Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Elife ; 122024 Apr 19.
Article En | MEDLINE | ID: mdl-38639990

CD4+ T cell activation is driven by five-module receptor complexes. The T cell receptor (TCR) is the receptor module that binds composite surfaces of peptide antigens embedded within MHCII molecules (pMHCII). It associates with three signaling modules (CD3γε, CD3δε, and CD3ζζ) to form TCR-CD3 complexes. CD4 is the coreceptor module. It reciprocally associates with TCR-CD3-pMHCII assemblies on the outside of a CD4+ T cells and with the Src kinase, LCK, on the inside. Previously, we reported that the CD4 transmembrane GGXXG and cytoplasmic juxtamembrane (C/F)CV+C motifs found in eutherian (placental mammal) CD4 have constituent residues that evolved under purifying selection (Lee et al., 2022). Expressing mutants of these motifs together in T cell hybridomas increased CD4-LCK association but reduced CD3ζ, ZAP70, and PLCγ1 phosphorylation levels, as well as IL-2 production, in response to agonist pMHCII. Because these mutants preferentially localized CD4-LCK pairs to non-raft membrane fractions, one explanation for our results was that they impaired proximal signaling by sequestering LCK away from TCR-CD3. An alternative hypothesis is that the mutations directly impacted signaling because the motifs normally play an LCK-independent role in signaling. The goal of this study was to discriminate between these possibilities. Using T cell hybridomas, our results indicate that: intracellular CD4-LCK interactions are not necessary for pMHCII-specific signal initiation; the GGXXG and (C/F)CV+C motifs are key determinants of CD4-mediated pMHCII-specific signal amplification; the GGXXG and (C/F)CV+C motifs exert their functions independently of direct CD4-LCK association. These data provide a mechanistic explanation for why residues within these motifs are under purifying selection in jawed vertebrates. The results are also important to consider for biomimetic engineering of synthetic receptors.


Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , Placenta , Pregnancy , Animals , Female , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Placenta/metabolism , Signal Transduction/genetics , Receptors, Antigen, T-Cell/metabolism , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , Phosphorylation , CD4 Antigens , Mammals/metabolism
2.
bioRxiv ; 2023 Nov 29.
Article En | MEDLINE | ID: mdl-37214965

CD4+ T cell activation is driven by 5-module receptor complexes. The T cell receptor (TCR) is the receptor module that binds composite surfaces of peptide antigens embedded within MHCII molecules (pMHCII). It associates with three signaling modules (CD3γε, CD3δε, and CD3ζζ) to form TCR-CD3 complexes. CD4 is the coreceptor module. It reciprocally associates with TCR-CD3-pMHCII assemblies on the outside of a CD4+ T cells and with the Src kinase, LCK, on the inside. Previously, we reported that the CD4 transmembrane GGXXG and cytoplasmic juxtamembrane (C/F)CV+C motifs found in eutherian (placental mammal) CD4 have constituent residues that evolved under purifying selection (Lee, et al., 2022). Expressing mutants of these motifs together in T cell hybridomas increased CD4-LCK association but reduced CD3ζ, ZAP70, and PLCγ1 phosphorylation levels, as well as IL-2 production, in response to agonist pMHCII. Because these mutants preferentially localized CD4-LCK pairs to non-raft membrane fractions, one explanation for our results was that they impaired proximal signaling by sequestering LCK away from TCR-CD3. An alternative hypothesis is that the mutations directly impacted signaling because the motifs normally play an LCK-independent role in signaling. The goal of this study was to discriminate between these possibilities. Using T cell hybridomas, our results indicate that: intracellular CD4-LCK interactions are not necessary for pMHCII-specific signal initiation; the GGXXG and (C/F)CV+C motifs are key determinants of CD4-mediated pMHCII-specific signal amplification; the GGXXG and (C/F)CV+C motifs exert their functions independently of direct CD4-LCK association. These data provide a mechanistic explanation for why residues within these motifs are under purifying selection in jawed vertebrates. The results are also important to consider for biomimetic engineering of synthetic receptors.

