High-resolution analysis of the human T-cell receptor repertoire

Eliana Ruggiero, Jan P Nicolay, Raffaele Fronza, Anne Arens, Anna Paruzynski, Ali Nowrouzi, Gökçe Ürenden, Christina Lulay, Sven Schneider, Sergij Goerdt, Hanno Glimm, Peter H Krammer, Manfred Schmidt, Christof von Kalle, Eliana Ruggiero, Jan P Nicolay, Raffaele Fronza, Anne Arens, Anna Paruzynski, Ali Nowrouzi, Gökçe Ürenden, Christina Lulay, Sven Schneider, Sergij Goerdt, Hanno Glimm, Peter H Krammer, Manfred Schmidt, Christof von Kalle

Abstract

Unbiased dissection of T-cell receptor (TCR) repertoire diversity at the nucleotide level could provide important insights into human immunity. Here we show that TCR ligation-anchored-magnetically captured PCR (TCR-LA-MC PCR) identifies TCR α- and β-chain diversity without sequence-associated or quantitative restrictions in healthy and diseased conditions. TCR-LA-MC PCR identifies convergent recombination events, classifies different stages of cutaneous T-cell lymphoma in vivo and demonstrates TCR reactivation after in vitro cytomegalovirus stimulation. TCR-LA-MC PCR allows ultra-deep data access to both physiological TCR diversity and mechanisms influencing clonality in all clinical settings with restricted or distorted TCR repertoires.

Figures

Figure 1. TCR-LA-MC PCR methodology.
Figure 1. TCR-LA-MC PCR methodology.
For TCR analysis, RNA was used as the starting material and gene-specific cDNA synthesis was performed using a biotinylated primer located in the constant (C) gene of the TCR chains. The resulting full-length cDNA fragments were magnetically captured on streptavidin beads and ligated to a known single-stranded linker cassette (ssLC). After exponential PCR amplification, sequencing adaptors were added and the PCR products were deep sequenced. C, constant gene; BC, sample-specific barcode.
Figure 2. Immunomonitoring after CMV stimulation.
Figure 2. Immunomonitoring after CMV stimulation.
TCR-LA-MC PCR was performed on RNA samples for two CMV-seropositive donors ((a) donor 1; (b) donor 2; upper panel, α-chain; lower panel, β-chain) at different time points after CMV stimulation. At day 15, an additional enrichment of the IFNγ-secreting fraction was performed. The 10 most predominant CMV-reactive TCR sequences identified at day 15 and their relative contribution to the total repertoire (sequence count) are shown. Retrieval frequency is indicated with a colour code ranging from light pink (0.01–0.1%) to black (>40%). We observed enrichment of defined TCR specificities over time. The number of T-cell clones (shown as %) contributing to the generation of those specificities also increased, indicating that a portion of the T-cell population is committed to the generation of specific TCR molecules. The CMV specificity of the analysed sequences was further confirmed by analysis of the enriched fraction. Two replicates were performed for day 0, 9 and 15 of each donor and sequencing results were combined. ✓, sequence was identified in the CMV enriched fraction; aa, amino acid; CDR3, complementarity-determining region 3; d, day; IFN, interferon; Seq, sequence.
Figure 3. Contributions of CDR3 clonotypes in…
Figure 3. Contributions of CDR3 clonotypes in Sézary patients.
The figure shows the contributions of the CDR3 aa sequences to the αβ TCR repertoire in HDs and in 10 Sézary patients. Each bar represents an individual CDR3 aa clonotype, with red and violet indicating the first and tenth most predominant sequences, respectively. Grey bars indicate the contribution of the remaining sequences identified in the analysed sample. TCR-LA-MC PCR results show that the stage of the disease correlated well with the observed TCR clonality. These results were confirmed by calculating SI and SA diversity indices (DI). The higher is the SA or the SI, the more diverse is the population. Two replicates were performed for each TCR chain of each patient and sequencing results were combined. aa, amino acid; CDR3, complementarity-determining region 3; HD, healthy donor; P, patient; SA, Shannon index; SI, Simpson index.

