of crosspriming immunogens for CD8(+) T cell responses is highly controversial. By using a panel of T cell receptor-like antibodies specific for viral peptides bound to mouse D(b) major histocompatibility complex class I molecules, we show that an exceptional peptide (PA(224-233)) expressed as a viral minigene product formed a sizeable cytosolic pool continuously presented for hours after protein synthesis was inhibited. PA(224-233) pool formation required active cytosolic heat-shock protein 90 but not ER g96 and uniquely enabled crosspriming by this peptide. These findings demonstrate that exceptional class I binding oligopeptides that escape proteolytic degradation are potent crosspriming agents. Thus, the feeble immunogenicity of natural proteasome products in crosspriming can be attributed to their evanescence in donor cells and not an absolute inability of cytosolic oligopeptides to be transferred to and presented by professional antigen-presenting cells.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/metabolism , HSP90 Heat-Shock Proteins/metabolism , Histocompatibility Antigens Class I/immunology , Influenza A virus/immunology , Peptides/immunology , Animals , Antibodies/immunology , Antigens, Viral/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line , Female , HSP90 Heat-Shock Proteins/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Mice , Peptides/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
4.
J Immunol ; 193(11): 5733-43, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25362181

ABSTRACT

Adoptive transfer of Ag-specific T lymphocytes is an attractive form of immunotherapy for cancers. However, acquiring sufficient numbers of host-derived tumor-specific T lymphocytes by selection and expansion is challenging, as these cells may be rare or anergic. Using engineered T cells can overcome this difficulty. Such engineered cells can be generated using a chimeric Ag receptor based on common formats composed from Ag-recognition elements such as αß-TCR genes with the desired specificity, or Ab variable domain fragments fused with T cell-signaling moieties. Combining these recognition elements are Abs that recognize peptide-MHC. Such TCR-like Abs mimic the fine specificity of TCRs and exhibit both the binding properties and kinetics of high-affinity Abs. In this study, we compared the functional properties of engineered T cells expressing a native low affinity αß-TCR chains or high affinity TCR-like Ab-based CAR targeting the same specificity. We isolated high-affinity TCR-like Abs recognizing HLA-A2-WT1Db126 complexes and constructed CAR that was transduced into T cells. Comparative analysis revealed major differences in function and specificity of such CAR-T cells or native TCR toward the same antigenic complex. Whereas the native low-affinity αß-TCR maintained potent cytotoxic activity and specificity, the high-affinity TCR-like Ab CAR exhibited reduced activity and loss of specificity. These results suggest an upper affinity threshold for TCR-based recognition to mediate effective functional outcomes of engineered T cells. The rational design of TCRs and TCR-based constructs may need to be optimized up to a given affinity threshold to achieve optimal T cell function.


Subject(s)
Antibodies/immunology , Cancer Vaccines , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Antigen, T-Cell, alpha-beta/immunology , Recombinant Fusion Proteins/immunology , T-Lymphocytes/physiology , Antibody Affinity , Cytotoxicity, Immunologic , Genetic Engineering , HLA-A2 Antigen/metabolism , Humans , Jurkat Cells , Neoplasms/immunology , Protein Binding , Signal Transduction , T-Cell Antigen Receptor Specificity
5.
J Immunol ; 192(9): 4164-73, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24683185

ABSTRACT

CD74, the cell-surface form of the MHC class II invariant chain, is a key inflammatory factor that is involved in various immune-mediated diseases as part of the macrophage migration inhibitory factor (MIF) binding complex. However, little is known about the natural regulators of CD74 in this context. In order to study the role of the HLA-DR molecule in regulating CD74, we used the HLA-DRα1 domain, which was shown to bind to and downregulate CD74 on CD11b(+) monocytes. We found that DRα1 directly inhibited binding of MIF to CD74 and blocked its downstream inflammatory effects in the spinal cord of mice with experimental autoimmune encephalomyelitis (EAE). Potency of the DRα1 domain could be destroyed by trypsin digestion but enhanced by addition of a peptide extension (myelin oligodendrocyte glycoprotein [MOG]-35-55 peptide) that provided secondary structure not present in DRα1. These data suggest a conformationally sensitive determinant on DRα1-MOG that is responsible for optimal binding to CD74 and antagonism of MIF effects, resulting in reduced axonal damage and reversal of ongoing clinical and histological signs of EAE. These results demonstrate natural antagonist activity of DRα1 for MIF that was strongly potentiated by the MOG peptide extension, resulting in a novel therapeutic, DRα1-MOG-35-55, that within the limitations of the EAE model may have the potential to treat autoimmune diseases such as multiple sclerosis.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , HLA-DR alpha-Chains/metabolism , Histocompatibility Antigens Class II/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Animals , Blotting, Western , Flow Cytometry , Humans , Mice , Mice, Transgenic , Monocytes/metabolism , Spinal Cord/metabolism
6.
J Immunol ; 192(5): 2109-19, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24489091

