Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1157263, 2023.
Article in English | MEDLINE | ID: mdl-37081876

ABSTRACT

Introduction: The rapid development of vaccines to prevent COVID-19 has raised the need to compare the capacity of different vaccines in terms of developing a protective humoral response. Previous studies have shown inconsistent results in this area, highlighting the importance of further research to evaluate the efficacy of different vaccines. Methods: This study utilized a highly sensitive and reliable flow cytometry method to measure the titers of IgG1 isotype antibodies in the blood of healthy volunteers after receiving one or two doses of various vaccines administered in Spain. The method was also used to simultaneously measure the reactivity of antibodies to the S protein of the original Wuhan strain and variants B.1.1.7 (Alpha), B.1.617.2 (Delta), and B.1.617.1 (Kappa). Results: Significant differences were observed in the titer of anti-S antibodies produced after a first dose of the vaccines ChAdOx1 nCov-19/AstraZeneca, mRNA-1273/Moderna, BNT162b2/Pfizer-BioNTech, and Ad26.COV.S/Janssen. Furthermore, a relative reduction in the reactivity of the sera with the Alpha, Delta, and Kappa variants, compared to the Wuhan strain, was observed after the second boosting immunization. Discussion: The findings of this study provide a comparison of different vaccines in terms of anti-S antibody generation and cast doubts on the convenience of repeated immunization with the same S protein sequence. The multiplexed capacity of the flow cytometry method utilized in this study allowed for a comprehensive evaluation of the efficacy of various vaccines in generating a protective humoral response. Future research could focus on the implications of these findings for the development of effective COVID-19 vaccination strategies.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Antibody Formation , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines , ChAdOx1 nCoV-19 , Spike Glycoprotein, Coronavirus , Vaccination , Antibodies
2.
Cancers (Basel) ; 15(4)2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36831533

ABSTRACT

Harnessing the immune system to fight cancer has become a reality with the clinical success of immune-checkpoint blockade (ICB) antibodies against PD(L)-1 and CTLA-4. However, not all cancer patients respond to ICB. Thus, there is a need to modulate the immune system through alternative strategies for improving clinical responses to ICB. The CD3-T cell receptor (TCR) is the canonical receptor complex on T cells. It provides the "first signal" that initiates T cell activation and determines the specificity of the immune response. The TCR confers the binding specificity whilst the CD3 subunits facilitate signal transduction necessary for T cell activation. While the mechanisms through which antigen sensing and signal transduction occur in the CD3-TCR complex are still under debate, recent revelations regarding the intricate 3D structure of the CD3-TCR complex might open the possibility of modulating its activity by designing targeted drugs and tools, including aptamers. In this review, we summarize the basis of CD3-TCR complex assembly and survey the clinical and preclinical therapeutic tools available to modulate CD3-TCR function for potentiating cancer immunotherapy.

3.
Proc Natl Acad Sci U S A ; 119(22): e2201907119, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35617435

ABSTRACT

Signaling via the T cell receptor (TCR) is critical during the development, maintenance, and activation of T cells. Quantitative aspects of TCR signaling have an important role during positive and negative selection, lineage choice, and ability to respond to small amounts of antigen. By using a mutant mouse line expressing a hypomorphic allele of the CD3ζ chain, we show here that the strength of pre-TCR­mediated signaling during T cell development determines the diversity of the TCRß repertoire available for positive and negative selection, and hence of the final αßTCR repertoire. This finding uncovers an unexpected, pre-TCR signaling­dependent and repertoire­shaping role for ß-selection beyond selection of in-frame rearranged TCRß chains. Our data furthermore support a model of pre-TCR signaling in which the arrangement of this receptor in stable nanoclusters determines its quantitative signaling capacity.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta , T-Lymphocytes , Animals , CD3 Complex/genetics , Cell Differentiation , Mice , Mice, Mutant Strains , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction , T-Lymphocytes/immunology
4.
Front Immunol ; 13: 836516, 2022.
Article in English | MEDLINE | ID: mdl-35401548