References

    1. Douek D. C. et al.. A novel approach to the analysis of specificity, clonality, and frequency of HIV-specific T cell responses reveals a potential mechanism for control of viral escape. J. Immunol. 168, 3099–3104 (2002).
    1. Freeman J. D., Warren R. L., Webb J. R., Nelson B. H. & Holt R. A. Profiling the T-cell receptor beta-chain repertoire by massively parallel sequencing. Genome Res. 19, 1817–1824 (2009).
    1. Warren R. L. et al.. Exhaustive T-cell repertoire sequencing of human peripheral blood samples reveals signatures of antigen selection and a directly measured repertoire size of at least 1 million clonotypes. Genome Res. 21, 790–797 (2011).
    1. Quigley M. F., Almeida J. R., Price D. A. & Douek D. C. Unbiased molecular analysis of T cell receptor expression using template-switch anchored RT-PCR. Curr. Protoc. Immunol Chapter 10, Unit10: 33. doi: 10.1002/0471142735.im1033s94 (2011).
    1. Bolotin D. A. et al.. Next generation sequencing for TCR repertoire profiling: platform-specific features and correction algorithms. Eur. J. Immunol. 42, 3073–3083 (2012).
    1. Wang C. et al.. High throughput sequencing reveals a complex pattern of dynamic interrelationships among human T cell subsets. Proc. Natl Acad. Sci. USA 107, 1518–1523 (2010).
    1. Robins H. S. et al.. Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. Blood 114, 4099–4107 (2009).
    1. Robins H. S. et al.. Overlap and effective size of the human CD8+ T cell receptor repertoire. Sci. Transl. Med. 2, 47ra64 (2010).
    1. Klarenbeek P. L. et al.. Human T-cell memory consists mainly of unexpanded clones. Immunol. Lett. 133, 42–48 (2010).
    1. Troutt A. B., McHeyzer-Williams M. G., Pulendran B. & Nossal G. J. Ligation-anchored PCR: a simple amplification technique with single-sided specificity. Proc. Natl Acad. Sci. USA 89, 9823–9825 (1992).
    1. Argaet V. P. et al.. Dominant selection of an invariant T cell antigen receptor in response to persistent infection by Epstein-Barr virus. J. Exp. Med. 180, 2335–2340 (1994).
    1. Schmidt M. et al.. High-resolution insertion-site analysis by linear amplification-mediated PCR (LAM-PCR). Nat. Methods 4, 1051–1057 (2007).
    1. Paruzynski A. et al.. Genome-wide high-throughput integrome analyses by nrLAM-PCR and next-generation sequencing. Nat. Protoc. 5, 1379–1395 (2010).
    1. Gabriel R. et al.. Comprehensive genomic access to vector integration in clinical gene therapy. Nat. Med. 15, 1431–1436 (2009).
    1. Greenaway H. Y. et al.. NKT and MAIT invariant TCRα sequences can be produced efficiently by VJ gene recombination. Immunobiology 218, 213–224 (2013).
    1. Yousfi Monod M., Giudicelli V., Chaume D. & Lefranc M. P. IMGT/JunctionAnalysis: the first tool for the analysis of the immunoglobulin and T cell receptor complex V-J and V-D-J JUNCTIONs. Bioinformatics 20, (Suppl 1): i379–i385 (2004).
    1. Rosenberg W. M., Moss P. A. & Bell J. I. Variation in human T cell receptor V beta and J beta repertoire: analysis using anchor polymerase chain reaction. Eur. J. Immunol. 22, 541–549 (1992).
    1. Gauss G. H. & Lieber M. R. Mechanistic constraints on diversity in human V(D)J recombination. Mol. Cell. Biol. 16, 258–269 (1996).
    1. Venturi V., Price D. A., Douek D. C. & Davenport M. P. The molecular basis for public T-cell responses? Nat. Rev. Immunol. 8, 231–238 (2008).
    1. Quigley M. F. et al.. Convergent recombination shapes the clonotypic landscape of the naive T-cell repertoire. Proc. Natl Acad. Sci. USA 107, 19414–19419 (2010).