ABSTRACT

T cell anergy is a key tolerance mechanism to mitigate unwanted T cell activation against self by rendering lymphocytes functionally inactive following Ag encounter. Ag plays an important role in anergy induction where high supraoptimal doses lead to the unresponsive phenotype. How T cells "measure" Ag dose and how this determines functional output to a given antigenic dose remain unclear. Using multiparametric phospho-flow and mass cytometry, we measured the intracellular phosphorylation-dependent signaling events at a single-cell resolution and studied the phosphorylation levels of key proximal human TCR activation- and inhibition-signaling molecules. We show that the intracellular balance and signal integration between these opposing signaling cascades serve as the molecular switch gauging Ag dose. An Ag density of 100 peptide-MHC complexes/cell was found to be the transition point between dominant activation and inhibition cascades, whereas higher Ag doses induced an anergic functional state. Finally, the neutralization of key inhibitory molecules reversed T cell unresponsiveness and enabled maximal T cell functions, even in the presence of very high Ag doses. This mechanism permits T cells to make integrated "measurements" of Ag dose that determine subsequent functional outcomes.


Subject(s)
Antigens/immunology , Clonal Anergy/physiology , Lymphocyte Activation/physiology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Antigens/pharmacology , Cell Line, Transformed , Clonal Anergy/drug effects , Dose-Response Relationship, Immunologic , HLA Antigens/immunology , Humans , Lymphocyte Activation/drug effects , Signal Transduction/drug effects , T-Lymphocytes/cytology
7.
Proc Natl Acad Sci U S A ; 109(46): 18885-90, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23112154

ABSTRACT

Human Langerhans cells (LCs) are highly efficient at priming cytolytic CD8(+) T cells compared with dermal CD14(+) dendritic cells (DCs). Here we show that dermal CD14(+) DCs instead prime a fraction of naïve CD8(+) T cells into cells sharing the properties of type 2 cytokine-secreting CD8(+) T cells (TC2). Differential expression of the CD8-antagonist receptors on dermal CD14(+) DCs, the Ig-like transcript (ILT) inhibitory receptors, explains the difference between the two types of DCs. Inhibition of CD8 function on LCs inhibited cytotoxic T lymphocytes (CTLs) and enhanced TC2 generation. In addition, blocking ILT2 or ILT4 on dermal CD14(+) DCs enhanced the generation of CTLs and inhibited TC2 cytokine production. Lastly, addition of soluble ILT2 and ILT4 receptors inhibited CTL priming by LCs. Thus, ILT receptor expression explains the polarization of CD8(+) T-cell responses by LCs vs. dermal CD14(+) DCs.


Subject(s)
Antigens, CD/immunology , Dermis/immunology , Langerhans Cells/metabolism , Lipopolysaccharide Receptors , Membrane Glycoproteins/immunology , Receptors, Immunologic/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, CD/biosynthesis , Antigens, CD/genetics , Dermis/cytology , Dermis/metabolism , Humans , Langerhans Cells/cytology , Langerhans Cells/immunology , Leukocyte Immunoglobulin-like Receptor B1 , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/metabolism
8.
J Pept Sci ; 20(6): 446-50, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24733719

ABSTRACT

The vast potential applications of biomolecules that bind inorganic surfaces led mostly to the isolation of short peptides that target selectively specific materials. The demonstrated differential affinity toward certain surfaces created the impression that the recognition capacity of short peptides may match that of rigid biomolecules. In the following, we challenge this view by comparing the capacity of antibody molecules to discriminate between the (100) and (111A) facets of a gallium arsenide semiconductor crystal with the capacity of short peptides to do the same. Applying selection from several peptide and single chain phage display libraries, we find a number of antibody molecules that bind preferentially a given crystal facet but fail to isolate, in dozens of attempts, a single peptide capable of such recognition. The experiments underscore the importance of rigidity to the recognition of inorganic flat targets and therefore set limitations on potential applications of short peptides in biomimetics.