3.
J Immunol ; 209(10): 2033-2041, 2022 11 15.
Article En | MEDLINE | ID: mdl-36426940

T cells play a central role in adaptive immunity by recognizing peptide Ags presented by MHC molecules (pMHC) via their clonotypic TCRs. αßTCRs are heterodimers, consisting of TCRα and TCRß subunits that are composed of variable (Vα, Vß) and constant (Cα, Cß) domains. Whereas the Vα, Vß, and Cß domains adopt typical Ig folds in the extracellular space, the Cα domain lacks a top ß sheet and instead has two loosely associated top strands (C- and F-strands) on its surface. Previous results suggest that this unique Ig-like fold mediates homotypic TCR interactions and influences signaling in vitro. To better understand why evolution has selected this unique structure, we asked, what is the fitness cost for development and function of mouse CD4+ T cells bearing a mutation in the Cα C-strand? In both TCR retrogenic and transgenic mice we observed increased single-positive thymocytes bearing mutant TCRs compared with those expressing wild-type TCRs. Furthermore, our analysis of mutant TCR transgenic mice revealed an increase in naive CD4+ T cells experiencing strong tonic TCR signals, increased homeostatic survival, and increased recruitment of responders to cognate pMHC class II upon immunization compared with the wild-type. The mutation did not, however, overtly impact CD4+ T cell proliferation or differentiation after immunization. We interpret these data as evidence that the unique Cα domain has evolved to fine-tune TCR signaling, particularly in response to weak interactions with self-pMHC class II.


DNA Repair , Receptors, Antigen, T-Cell , Animals , Mice , Cell Membrane , Thymocytes , Mice, Transgenic
4.
Elife ; 112022 07 21.
Article En | MEDLINE | ID: mdl-35861317

CD4+ T cells use T cell receptor (TCR)-CD3 complexes, and CD4, to respond to peptide antigens within MHCII molecules (pMHCII). We report here that, through ~435 million years of evolution in jawed vertebrates, purifying selection has shaped motifs in the extracellular, transmembrane, and intracellular domains of eutherian CD4 that enhance pMHCII responses, and covary with residues in an intracellular motif that inhibits responses. Importantly, while CD4 interactions with the Src kinase, Lck, are viewed as key to pMHCII responses, our data indicate that CD4-Lck interactions derive their importance from the counterbalancing activity of the inhibitory motif, as well as motifs that direct CD4-Lck pairs to specific membrane compartments. These results have implications for the evolution and function of complex transmembrane receptors and for biomimetic engineering.


CD4 Antigens , Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , Animals , CD3 Complex/metabolism , CD4 Antigens/genetics , CD4 Antigens/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Lymphocyte Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , src-Family Kinases/metabolism
5.
Proc Natl Acad Sci U S A ; 117(46): 28950-28959, 2020 11 17.
Article En | MEDLINE | ID: mdl-33139567

T cells express clonotypic T cell receptors (TCRs) that recognize peptide antigens in the context of class I or II MHC molecules (pMHCI/II). These receptor modules associate with three signaling modules (CD3γε, δε, and ζζ) and work in concert with a coreceptor module (either CD8 or CD4) to drive T cell activation in response to pMHCI/II. Here, we describe a first-generation biomimetic five-module chimeric antigen receptor (5MCAR). We show that 1) chimeric receptor modules built with the ectodomains of pMHCII assemble with CD3 signaling modules into complexes that redirect cytotoxic T lymphocyte (CTL) specificity and function in response to the clonotypic TCRs of pMHCII-specific CD4+ T cells, and 2) surrogate coreceptor modules enhance the function of these complexes. Furthermore, we demonstrate that adoptively transferred 5MCAR-CTLs can mitigate type I diabetes by targeting autoimmune CD4+ T cells in NOD mice. This work provides a framework for the construction of biomimetic 5MCARs that can be used as tools to study the impact of particular antigen-specific T cells in immune responses, and may hold potential for ameliorating diseases mediated by pathogenic T cells.


