Skip to main content
Erschienen in: Current Rheumatology Reports 12/2022

Open Access 05.10.2022 | Spondyloarthritis (MA Khan and N Akkoc, Section Editors)

T cell Repertoire Profiling and the Mechanism by which HLA-B27 Causes Ankylosing Spondylitis

verfasst von: Jose Garrido-Mesa, Matthew A. Brown

Erschienen in: Current Rheumatology Reports | Ausgabe 12/2022

Abstract

Purpose of Review

Ankylosing spondylitis (AS) is strongly associated with the HLA-B27 gene. The canonical function of HLA-B27 is to present antigenic peptides to CD8 lymphocytes, leading to adaptive immune responses. The ‘arthritogenic peptide’ theory as to the mechanism by which HLA-B27 induces ankylosing spondylitis proposes that HLA-B27 presents peptides derived from exogenous sources such as bacteria to CD8 lymphocytes, which subsequently cross-react with antigens at the site of inflammation of the disease, causing inflammation. This review describes findings of studies in AS involving profiling of T cell expansions and discusses future research opportunities based on these findings.

Recent Findings

Consistent with this theory, there is an expanding body of data showing that expansion of a restricted pool of CD8 lymphocytes is found in most AS patients yet only in a small proportion of healthy HLA-B27 carriers.

Summary

These exciting findings strongly support the theory that AS is driven by presentation of antigenic peptides to the adaptive immune system by HLA-B27. They point to new potential approaches to identify the exogenous and endogenous antigens involved and to potential therapies for the disease.
Hinweise
This article is part of the Topical Collection on Spondyloarthritis