Subject(s)
Antibodies/chemistry , Arsenicals/chemistry , Gallium/chemistry , Oligopeptides/chemistry , Antibodies/immunology , Arsenicals/immunology , Enzyme-Linked Immunosorbent Assay , Gallium/immunology , Semiconductors , Surface Properties
9.
J Exp Med ; 204(5): 1131-44, 2007 May 14.
Article in English | MEDLINE | ID: mdl-17485514

ABSTRACT

CD1d-restricted lymphocytes recognize a broad lipid range. However, how CD1d-restricted lymphocytes translate T cell receptor (TCR) recognition of lipids with similar group heads into distinct biological responses remains unclear. Using a soluble invariant NKT (iNKT) TCR and a newly engineered antibody specific for alpha-galactosylceramide (alpha-GalCer)-human CD1d (hCD1d) complexes, we measured the affinity of binding of iNKT TCR to hCD1d molecules loaded with a panel of alpha-GalCer analogues and assessed the rate of dissociation of alpha-GalCer and alpha-GalCer analogues from hCD1d molecules. We extended this analysis by studying iNKT cell synapse formation and iNKT cell activation by the same panel of alpha-GalCer analogues. Our results indicate the unique role of the lipid chain occupying the hCD1d F' channel in modulating TCR binding affinity to hCD1d-lipid complexes, the formation of stable immunological synapse, and cell activation. These data are consistent with previously described conformational changes between empty and loaded hCD1d molecules (Koch, M., V.S. Stronge, D. Shepherd, S.D. Gadola, B. Mathew, G. Ritter, A.R. Fersht, G.S. Besra, R.R. Schmidt, E.Y. Jones, and V. Cerundolo. 2005. Nat. Immunol 6:819-826), suggesting that incomplete occupation of the hCD1d F' channel results in conformational differences at the TCR recognition surface. This indirect effect provides a general mechanism by which lipid-specific lymphocytes are capable of recognizing both the group head and the length of lipid antigens, ensuring greater specificity of antigen recognition.


Subject(s)
Antigens, CD1/metabolism , Glycolipids/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Models, Molecular , Receptors, Antigen, T-Cell/metabolism , Antigens, CD1d , Calcium/metabolism , Cell Line , Enzyme-Linked Immunosorbent Assay , Galactosylceramides/immunology , Galactosylceramides/metabolism , Humans , Molecular Structure , Protein Binding
10.
Eur J Immunol ; 42(4): 842-50, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22531911

ABSTRACT

Short-lived protein translation products are proposed to be a major source of substrates for major histocompatibility complex (MHC) class I antigen processing and presentation; however, a direct link between protein stability and the presentation level of MHC class I-peptide complexes has not been made. We have recently discovered that the peptide Tyr((369-377)) , derived from the tyrosinase protein is highly presented by HLA-A2 on the surface of melanoma cells. To examine the molecular mechanisms responsible for this presentation, we compared characteristics of tyrosinase in melanoma cells lines that present high or low levels of HLA-A2-Tyr((369-377)) complexes. We found no correlation between mRNA levels and the levels of HLA-A2-Tyr((369-377)) presentation. Co-localization experiments revealed that, in cell lines presenting low levels of HLA-A2-Tyr((369-377)) complexes, tyrosinase co-localizes with LAMP-1, a melanosome marker, whereas in cell lines presenting high HLA-A2-Tyr((369-377)) levels, tyrosinase localizes to the endoplasmic reticulum. We also observed differences in tyrosinase molecular weight and glycosylation composition as well as major differences in protein stability (t(1/2) ). By stabilizing the tyrosinase protein, we observed a dramatic decrease in HLA-A2-tyrosinase presentation. Our findings suggest that aberrant processing and instability of tyrosinase are responsible for the high presentation of HLA-A2-Tyr((369-377)) complexes and thus shed new light on the relationship between intracellular processing, stability of proteins, and MHC-restricted peptide presentation.


