T cell receptor specificity for major histocompatibility complex proteins

https://doi.org/10.1016/j.coi.2008.03.008Get rights and content

The ligands for alpha beta T cell receptors (αβTCRs) are usually major histocompatibility complex (MHC) proteins bound to peptides. Although there is evidence that T cell receptor variable regions have been selected evolutionarily to bind MHC, the rules governing this interaction have not previously been apparent. However, recent solved structures of T cell receptors with related variable regions bound to MHC plus peptides suggest that some amino acids in variable region CDR1 and CDR2s almost always react in a consistent way with MHC. These amino acids may therefore have been selected evolutionarily to predispose T cell receptors toward recognition of MHC ligands.

Introduction

Proteins have been evolutionarily selected to react specifically with particular targets, other proteins, nucleic acids, substrates for enzymes, and so on. This phenomenon is not difficult to understand for most proteins, which have well-defined sequences and a relatively small collection of ligands that are usually created directly or indirectly by the genome of their host. However, the phenomenon is more complicated for the antigen-recognizing proteins of the specific immune system, which are germline encoded, have related and very variable sequences but a huge range of unpredictable ligands that are not encoded by their host. For antibody proteins, the explanation is quite straightforward, the variability created during the assembly of immunoglobulin genes coupled with selection by antigen and somatic mutation during response picks out antibody proteins that can bind virtually any antigen. The situation for alpha beta T cell receptors (αβTCRs) is less easily understood. The variability is created only during gene rearrangement, not somatic mutation, and the repertoire of αβTCRs on mature T cells is tremendously biased toward recognition of antigens in the form of peptides bound to major histocompatibility complex (MHC) proteins.

How is this bias created? There are two explanations. One possibility is that the specificities of αβTCRs, like those of immunoglobulins, are intrinsically random. T cell receptors could react with any ligand. However, in order to turn into mature T cells, developing thymocytes must react with MHC/peptides in the thymus. Thus, the bias for MHC reactivity on mature T cells could be caused entirely by positive selection from pool of random TCRs. Plenty of proteins are expressed on thymus cortical epithelial cells, the major cells participating in positive selection. Of all of these, why are TCRs selected for reaction with MHC? Perhaps positive selection requires co-engagement of CD4 or CD8 [1, 2], and these proteins react with MHC, not other proteins on thymus epithelial cells. Alternatively thymocytes may be selected by reaction with all the other proteins on cortical epithelial cells, but then all those with high avidity/affinity are deleted by negative selection. In this case some mature T cells should bear αβTCRs with low affinity for proteins such as ICAM-1. These T cells will be invisible because they will never encounter a peripheral variant of their target ligand for which they have high affinity. However, to some extent a test of this idea has been performed, searching for T cells specific for the MHC-related protein, DM, and such cells were not found [3].

Finally, αβTCRs may react with MHC because they have a built-in affinity for these proteins [4]. If that happens, then probably it will operate via the CDR1 and 2 sequences of the TCR α and β chains since these are germline encoded, and since, in the solved structures of TCRs bound to MHC, these portions of the TCR contact MHC helix [5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17••, 18, 19••]. Against this idea is the fact that every species contains many TCR α and β variable regions and, although these fall into families, there is no close resemblance between the different groups [20]. Also, until recently, the solved structures of TCRs bound to MHC/peptide have failed to reveal any pattern whereby particular V region amino acids contact MHC in a reproducible way [5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17••, 18, 19••]. In favor of the idea is the fact that the TCR V region sequences are relatively conserved from man to mouse [20, 21], indicating functions for the various V families, and the fact that TCRs nearly always engage MHC diagonally [22], suggesting either that docking of CD4 and CD8 requires the diagonal (see above) or that some conserved feature of TCRs and MHC drives the phenomenon.

With the increasing numbers of solved structures of TCRs bound to MHC, evidence for conserved interactions is accumulating.

Section snippets

If TCR V regions are evolutionarily designed to react with MHC, why is not the nature of the design more evident from the solved structures of TCRs bound to MHC?

Probably there are several reasons why the evolutionarily selected rules, if they exist, have been hard to detect. First, both mouse and man, the two species from which all solved structures of TCR/MHC/peptide have been derived, code for many TCR Vαs and Vβs [20], and, particularly for Vα, there are not a lot of structures that include the same V regions. If we assume that different V regions have been evolutionarily selected to react with MHC in different ways, the lack, until recently, of

If evolutionarily selected rules govern V region engagement with MHC, what are they?

In the past couple of years a number of TCR/MHC/peptide structures have been reported in which the TCRs use the same or related V regions [17••, 19••, 34]. This has allowed investigators to search for conserved interactions between V region CDRs and MHC. In the first analysis of this type, Maynard et al. solved the structure of a mouse TCR, 172.10, bound to its ligand, IAu plus a peptide from myelin basic protein [17••]. 172.10 uses Vα2.3 and Vβ8.2, these are similar or identical to, those used

What about autoreactive and other unusual TCR/MHC interactions?