ABSTRACT

Background: COVID-19 can generate a broad spectrum of severity and symptoms. Many studies analysed the determinants of severity but not among some types of symptoms. More importantly, very few studies analysed patients highly exposed to the virus that nonetheless remain uninfected. Methods: We analysed serum levels of ACE2, Angiotensin II and anti-Spike antibodies in 2 different cohorts at high risk of viral exposure, highly exposed but uninfected subjects, either high risk health care workers or persons cohabiting with infected close relatives and seropositive patients with symptoms. We tested the ability of the sera of these subjects to neutralize lentivirus pseudotyped with the Spike-protein. Results: We found that the serum levels of ACE2 are significantly higher in highly exposed but uninfected subjects. Moreover, sera from this seronegative persons can neutralize SARS-CoV-2 infection in cellular assays more strongly that sera from non-exposed negative controls eventhough they do not have anti-CoV-2 IgG antibodies suggesting that high levels of ACE2 in serum may somewhat protect against an active infection without generating a conventional antibody response. Finally, we show that among patients with symptoms, ACE2 levels were significantly higher in infected patients who developed cutaneous as compared with respiratory symptoms and ACE2 was also higher in those with milder symptoms. Conclusions: These findings suggest that soluble ACE2 could be used as a potential biomarker to predict SARS-CoV-2 infection risk and to discriminate COVID-19 disease subtypes.


Subject(s)
COVID-19 , Angiotensin-Converting Enzyme 2 , Antibodies, Neutralizing , Antibodies, Viral , Humans , SARS-CoV-2 , Spike Glycoprotein, Coronavirus
5.
Cell Rep ; 34(11): 108861, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33730591

ABSTRACT

T cells form immunological synapses with professional antigen-presenting cells (APCs) resulting in T cell activation and the acquisition of peptide antigen-MHC (pMHC) complexes from the plasma membrane of the APC. They thus become APCs themselves. We investigate the functional outcome of T-T cell antigen presentation by CD4 T cells and find that the antigen-presenting T cells (Tpres) predominantly differentiate into regulatory T cells (Treg), whereas T cells that have been stimulated by Tpres cells predominantly differentiate into Th17 pro-inflammatory cells. Using mice deficient in pMHC uptake by T cells, we show that T-T antigen presentation is important for the development of experimental autoimmune encephalitis and Th17 cell differentiation in vivo. By varying the professional APC:T cell ratio, we can modulate Treg versus Th17 differentiation in vitro and in vivo, suggesting that T-T antigen presentation underlies proinflammatory responses in conditions of antigen scarcity.


Subject(s)
Antigen Presentation/immunology , Antigens/metabolism , Cell Polarity/immunology , Th17 Cells/immunology , Animals , CD28 Antigens/metabolism , Cell Differentiation/immunology , Cell Membrane/metabolism , Dendritic Cells/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , Genome , Histocompatibility Antigens Class II/immunology , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Transcription, Genetic , Trogocytosis , rho GTP-Binding Proteins/deficiency , rho GTP-Binding Proteins/metabolism
6.
EMBO Mol Med ; 13(3): e13549, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33471406

ABSTRACT

A correct identification of seropositive individuals for the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is of paramount relevance to assess the degree of protection of a human population to present and future outbreaks of the COVID-19 pandemic. We describe here a sensitive and quantitative flow cytometry method using the cytometer-friendly non-adherent Jurkat T-cell line that stably expresses the full-length native spike "S" protein of SARS-CoV-2 and a truncated form of the human EGFR that serves a normalizing role. S protein and huEGFRt coding sequences are separated by a T2A self-cleaving sequence, allowing to accurately quantify the presence of anti-S immunoglobulins by calculating a score based on the ratio of fluorescence intensities obtained by double-staining with the test sera and anti-EGFR. The method allows to detect immune individuals regardless of the result of other serological tests or even repeated PCR monitoring. As examples of its use, we show that as much as 28% of the personnel working at the CBMSO in Madrid is already immune. Additionally, we show that anti-S antibodies with protective neutralizing activity are long-lasting and can be detected in sera 8 months after infection.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19 Serological Testing/methods , COVID-19/diagnosis , COVID-19/immunology , Flow Cytometry/methods , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Adult , COVID-19/virology , COVID-19 Serological Testing/statistics & numerical data , Enzyme-Linked Immunosorbent Assay , ErbB Receptors/genetics , Female , Flow Cytometry/statistics & numerical data , Hep G2 Cells , Humans , Jurkat Cells , Male , Middle Aged , Neutralization Tests , Pandemics , Polymerase Chain Reaction , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics
7.
EMBO J ; 39(15): e104749, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32525588