Antigens/metabolism , Biomimetics/methods , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism , Animals , Antigens/immunology , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Female , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Pancreas/immunology , Pancreas/pathology , Receptors, Antigen, T-Cell , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology
6.
Cell Rep ; 22(5): 1263-1275, 2018 01 30.
Article En | MEDLINE | ID: mdl-29386113

CD4+ T cells convert the time that T cell receptors (TCRs) interact with peptides embedded within class II major histocompatibility complex molecules (pMHCII) into signals that direct cell-fate decisions. In principle, TCRs relay information to intracellular signaling motifs of the associated CD3 subunits, while CD4 recruits the kinase Lck to those motifs upon coincident detection of pMHCII. But the mechanics by which this occurs remain enigmatic. In one model, the TCR and CD4 bind pMHCII independently, while in another, CD4 interacts with a composite surface formed by the TCR-CD3 complex bound to pMHCII. Here, we report that the duration of TCR-pMHCII interactions impact CD4 binding to MHCII. In turn, CD4 increases TCR confinement to pMHCII via reciprocal interactions involving membrane distal and proximal CD4 ectodomains. The data suggest that a precisely assembled macrocomplex functions to reliably convert TCR-pMHCII confinement into reproducible signals that orchestrate adaptive immunity.


CD4 Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Lymphocyte Activation/immunology , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , Adaptive Immunity/immunology , Animals , CD4 Antigens/chemistry , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Female , Histocompatibility Antigens Class II/immunology , Male , Mice , Mice, Transgenic , Protein Domains , Receptor-CD3 Complex, Antigen, T-Cell/chemistry , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Signal Transduction/immunology
7.
J Immunol ; 196(11): 4713-22, 2016 06 01.
Article En | MEDLINE | ID: mdl-27183595

TCRs relay information about peptides embedded within MHC molecules (pMHC) to the ITAMs of the associated CD3γε, CD3δε, and CD3ζζ signaling modules. CD4 then recruits the Src kinase p56(Lck) (Lck) to the TCR-CD3 complex to phosphorylate the ITAMs, initiate intracellular signaling, and drive CD4(+) T cell fate decisions. Whereas the six ITAMs of CD3ζζ are key determinants of T cell development, activation, and the execution of effector functions, multiple models predict that CD4 recruits Lck proximal to the four ITAMs of the CD3 heterodimers. We tested these models by placing FRET probes at the cytosolic juxtamembrane regions of CD4 and the CD3 subunits to evaluate their relationship upon pMHC engagement in mouse cell lines. The data are consistent with a compact assembly in which CD4 is proximal to CD3δε, CD3ζζ resides behind the TCR, and CD3γε is offset from CD3δε. These results advance our understanding of the architecture of the TCR-CD3-pMHC-CD4 macrocomplex and point to regions of high CD4-Lck + ITAM concentrations therein. The findings thus have implications for TCR signaling, as phosphorylation of the CD3 ITAMs by CD4-associated Lck is important for CD4(+) T cell fate decisions.


CD4 Antigens/immunology , Cell Membrane/immunology , Cytosol/immunology , Major Histocompatibility Complex/immunology , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Cell Line, Tumor , Fluorescence Resonance Energy Transfer , Humans , Mice
9.
Proc Natl Acad Sci U S A ; 113(11): 3000-5, 2016 Mar 15.
Article En | MEDLINE | ID: mdl-26831112

How T cells become restricted to binding antigenic peptides within class I or class II major histocompatibility complex molecules (pMHCI or pMHCII, respectively) via clonotypic T-cell receptors (TCRs) remains debated. During development, if TCR-pMHC interactions exceed an affinity threshold, a signal is generated that positively selects the thymocyte to become a mature CD4(+) or CD8(+) T cell that can recognize foreign peptides within MHCII or MHCI, respectively. But whether TCRs possess an intrinsic, subthreshold specificity for MHC that facilitates sampling of the peptides within MHC during positive selection or T-cell activation is undefined. Here we asked if increasing the frequency of lymphocyte-specific protein tyrosine kinase (Lck)-associated CD4 molecules in T-cell hybridomas would allow for the detection of subthreshold TCR-MHC interactions. The reactivity of 10 distinct TCRs was assessed in response to selecting and nonselecting MHCII bearing cognate, null, or "shaved" peptides with alanine substitutions at known TCR contact residues: Three of the TCRs were selected on MHCII and have defined peptide specificity, two were selected on MHCI and have a known pMHC specificity, and five were generated in vitro without defined selecting or cognate pMHC. Our central finding is that IL-2 was made when each TCR interacted with selecting or nonselecting MHCII presenting shaved peptides. These responses were abrogated by anti-CD4 antibodies and mutagenesis of CD4. They were also inhibited by anti-MHC antibodies that block TCR-MHCII interactions. We interpret these data as functional evidence for TCR-intrinsic specificity for MHCII.