Subject(s)
Antigen Presentation , HLA-A2 Antigen/immunology , Melanoma/immunology , Monophenol Monooxygenase/immunology , Neoplasm Proteins/immunology , Cell Line, Tumor , Glycosylation , HLA-A2 Antigen/metabolism , Humans , Lysosomal Membrane Proteins/immunology , Lysosomal Membrane Proteins/metabolism , Melanoma/metabolism , Melanosomes/immunology , Melanosomes/metabolism , Monophenol Monooxygenase/metabolism , Neoplasm Proteins/metabolism , Peptides/immunology , Peptides/metabolism , Protein Stability , RNA, Messenger/immunology , RNA, Messenger/metabolism
11.
J Autoimmun ; 47: 83-93, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24090977

ABSTRACT

The trimolecular complex composed of autoreactive T-cell receptor, MHC class II, and an autoantigenic peptide plays a central role in the activation of pathogenic Islet-specific CD4+ T cells in type 1 diabetes (T1D). We isolated and characterized novel antibodies against autoreactive T-cell epitopes associated with T1D. Our antibodies mimic the specificity of the T-cell receptor (TCR), while binding MHC class II/peptide complexes in an autoantigen peptide specific, MHC-restricted manner. The isolated TCR-like antibodies were directed against the minimal T-cell epitope GAD-555-567 in the context of the HLA-DR4-diabetic-associated molecule. A representative high-affinity TCR-like antibody clone (G3H8) enabled the detection of intra- and extra-cellular DR4/GAD-555-567 complexes in antigen presenting cells. I561M single mutation at the central position (P5) of the GAD-555-567 peptide abolished the binding of G3H8 to the DR4/GAD complex, demonstrating its high fine TCR-like specificity. The G3H8 TCR-like antibody significantly inhibited GAD-555-567 specific, DR4 restricted T-cell response in vitro and in vivo in HLA-DR4 transgenic mice. Our findings constitute a proof-of-concept for the utility of TCR-like antibodies as antigen-specific immunomodulation agents for regulating pathogenic T-cells and suggest that TCR-like antibodies targeting autoreactive MHC class II epitopes are valuable research tools that enable studies related to antigen presentation as well as novel therapeutic agents that may be used to modulate autoimmune disorders such as T1D.


Subject(s)
Antibodies/therapeutic use , Diabetes Mellitus, Type 1/therapy , Epitopes, T-Lymphocyte/immunology , Glutamate Decarboxylase/immunology , Immunomodulation , Receptors, Antigen, T-Cell/immunology , Recombinant Proteins/therapeutic use , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cell Proliferation , Epitopes, T-Lymphocyte/genetics , Glutamate Decarboxylase/genetics , HEK293 Cells , HLA-DR4 Antigen/genetics , Humans , Immunoglobulin G/immunology , Interleukin-2/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/genetics , Peptide Fragments/immunology , Protein Binding/genetics , Protein Binding/immunology
12.
J Autoimmun ; 40: 96-110, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23026773

ABSTRACT

Treatment with partial (p)MHC class II-ß1α1 constructs (also referred to as recombinant T-cell receptor ligands - RTL) linked to antigenic peptides can induce T-cell tolerance, inhibit recruitment of inflammatory cells and reverse autoimmune diseases. Here we demonstrate a novel regulatory pathway that involves RTL binding to CD11b(+) mononuclear cells through a receptor comprised of MHC class II invariant chain (CD74), cell-surface histones and MHC class II itself for treatment of experimental autoimmune encephalomyelitis (EAE). Binding of RTL constructs with CD74 involved a previously unrecognized MHC class II-α1/CD74 interaction that inhibited CD74 expression, blocked activity of its ligand, macrophage migration inhibitory factor, and reduced EAE severity. These findings implicate binding of RTL constructs to CD74 as a key step in both antigen-driven and bystander T-cell tolerance important in treatment of inflammatory diseases.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Autoimmune Diseases/immunology , Histocompatibility Antigens Class II/metabolism , Monocytes/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/biosynthesis , CD11b Antigen/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Histocompatibility Antigens Class II/biosynthesis , Histocompatibility Antigens Class II/immunology , Immune Tolerance , Intramolecular Oxidoreductases , Macrophage Migration-Inhibitory Factors , Membrane Proteins , Mice , Receptors, Antigen, T-Cell , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/immunology
13.
Nano Lett ; 12(9): 4992-6, 2012 Sep 12.
Article in English | MEDLINE | ID: mdl-22900991