One might predict that autoreactive TCRs and their close relatives, specific for tumor antigens that have survived the rigors of negative selection, would react with MHC and peptide in odd ways. Such TCRs may fail to use the evolutionarily conserved rules. This idea turns out to be only partially true. For example, some of the TCRs used in the analyses that found conserved interactions by the Garcia group are autoreactive, binding IAu plus a myelin basic protein peptide. Nevertheless these TCRs

Conclusions

Antigen-specific receptors on T cells are constrained in many ways: by the ability of the host genome to code for them, by positive and negative selection, and by the requirement that at least most of them must be able to react with MHC proteins. It has long been believed that this last property is imposed on them not only by positive selection but also by the fact that the variable regions of T cell receptors have been designed to react with MHCs. However, until recently there has been little

Conflict of interest

The authors have no conflicts of interest with the subject of this article.

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

This work was supported partly by USPHS grants AI-17134, AI-18785 and AI-22295. The authors would like to thank all members of their laboratory for their helpful comments and putting up, for the past 25 years, with the authors’ obsession with the subject of this article.

References (36)

  • H.J. Kim et al.

    Coevolution of TCR-MHC interactions: conserved MHC tertiary structure is not sufficient for interactions with the TCR

    Proc Natl Acad Sci U S A

    (2005)
  • N.K. Jerne

    The somatic generation of immune recognition

    Eur J Immunol

    (1971)
  • K.C. Garcia et al.

    An alphabeta T cell receptor structure at 2.5 A and its orientation in the TCR-MHC complex

    Science

    (1996)
  • D.N. Garboczi et al.

    Structure of the complex between human T-cell receptor, viral peptide and HLA-A2

    Nature

    (1996)
  • K.C. Garcia et al.

    Structural basis of plasticity in T cell receptor recognition of a self peptide-MHC antigen

    Science

    (1998)
  • E.L. Reinherz et al.

    The crystal structure of a T cell receptor in complex with peptide and MHC class II

    Science

    (1999)
  • J. Hennecke et al.

    Structure of a covalently stabilized complex of a human alphabeta T-cell receptor, influenza HA peptide and MHC class II molecule, HLA-DR1

    EMBO J

    (2000)
  • J. Hennecke et al.

    Structure of a complex of the human alpha/beta T cell receptor (TCR) HA1.7, influenza hemagglutinin peptide, and major histocompatibility complex class II molecule, HLA-DR4 (DRA*0101 and DRB1*0401): insight into TCR cross-restriction and alloreactivity

    J Exp Med

    (2002)
  • Cited by (22)

    • Anti-tumor mechanism of eicosapentaenoic acid (EPA) on ovarian tumor model by improving the immunomodulatory activity in F344 rats

      2020, Journal of Functional Foods
      Citation Excerpt :

      MHC-II is mainly involved in the presentation of exogenous antigens. Just as CD8 T cells can only recognize antigenic fragments bound by MHC- I molecules, CD4 T cells can only recognize class II molecular binding point antigen fragments (Huang et al., 2013; Marrack, Rubtsova, Scott-Browne, & Kappler, 2008; Venalis & Lundberg, 2014). Many nutrients are capable of modulating the immune system.

    • Structural basis of chronic beryllium disease: Linking allergic hypersensitivity and autoimmunity

      2014, Cell
      Citation Excerpt :

      A summary of the atom-to-atom contacts and the complete details of the contacts are listed in Table 1 and Table S2, respectively. Typically, TCRs bind to MHCII ligands in a diagonal mode that places the CDR3 loops of the V domains over the peptide, while CDR1 and CDR2 loops make multiple interactions with the MHCII helices (see Marrack et al., 2008; Wilson and Garcia, 1997). The AV22 TCR also takes this diagonal orientation on the DP2 complex (Figure 4A), but there are some unusual features of the interface.

    • Immunological challenges for peptide-based immunotherapy in glioblastoma

      2014, Cancer Treatment Reviews
      Citation Excerpt :

      Finally, the peptide: HLA complex is transported to the cell surface through the Golgi apparatus [86]. For surface expression by an HLA molecule, certain amino acids at specific positions of the peptide have to bind into so called “anchoring pockets” of the HLA molecule [18,87]. Depending on its polymorphic regions, every HLA allele requires certain amino acid combinations for their anchoring pockets [88].

    • Subtle changes in TCRα CDR1 profoundly increase the sensitivity of CD4 T cells

      2013, Molecular Immunology
      Citation Excerpt :

      It has long been known that the variable CDR loops form the binding footprint for TCR to contact the pMHC complex (Garcia et al., 1999; Huseby et al., 2006). More recently it has been proposed that the TCR maintains germline-encoded affinity for MHC by interactions between key residues on the MHC helices and CDRs 1 and 2 (Adams et al., 2011; Marrack et al., 2008). CDRs 1 and 2 differ in the V gene segments, and it has been shown that introduction of mutations in these CDR loops can result in generation of high affinity TCRs (Chlewicki et al., 2005; Manning et al., 1999), potentially as a consequence of an optimal binding conformation or enhanced TCR:pMHC stability (Adams et al., 2011; Burrows et al., 2010; Dai et al., 2008; Holler and Kranz, 2004; Willcox et al., 1999).

    • Antigen receptor genes, gene products, and co-receptors

      2013, Clinical Immunology: Principles and Practice: Fourth Edition
    View all citing articles on Scopus
    View full text