Histocompatibility Antigens Class II/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Cell Antigen Receptor Specificity , Amino Acid Sequence , Animals , Antigen Presentation , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CHO Cells , Cell Line , Cell Membrane/immunology , Coculture Techniques , Cricetulus , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Hybridomas , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Immunological , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Recombinant Fusion Proteins/immunology , Sequence Deletion
10.
Immunity ; 43(2): 227-39, 2015 Aug 18.
Article En | MEDLINE | ID: mdl-26231119

The eight-subunit T cell receptor (TCR)-CD3 complex is the primary determinant for T cell fate decisions. Yet how it relays ligand-specific information across the cell membrane for conversion to chemical signals remains unresolved. We hypothesized that TCR engagement triggers a change in the spatial relationship between the associated CD3ζζ subunits at the junction where they emerge from the membrane into the cytoplasm. Using three in situ proximity assays based on ID-PRIME, FRET, and EPOR activity, we determined that the cytosolic juxtamembrane regions of the CD3ζζ subunits are spread apart upon assembly into the TCR-CD3 complex. TCR engagement then triggered their apposition. This mechanical switch resides upstream of the CD3ζζ intracellular motifs that initiate chemical signaling, as well as the polybasic stretches that regulate signal potentiation. These findings provide a framework from which to examine triggering events for activating immune receptors and other complex molecular machines.


CD3 Complex/metabolism , Cell Membrane/metabolism , Cytoplasm/metabolism , Multiprotein Complexes/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Animals , CD3 Complex/genetics , Humans , Hybridomas , Mechanotransduction, Cellular , Mice , Multiprotein Complexes/genetics , Protein Conformation , Protein Engineering , Protein Multimerization/genetics , Protein Multimerization/immunology , Protein Structure, Tertiary/genetics , Receptor Cross-Talk , Receptors, Antigen, T-Cell/genetics , Signal Transduction/genetics
11.
PLoS One ; 10(7): e0132333, 2015.
Article En | MEDLINE | ID: mdl-26147390

CD4 interactions with class II major histocompatibility complex (MHC) molecules are essential for CD4+ T cell development, activation, and effector functions. While its association with p56lck (Lck), a Src kinase, is important for these functions CD4 also has an Lck-independent role in TCR signaling that is incompletely understood. Here, we identify a conserved GGxxG motif in the CD4 transmembrane domain that is related to the previously described GxxxG motifs of other proteins and predicted to form a flat glycine patch in a transmembrane helix. In other proteins, these patches have been reported to mediate dimerization of transmembrane domains. Here we show that introducing bulky side-chains into this patch (GGxxG to GVxxL) impairs the Lck-independent role of CD4 in T cell activation upon TCR engagement of agonist and weak agonist stimulation. However, using Forster's Resonance Energy Transfer (FRET), we saw no evidence that these mutations decreased CD4 dimerization either in the unliganded state or upon engagement of pMHC concomitantly with the TCR. This suggests that the CD4 transmembrane domain is either mediating interactions with an unidentified partner, or mediating some other function such as membrane domain localization that is important for its role in T cell activation.


Lymphocyte Activation/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Protein Multimerization/physiology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Amino Acid Motifs , Animals , CD4 Antigens , CHO Cells , Cricetinae , Cricetulus , Fluorescence Resonance Energy Transfer , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/cytology
12.
Elife ; 4: e05949, 2015 Jul 14.
Article En | MEDLINE | ID: mdl-26173205

T-cell recognition of self and foreign peptide antigens presented in major histocompatibility complex molecules (pMHC) is essential for life-long immunity. How the ability of the CD4(+) T-cell compartment to bind self- and foreign-pMHC changes over the lifespan remains a fundamental aspect of T-cell biology that is largely unexplored. We report that, while old mice (18-22 months) contain fewer CD4(+) T-cells compared with adults (8-12 weeks), those that remain have a higher intrinsic affinity for self-pMHC, as measured by CD5 expression. Old mice also have more cells that bind individual or multiple distinct foreign-pMHCs, and the fold increase in pMHC-binding populations is directly related to their CD5 levels. These data demonstrate that the CD4(+) T-cell compartment preferentially accumulates promiscuous constituents with age as a consequence of higher affinity T-cell receptor interactions with self-pMHC.


Autoantigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Self Tolerance , Animals , CD5 Antigens/analysis , Mice
...