ABSTRACT

Nanoscale organization of surface ligands often has a critical effect on cell-surface interactions. We have developed an experimental system that allows a high degree of control over the 2-D spatial distribution of ligands. As a proof of concept, we used the developed system to study how T-cell activation is independently affected by antigen density and antigen amount per cell. Arrays of submicrometer gold islands at varying surface coverage were defined on silicon by electron beam lithography (EBL). The gold islands were functionalized with alkanethiol self-assembled monolayers (SAMs) containing a small antigen, 2,4,6-trinotrophenyl (TNP), at various densities. Genetically engineered T-cell hybridomas expressing TNP-specific chimeric T-cell antigen receptor (CAR) were cultured on the SAMs, and their activation was assessed by IL-2 secretion and CD69 expression. It was found that, at constant antigen density, activation increased monotonically with the amount of antigen, while at constant antigen amount activation was maximal at an intermediate antigen density, whose value was independent of the amount of antigen.


Subject(s)
Alkanes/chemistry , Gold/chemistry , Immunoassay/methods , Nanoparticles/chemistry , Protein Array Analysis/methods , Protein Interaction Mapping/methods , Sulfhydryl Compounds/chemistry , Materials Testing , Molecular Imprinting/methods , Nanoparticles/ultrastructure
14.
Eur J Immunol ; 41(5): 1465-79, 2011 May.
Article in English | MEDLINE | ID: mdl-21469129

ABSTRACT

Antigen-presenting cell-associated four-domain MHC class II (MHC-II) molecules play a central role in activating autoreactive CD4(+) T cells involved in multiple sclerosis (MS) and type 1 diabetes (T1D). In contrast, two-domain MHC-II structures with the same covalently attached self-peptide (recombinant T-cell receptor ligands (RTLs)) can regulate pathogenic CD4(+) T cells and reverse clinical signs of experimental autoimmune diseases. RTL1000, which is composed of the ß1α1 domains of human leukocyte antigen (HLA)-DR2 linked to the encephalitogenic human myelin oligodendrocyte glycoprotein (MOG)-35-55 peptide, was recently shown to be safe and well tolerated in a phase I clinical trial in MS. To evaluate the opposing biological effects of four- versus two-domain MHC-II structures, we screened phage Fab antibodies (Abs) for the neutralizing activity of RTL1000. Five different TCR-like Abs were identified that could distinguish between the two- versus four-domain MHC-peptide complexes while the cognate TCR was unable to make such a distinction. Moreover, Fab detection of native two-domain HLA-DR structures in human plasma implies that there are naturally occurring regulatory MHC-peptide complexes. These results demonstrate for the first time distinct conformational determinants characteristic of activating versus tolerogenic MHC-peptide complexes involved in human autoimmunity.


Subject(s)
Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Autoimmunity/immunology , Diabetes Mellitus, Type 1/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glycoproteins , HLA-DR2 Antigen/chemistry , HLA-DR2 Antigen/genetics , HLA-DR2 Antigen/immunology , Humans , Immune Tolerance , Immunoglobulin Fab Fragments/immunology , Major Histocompatibility Complex/immunology , Mice , Mice, Transgenic , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/immunology , Recombinant Proteins/immunology
15.
Expert Rev Mol Med ; 14: e6, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22361332

ABSTRACT

Tumour and virus-infected cells are recognised by CD8+ cytotoxic T cells that, in response, are activated to eliminate these cells. In order to be activated, the clonotypic T-cell receptor (TCR) needs to encounter a specific peptide antigen presented by the membrane surface major histocompatibility complex (MHC) molecule. Cells that have undergone malignant transformation or viral infection present peptides derived from tumour-associated antigens or viral proteins on their MHC class I molecules. Therefore, disease-specific MHC-peptide complexes are desirable targets for immunotherapeutic approaches. One such approach transforms the unique fine specificity but low intrinsic affinity of TCRs to MHC-peptide complexes into high-affinity soluble antibody molecules endowed with a TCR-like specificity towards tumour or viral epitopes. These antibodies, termed TCR-like antibodies, are being developed as a new class of immunotherapeutics that can target tumour and virus-infected cells and mediate their specific killing. In addition to their therapeutic capabilities, TCR-like antibodies are being developed as diagnostic reagents for cancer and infectious diseases, and serve as valuable research tools for studying MHC class I antigen presentation.