ABSTRACT

CCR5 is not only a coreceptor for HIV-1 infection in CD4+ T cells, but also contributes to their functional fitness. Here, we show that by limiting transcription of specific ceramide synthases, CCR5 signaling reduces ceramide levels and thereby increases T-cell antigen receptor (TCR) nanoclustering in antigen-experienced mouse and human CD4+ T cells. This activity is CCR5-specific and independent of CCR5 co-stimulatory activity. CCR5-deficient mice showed reduced production of high-affinity class-switched antibodies, but only after antigen rechallenge, which implies an impaired memory CD4+ T-cell response. This study identifies a CCR5 function in the generation of CD4+ T-cell memory responses and establishes an antigen-independent mechanism that regulates TCR nanoclustering by altering specific lipid species.


Subject(s)
Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Ceramides/immunology , Immunologic Memory , Receptors, CCR5/deficiency , Animals , Antigens/genetics , CD4-Positive T-Lymphocytes/cytology , Ceramides/genetics , HEK293 Cells , Humans , Mice , Mice, Knockout , Receptors, CCR5/immunology
8.
J Exp Med ; 216(10): 2427-2447, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31324740

ABSTRACT

Signal strength controls the outcome of αß T cell selection in the thymus, resulting in death if the affinity of the rearranged TCR is below the threshold for positive selection, or if the affinity of the TCR is above the threshold for negative selection. Here we show that deletion of the GTPase RRAS2 results in exacerbated negative selection and above-normal expression of positive selection markers. Furthermore, Rras2-/- mice are resistant to autoimmunity both in a model of inflammatory bowel disease (IBD) and in a model of myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE). We show that MOG-specific T cells in Rras2-/- mice have reduced affinity for MOG/I-Ab tetramers, suggesting that enhanced negative selection leads to selection of TCRs with lower affinity for the self-MOG peptide. An analysis of the TCR repertoire shows alterations that mostly affect the TCRα variable (TRAV) locus with specific VJ combinations and CDR3α sequences that are absent in Rras2-/- mice, suggesting their involvement in autoimmunity.


Subject(s)
Clonal Selection, Antigen-Mediated , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor/immunology , Membrane Proteins/immunology , Monomeric GTP-Binding Proteins/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology , Membrane Proteins/genetics , Mice , Mice, Knockout , Monomeric GTP-Binding Proteins/genetics , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Myelin-Oligodendrocyte Glycoprotein/pharmacology
9.
Front Immunol ; 5: 132, 2014.
Article in English | MEDLINE | ID: mdl-24817867

ABSTRACT

T cell activation, initiated by T cell receptor (TCR) mediated recognition of pathogen-derived peptides presented by major histocompatibility complex class I or II molecules (pMHC), shows exquisite specificity and sensitivity, even though the TCR-pMHC binding interaction is of low affinity. Recent experimental work suggests that TCR pre-clustering may be a mechanism via which T cells can achieve such high sensitivity. The unresolved stoichiometry of the TCR makes TCR-pMHC binding and TCR triggering, an open question. We formulate a mathematical model to characterize the pre-clustering of T cell receptors (TCRs) on the surface of T cells, motivated by the experimentally observed distribution of TCR clusters on the surface of naive and memory T cells. We extend a recently introduced stochastic criterion to compute the timescales of T cell responses, assuming that ligand-induced cross-linked TCR is the minimum signaling unit. We derive an approximate formula for the mean time to signal initiation. Our results show that pre-clustering reduces the mean activation time. However, additional mechanisms favoring the existence of clusters are required to explain the difference between naive and memory T cell responses. We discuss the biological implications of our results, and both the compatibility and complementarity of our approach with other existing mathematical models.