    1. Venturi V. et al.. Sharing of T cell receptors in antigen-specific responses is driven by convergent recombination. Proc. Natl Acad. Sci. USA 103, 18691–18696 (2006).
    1. Li H., Ye C., Ji G. & Han J. Determinants of public T cell responses. Cell Res. 22, 33–42 (2012).
    1. Price D. A. et al.. Avidity for antigen shapes clonal dominance in CD8+ T cell populations specific for persistent DNA viruses. J. Exp. Med. 202, 1349–1361 (2005).
    1. Lehner P. J. et al.. Human HLA-A0201-restricted cytotoxic T lymphocyte recognition of influenza A is dominated by T cells bearing the V beta 17 gene segment. J. Exp. Med. 181, 79–91 (1995).
    1. Trautmann L. et al.. Selection of T cell clones expressing high-affinity public TCRs within Human cytomegalovirus-specific CD8 T cell responses. J. Immunol. 175, 6123–6132 (2005).
    1. Miles J. J. et al.. Genetic and structural basis for selection of a ubiquitous T cell receptor deployed in Epstein-Barr virus infection. PLoS Pathog. 6, e1001198 (2010).
    1. Moss P. A. et al.. Extensive conservation of alpha and beta chains of the human T-cell antigen receptor recognizing HLA-A2 and influenza A matrix peptide. Proc. Natl Acad. Sci. USA 88, 8987–8990 (1991).
    1. Sinclair E. et al.. CMV antigen-specific CD4+ and CD8+ T cell IFNgamma expression and proliferation responses in healthy CMV-seropositive individuals. Viral Immunol. 17, 445–454 (2004).
    1. Slezak S. L. et al.. CMV pp65 and IE-1 T cell epitopes recognized by healthy subjects. J. Transl. Med. 5, 17 (2007).
    1. Kern F. et al.. Cytomegalovirus (CMV) phosphoprotein 65 makes a large contribution to shaping the T cell repertoire in CMV-exposed individuals. J. Infect. Dis. 185, 1709–1716 (2002).
    1. Wills M. R. et al.. The human cytotoxic T-lymphocyte (CTL) response to cytomegalovirus is dominated by structural protein pp65: frequency, specificity, and T-cell receptor usage of pp65-specific CTL. J. Virol. 70, 7569–7579 (1996).
    1. Olsen E. et al.. Revisions to the staging and classification of mycosis fungoides and Sezary syndrome: a proposal of the International Society for Cutaneous Lymphomas (ISCL) and the cutaneous lymphoma task force of the European Organization of Research and Treatment of Cancer (EORTC). Blood 110, 1713–1722 (2007).
    1. Yawalkar N. et al.. Profound loss of T-cell receptor repertoire complexity in cutaneous T-cell lymphoma. Blood 102, 4059–4066 (2003).
    1. Martins V. C. et al.. Thymus-autonomous T cell development in the absence of progenitor import. J. Exp. Med. 209, 1409–1417 (2012).
    1. Willenbrock K. et al.. Analysis of T-cell subpopulations in T-cell non-Hodgkin's lymphoma of angioimmunoblastic lymphadenopathy with dysproteinemia type by single target gene amplification of T cell receptor- beta gene rearrangements. Am. J. Pathol. 158, 1851–1857 (2001).
    1. Bolotin D. A. et al.. MiTCR: software for T-cell receptor sequencing data analysis. Nat. Methods 10, 813–814 (2013).
    1. Kent W. J. BLAT—the BLAST-like alignment tool. Genome Res. 12, 656–664 (2002).
    1. Lefranc M. P. et al.. IMGT, the international ImMunoGeneTics information system. Nucleic Acids Res. 33, D593–D597 (2005).
    1. Smith T. F. & Waterman M. S. Identification of common molecular subsequences. J. Mol. Biol. 147, 195–197 (1981).
    1. Gotoh O. An improved algorithm for matching biological sequences. J. Mol. Biol. 162, 705–708 (1982).
    1. Shannon C. E. The mathematical theory of communication. 1963. MD Comput. 14, 306–317 (1997).
    1. Simpson E. H. Measurement of diversity. Nature 163, 688 (1949).

Source: PubMed

3
Subskrybuj