Subject(s)
Antigens, Neoplasm/therapeutic use , Immunotherapy/methods , Neoplasms/drug therapy , Receptors, Antigen, T-Cell , Virus Diseases/drug therapy , Animals , Antibody Affinity/immunology , Antigen Presentation , Antigens, Neoplasm/immunology , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Histocompatibility Antigens Class I/immunology , Humans , Neoplasm Proteins/immunology , Neoplasms/immunology , Neoplasms/pathology , Peptides/immunology , Viral Proteins/immunology , Virus Diseases/immunology , Virus Diseases/pathology
16.
Blood ; 116(10): 1685-97, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20530286

ABSTRACT

We evaluated human CD8(+) T-cell responses generated by targeting antigens to dendritic cells (DCs) through various lectin receptors. We found the immunoreceptor tyrosine-based inhibitory motif-containing DC immunoreceptor (DCIR) to mediate potent cross-presentation. A single exposure to a low dose of anti-DCIR-antigen conjugate initiated antigen-specific CD8(+) T-cell immunity by all human DC subsets including ex vivo-generated DCs, skin-isolated Langerhans cells, and blood myeloid DCs and plasmacytoid DCs. The delivery of influenza matrix protein (FluMP) through DCIR resulted in expansion of FluMP-specific memory CD8(+) T cells. Enhanced specific CD8(+) T-cell responses were observed when an antigen was delivered to the DCs via DCIR, compared with those induced by a free antigen, or antigen conjugated to a control monoclonal antibody or delivered via DC-SIGN, another lectin receptor. DCIR targeting also induced primary CD8(+) T-cell responses against self (MART-1) and viral (HIV gag) antigens. Addition of Toll-like receptor (TLR) 7/8 agonist enhanced DCIR-mediated cross-presentation as well as cross-priming, particularly when combined with a CD40 signal. TLR7/8 activation was associated with increased expansion of the primed CD8(+) T cells, high production of interferon-γ and tumor necrosis factor-α, and reduced levels of type 2-associated cytokines. Thus, antigen targeting via the human DCIR receptor allows activation of specific CD8(+) T-cell immunity.


Subject(s)
Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lectins, C-Type/immunology , Membrane Glycoproteins/immunology , Receptors, Immunologic/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cross-Priming/drug effects , Cross-Priming/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Flow Cytometry , Humans , Langerhans Cells/cytology , Langerhans Cells/immunology , Langerhans Cells/metabolism , Lectins, C-Type/metabolism , MART-1 Antigen , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Neoplasm Proteins/immunology , Quinolines/pharmacology , Receptors, Immunologic/metabolism , Thiazoles/pharmacology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , gag Gene Products, Human Immunodeficiency Virus/immunology
17.
Nano Lett ; 11(11): 4997-5001, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-21985491

ABSTRACT

Seamless embedment of electronic devices in biological systems is expected to add the outstanding computing power, memory, and speed of electronics to the biochemical toolbox of nature. Such amalgamation requires transduction of electronic signals into biochemical cues that affect cells. Inspired by biology, where pathways are directed by molecular recognition, we propose and demonstrate a generic electrical-to-biological transducer comprising a two-state electronic antigen and a chimeric cell receptor engineered to bind the antigen exclusively in its "on" state. T-cells expressing these receptors remain inactivated with the antigen in its "off" state. Switching the antigen to its "on" state by an electrical signal leads to its recognition by the T-cells and correspondingly to cell activation.


Subject(s)
Receptors, Antigen, T-Cell/radiation effects , Single-Chain Antibodies/radiation effects , T-Lymphocytes/radiation effects , Cells, Cultured , Electromagnetic Fields , Humans , Receptors, Antigen, T-Cell/chemistry , Single-Chain Antibodies/chemistry , T-Lymphocytes/chemistry
18.
Cells ; 12(1)2022 12 21.
Article in English | MEDLINE | ID: mdl-36611821