10.
J Immunol ; 192(1): 52-8, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24307729

ABSTRACT

Antigenic T cell stimulation requires interaction between the TCR of the T cell and cognate peptide-MHC molecules presented by the APC. Although studies with TCR-specific Abs and soluble peptide-MHC ligands have shown that the TCR needs to be crosslinked by two or more ligands to induce T cell stimulation, it is not understood how several MHC molecules loaded with the cognate antigenic peptide can produce crosslinking under physiological conditions. We show at the molecular level that large clusters of cognate peptide-MHC are formed at the surface of murine professional and nonprofessional APCs upon virus infection and that these clusters impinge on the stimulatory capacity of the APC. These clusters are formed by tight apposition of cognate peptide-MHC complexes in a configuration that is compatible with simultaneous engagement of two or more TCRs. This suggests that physiological expression of Ag allows formation of multivalent ligands for the TCR that permit TCR crosslinking and T cell activation.


Subject(s)
HLA Antigens/immunology , HLA Antigens/metabolism , Peptides/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Antigen Presentation/immunology , Humans , Mice , Peptides/chemistry , Peptides/immunology , Protein Binding/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/virology , Vaccinia virus/immunology
11.
Methods Cell Biol ; 117: 391-410, 2013.
Article in English | MEDLINE | ID: mdl-24143989

ABSTRACT

T cells show high sensitivity for antigen, even though their T-cell antigen receptor (TCR) has a low affinity for its ligand, a major histocompatibility complex molecule presenting a short pathogen-derived peptide. Over the past few years, it has become clear that these paradoxical properties rely at least in part on the organization of cell surface-expressed TCRs in TCR nanoclusters. We describe a protocol, comprising immunogold labeling, cell surface replica generation, and electron microscopy (EM) analysis that allows nanoscale resolution of the distribution of TCRs and other cell surface molecules of cells grown in suspension. Unlike most of the light microscopy-based single-molecule resolution techniques, this technique permits visualization of these molecules on cell surfaces that do not adhere to an experimental support. Given the potential of adhesion-induced receptor redistributions, our technique is a relevant complement to the substrate adherence-dependent techniques. Furthermore, it does not rely on introduction of fluorescently labeled recombinant molecules and therefore allows direct analysis of nonmanipulated primary cells.


Subject(s)
B-Lymphocytes/ultrastructure , Cell Membrane/ultrastructure , Receptors, Antigen, T-Cell/ultrastructure , Staining and Labeling/methods , T-Lymphocytes/ultrastructure , Aluminum Silicates/chemistry , B-Lymphocytes/metabolism , Cell Adhesion , Cell Line , Cell Membrane/metabolism , Freeze Etching , Gene Expression , Gold Colloid/chemistry , Humans , Immunohistochemistry , Microscopy, Electron, Transmission , Protein Multimerization , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Surface Properties , T-Lymphocytes/metabolism
12.
PLoS Comput Biol ; 9(7): e1003102, 2013.
Article in English | MEDLINE | ID: mdl-23935465

ABSTRACT

Conventional and regulatory T cells develop in the thymus where they are exposed to samples of self-peptide MHC (pMHC) ligands. This probabilistic process selects for cells within a range of responsiveness that allows the detection of foreign antigen without excessive responses to self. Regulatory T cells are thought to lie at the higher end of the spectrum of acceptable self-reactivity and play a crucial role in the control of autoimmunity and tolerance to innocuous antigens. While many studies have elucidated key elements influencing lineage commitment, we still lack a full understanding of how thymocytes integrate signals obtained by sampling self-peptides to make fate decisions. To address this problem, we apply stochastic models of signal integration by T cells to data from a study quantifying the development of the two lineages using controllable levels of agonist peptide in the thymus. We find two models are able to explain the observations; one in which T cells continually re-assess fate decisions on the basis of multiple summed proximal signals from TCR-pMHC interactions; and another in which TCR sensitivity is modulated over time, such that contact with the same pMHC ligand may lead to divergent outcomes at different stages of development. Neither model requires that T(conv) and T(reg) are differentially susceptible to deletion or that the two lineages need qualitatively different signals for development, as have been proposed. We find additional support for the variable-sensitivity model, which is able to explain apparently paradoxical observations regarding the effect of partial and strong agonists on T(conv) and T(reg) development.


Subject(s)
Models, Biological , T-Lymphocytes/metabolism , Thymus Gland/immunology , Animals , Cell Lineage , Major Histocompatibility Complex , Mice , T-Lymphocytes/cytology , Thymus Gland/cytology
13.
Immunity ; 35(3): 375-87, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21903423

ABSTRACT

Although memory T cells respond more vigorously to stimulation and they are more sensitive to low doses of antigen than naive T cells, the molecular basis of this increased sensitivity remains unclear. We have previously shown that the T cell receptor (TCR) exists as different-sized oligomers on the surface of resting T cells and that large oligomers are preferentially activated in response to low antigen doses. Through biochemistry and electron microscopy, we now showed that previously stimulated and memory T cells have more and larger TCR oligomers at the cell surface than their naive counterparts. Reconstitution of cells and mice with a point mutant of the CD3ζ subunit, which impairs TCR oligomer formation, demonstrated that the increased size of TCR oligomers was directly responsible for the increased sensitivity of antigen-experienced T cells. Thus, we propose that an "avidity maturation" mechanism underlies T cell antigenic memory.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Oligodeoxyribonucleotides , Receptors, Antigen, T-Cell/immunology , Animals , CD3 Complex/genetics , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/immunology , Receptors, Antigen, T-Cell/genetics
14.
Sci Signal ; 4(161): mr3, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21343616

ABSTRACT

Over the past years, a holistic approach has been applied to the study of the field of receptor signaling, permitting the analysis of how the interaction between receptors and their cellular environment determines receptor function and the study of the role of these receptors, under both normal and pathophysiological conditions, in whole organisms. This has been facilitated by the development of high-resolution microscopy techniques, which allow single-molecule or spatiotemporal resolution, or both, of signaling processes at the cellular and organismal levels. Concurrently, the role of these signaling pathways can be tested in increasingly sophisticated murine disease models. Finally, computational approaches aid in predicting and understanding receptor behavior. The program of the Madrid meeting reflected this integrated approach, highlighting signaling by and dynamics and regulation of immune cell receptors, the T cell receptor and B cell receptor, and signaling by and regulation of G protein-coupled receptors.


Subject(s)
Metabolic Diseases/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Humans , Mice
15.
Sci Signal ; 3(114): pe11, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20332426

ABSTRACT

The T cell antigen receptor (TCR) serves as a paradigm for how membrane receptors transmit signals to the cytoplasm because it controls many aspects of T cell differentiation and function by detecting atom-sized variations in the quality of the ligand that is recognized. The mechanisms that underlie the different signaling outcomes are unclear. Studies that suggest a ligand-tailored, qualitatively different signal are confronted with evidence that favors a quantitative model, and studies of TCR-dependent T cell differentiation in the thymus are no exception. Mature T cells with an alphabeta TCR are classified according to two major distinct subsets based on the mutually exclusive presence of the co-receptors CD4 and CD8, which play essential roles in recognition of the major histocompatibility complex (MHC) class II and I ligands, respectively, and in the recruitment of the tyrosine kinase Lck to the TCR complex. Mature CD4(+) and CD8(+) T cells derive from a common precursor in the thymus, a double-positive (DP) thymocyte, which has both co-receptors. Early signaling models suggested that the differential capacity of CD4 and CD8 to recruit Lck to the TCR underlay lineage decision. A study now shows that differentiation into the CD8(+) lineage requires the TCR-induced increased abundance of the tyrosine kinase zeta chain-associated protein kinase of 70 kD (Zap70). This finding, together with the known importance of Lck in the determination of CD4(+) and CD8(+) lineages, enables us to propose that a balance between the activation of these two kinases by the TCR determines lineage decisions.