ABSTRACT

The impressive clinical success of cancer immunotherapy has motivated the continued search for new targets that may serve to guide potent effector functions in an attempt to efficiently kill malignant cells. The intracellular proteome is an interesting source for such new targets, such as neo-antigens and others, with growing interest in their application for cell-based immunotherapies. These intracellular-derived targets are peptides presented by MHC class I molecules on the cell surface of malignant cells. These disease-specific class I HLA-peptide complexes can be targeted by specific TCRs or by antibodies that mimic TCR-specificity, termed TCR-like (TCRL) antibodies. Adoptive cell transfer of TCR engineered T cells and T-cell-receptor-like based CAR-T cells, targeted against a peptide-MHC of interest, are currently tested as cancer therapeutic agents in pre-clinical and clinical trials, along with soluble TCR- and TCRL-based agents, such as immunotoxins and bi-specific T cell engagers. Targeting the intracellular proteome using TCRL- and TCR-based molecules shows promising results in cancer immunotherapy, as exemplified by the success of the anti-gp100/HLA-A2 TCR-based T cell engager, recently approved by the FDA for the treatment of unresectable or metastatic uveal melanoma. This review is focused on the selection and isolation processes of TCR- and TCRL-based targeting moieties, with a spotlight on pre-clinical and clinical studies, examining peptide-MHC targeting agents in cancer immunotherapy.


Subject(s)
Melanoma , Receptors, Chimeric Antigen , Humans , T-Lymphocytes , Receptors, Chimeric Antigen/metabolism , Proteome/metabolism , Receptors, Antigen, T-Cell/metabolism , Immunotherapy , Peptides/metabolism , Melanoma/metabolism , Antibodies/metabolism
19.
Eur J Immunol ; 40(6): 1552-65, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20306470

ABSTRACT

There are no direct means to study class I MHC presentation in human normal or diseased cells. Using CMV-infected human cells and applying novel mAb that mimic T-cell receptor specificity directed toward the immunogenic epitope of the viral pp65 protein presented on HLA-A2 molecules, we directly imaged the dynamics of Ag presentation in infected cells. We demonstrate that following infection large intracellular pools of HLA-A2/pp65 complexes are localized to the Golgi. These HLA-A2/pp65 pools account for the majority of total HLA-A2 molecules in infected cells. Interestingly, these large pools are sequestered inside infected cells and only a small portion of them are exported to the cell surface. Virus-induced class I MHC down-regulation did not affect the intracellular pool of HLA-A2/pp65 complexes. Our data also suggest that proteasome function influences the release of class I complexes to the membrane. We present herein a new and direct molecular tool to study the dynamics of viral Ag presentation that may further elucidate the balance between immune response versus viral escape.


Subject(s)
Antibodies, Monoclonal , Antigen Presentation/immunology , Cytomegalovirus Infections/immunology , HLA-A2 Antigen/immunology , Phosphoproteins/immunology , Viral Matrix Proteins/immunology , Antibodies, Viral/immunology , Cell Separation , Cytomegalovirus Infections/metabolism , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , HLA-A2 Antigen/metabolism , Humans , Microscopy, Confocal , Molecular Mimicry , Phosphoproteins/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity/immunology , Viral Matrix Proteins/metabolism
20.
J Immunol ; 182(10): 6328-41, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414786

ABSTRACT

Peptide Ags presented by class I MHC molecules on human melanomas and that are recognized by CD8(+) T cells are the subjects of many studies of antitumor immunity and represent attractive candidates for therapeutic approaches. However, no direct quantitative measurements exist to reveal their expression hierarchy on the cell surface. Using novel recombinant Abs which bind these Ags with a peptide-specific, MHC-restricted manner, we demonstrate a defined pattern of expression hierarchy of peptide-HLA-A2 complexes derived from three major differentiation Ags: gp100, Melan-A/Mart-1, and tyrosinase. Studying melanoma cell lines derived from multiple patients, we reveal a surprisingly high level of presentation of tyrosinase-derived complexes and moderate to very low expression of complexes derived from other Ags. No correlation between Ag presentation and mRNA expression was found; however, protein stability may play a major role. These results provide new insights into the characteristics of Ag presentation and are particularly important when such targets are being considered for immunotherapy. These results may shed new light on relationships between Ag presentation and immune response to cancer Ags.


Subject(s)
Antigen Presentation/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Melanoma/immunology , Monophenol Monooxygenase/immunology , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Humans , MART-1 Antigen , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , gp100 Melanoma Antigen
SELECTION OF CITATIONS
SEARCH DETAIL