Subject(s)
Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Models, Immunological , Signal Transduction/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
16.
Proc Natl Acad Sci U S A ; 103(25): 9625-30, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-16766661

ABSTRACT

T cell antigen receptor (TCR) triggering determines the fate of immature thymocytes. The affinity of the TCR for its endogenous peptide/MHC ligands serves as a signal for positive or negative selection through mechanisms that are still little understood. We have used a conformation-specific antibody to demonstrate that recognition of TCR ligands that lead to negative selection induces a conformational change in the TCR in situ. In contrast, this conformational change is elicited in only a small percentage of immature thymocytes during positive selection. Using a TUNEL assay, we demonstrate that the conformational change in the TCR is strongly linked to activation of programmed cell death in conditions leading to negative selection. Furthermore, the few conformational change-positive thymocytes detected in conditions that preferably lead to positive selection are also TUNEL-positive. Thus, the conformational change in the TCR may underlie the discrimination of ligands leading to positive and negative selection.


Subject(s)
Receptors, Antigen, T-Cell/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Ligands , Mice , Mice, Transgenic , Protein Conformation , Receptors, Antigen, T-Cell/chemistry
17.
EMBO Rep ; 7(5): 490-5, 2006 May.
Article in English | MEDLINE | ID: mdl-16670682

ABSTRACT

The T-cell antigen receptor (TCR x CD3) is a multi-subunit complex that is responsible for triggering an adaptive immune response. It shows high specificity and sensitivity, while having a low affinity for the ligand. Furthermore, T cells respond to antigen over a wide concentration range. The stoichiometry and architecture of TCR x CD3 in the membrane have been under intense scrutiny because they might be the key to explaining its paradoxical properties. This review highlights new evidence that TCR x CD3 is found on intact unstimulated T cells in a monovalent form (one ligand-binding site per receptor) as well as in several distinct multivalent forms. This is in contrast to the TCR x CD3 stoichiometries determined by several biochemical means; however, these data can be explained by the effects of different detergents on the integrity of the receptor. Here, we discuss a model in which the multivalent receptors are important for the detection of low concentrations of ligand and therefore confer sensitivity, whereas the co-expressed monovalent TCR x CD3s allow a wide dynamic range.


Subject(s)
Receptor Aggregation/immunology , Receptors, Antigen, T-Cell/chemistry , Signal Transduction/immunology , Animals , Humans , Protein Conformation , Receptor-CD3 Complex, Antigen, T-Cell/chemistry , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , Receptor-CD3 Complex, Antigen, T-Cell/physiology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/physiology
18.
J Exp Med ; 202(4): 493-503, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16087711

ABSTRACT

A long-standing paradox in the study of T cell antigen recognition is that of the high specificity-low affinity T cell receptor (TCR)-major histocompatibility complex peptide (MHCp) interaction. The existence of multivalent TCRs could resolve this paradox because they can simultaneously improve the avidity observed for monovalent interactions and allow for cooperative effects. We have studied the stoichiometry of the TCR by Blue Native-polyacrylamide gel electrophoresis and found that the TCR exists as a mixture of monovalent (alphabetagammaepsilondeltaepsilonzetazeta) and multivalent complexes with two or more ligand-binding TCRalpha/beta subunits. The coexistence of monovalent and multivalent complexes was confirmed by electron microscopy after label fracture of intact T cells, thus ruling out any possible artifact caused by detergent solubilization. We found that although only the multivalent complexes become phosphorylated at low antigen doses, both multivalent and monovalent TCRs are phosphorylated at higher doses. Thus, the multivalent TCRs could be responsible for sensing low concentrations of antigen, whereas the monovalent TCRs could be responsible for dose-response effects at high concentrations, conditions in which the multivalent TCRs are saturated. Thus, besides resolving TCR stoichiometry, these data can explain how T cells respond to a wide range of MHCp concentrations while maintaining high sensitivity.


Subject(s)
Antigen Presentation/immunology , Antigens/immunology , Major Histocompatibility Complex/immunology , Peptides/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Animals , Cell Line , Cell Proliferation , Dose-Response Relationship, Immunologic , Humans , Hybridomas , Lymphocyte Activation/immunology , Phosphorylation , Rabbits , T-Lymphocytes/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...