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Adaptive immunity is orchestrated by peptides (epitopes or antigens) presented by histocompatibility antigens (HLAs) on antigen-presenting cells (APCs). This peptide-HLA (pHLA) pair is specifically recognised by immune receptors of lymphocytes (T cell — TCR; B cells — BCR or immunoglobulins). The sum of all immune receptors of one individual is termed the immune repertoire, which provides coverage against the diverse epitome. The epitome and immune repertoire define the host and the potential symbionts in immunological terms and changes greatly with the onset and progression of diseases. This is the reason why studying the immune repertoire is gaining more and more interest within immune disorders/IMIDs. By providing a global picture of the adaptive immune system, repertoire analyses have potential in both fields, as diagnostic biomarkers and for therapeutic development.
Ankylosing spondylitis (AS) is one of such pathologies where immune repertoire studies could make a significant impact. AS is the prototypic disease of a group of related disorders termed ‘seronegative spondyloarthropathies’ that also includes psoriatic arthritis, reactive arthritis (in response to bacterial urinary or gastrointestinal infection) and arthritis complicating inflammatory bowel disease. In total, these disease affects ~ 2–3% of European-descent populations and at least 1% of Asian populations [1]. AS causes significant back pain, stiffness, reduced function and eventual fusion of the spine and pelvis. AS first presents in early adulthood, initially with no changes visible on plain radiography of the sacroiliac joints (termed ‘non-radiographic axial spondyloarthritis’ (nr-axSpA), with characteristic sacroiliac radiographic changes developing subsequently and defining development of AS itself. Common extraskeletal associations of AS include acute anterior uveitis, inflammatory bowel disease and psoriasis. AS has a lifelong detrimental impact on patients, with depression and anxiety correlating with disease activity measures [2], absenteeism from work and high unemployment rates (40%). Furthermore, AS is also associated with early mortality [3], and current treatments do not change long-term prognosis.
AS is highly familial, with an increased risk in siblings of AS patients 82 × higher than the disease prevalence in the general community [4]. Twin studies [5, 6] suggest that > 90% of AS susceptibility is genetic in origin whilst the environmental trigger is likely ubiquitous. To date, 116 independent genomic loci having been robustly identified, contributing ~ 30% of the overall genetic risk, with ~ 20% being due to HLA-B*27 [7, 8, 9, 1011, 12•], one of the strongest genetic associations seen with any polygenic human disease (OR = 60, P < 10−300) (observed in > 80% of cases [13]). GWAS findings show that AS involves both the adaptive and innate immune systems, and that the IL-23 and TNF pathways are the major effector pathways involved [14].
HLA-B27 is a member of the HLA Class I family of MHC genes whose role is to present peptide antigens to CD8 T cells (Fig. 1). In AS, psoriasis and Behcet’s disease, there is robust evidence of gene–gene interaction between the HLA-I risk allele and ERAP1, indicating that they must operate closely together to influence disease risk [8, 15, 16]. The M1-aminopeptidase genes ERAP1 and/or ERAP2 are involved in the HLA-I antigenic processing and presentation (APP) pathway. These ERAP1 variants in AS patients exhibit significantly increased catalytic activity, resulting in over-trimming and creation of ‘unusual peptides’ presented by HLA-B27, which may not normally be generated in healthy situations. AS-associated ERAP2 variants break down peptides of 8 amino acids, and ERAP1 variant peptides longer than 9 amino acids [17]. This indicates that the peptides bound by HLA-B27 involved in AS-aetiopathogenesis are 9 amino acids in length, even though HLA-B27 has been shown to be able to present longer peptides.
In addition to altered peptidome due to HLA-B*27-ERAP1/2 interaction, there is evidence to suggest that, overall, AS and HLA-B*27 may be associated with defective gut immunity, enabling greater bacterial transgression across the gut mucosa. AS gut bacterial profiles are distinct from healthy controls [18, 19, 20•, 21], with evidence suggesting that these differences are driven by immunogenetic effects of associated genetic variants rather than being solely secondary to the disease itself (Fig. 2) [20•, 22••]. Peptide elution studies have shown an enrichment of bacterial peptides homologous to known HLA-B27-presented epitopes in the stools of patients with AS, suggesting a failure in clearance of these bacteria, and CD8 responses were observed to several peptides presented by APCs-B*27 + , but not B27-negative donors, consistent with these peptides driving adaptive immune activation in AS [20•]. Furthermore, reactive arthritis (ReA), triggered upon bacterial infection, proceeds to AS in ~ 10% of patients [23]. Paradoxically, the onset of many immune-mediated diseases is thought to involve an infectious stage, during which dampened immune responsiveness would be disadvantageous in clearing inflammatory stimuli. This is a feature of AS, for example, as shown by reports of deficient control of EBV infection [24, 25]. The underlying mechanisms involve known genetic associations, such as TLR4 [9] and MEFV [12•] involved in innate immune responses to bacterial components influencing a ‘reverse interferon signature’ in monocyte, macrophages and DCs from AS [26], as well as from HLA-B27/hb2m Tg rat model of SpA [27]. The initial infectious stage is followed by hyperactivation and disrupted self-tolerance in the context of innate immune activation and expansion of self-reactive Th17 cells or microbial mimicry (extensively reviewed [28]). CD8 T cells are therefore maintained in a state of heightened activation, potentially through persistent exposure to bacterial adjuvant, and fail to undergo senescence, leading to less productive responses to foreign antigen [25, 29••]. This further influences immune priming and results in greater proinflammatory cytokine (TNF, IL-23) production, contributing to the clinical features of joint and gut inflammation commonly found in AS [20•, 26, 27, 30]. In turn, exposure to gut-derived bacterial components lead to reduced integrity of the gut vascular barrier [31, 32], bacterial invasion of the intestinal epithelium [30] and disturbed microbiome that increase carriage of peptide pools presentable by HLA-B*27[20•], creating a deleterious cycle. These factors in combination affect the epitope repertoire available for T cell engagement [33, 34] and, consequently, alters the generation of adaptive immune diversity.
The exact mechanism by which HLA-B*27 induces AS is not known, but the leading theory proposes the ‘arthritogenic peptide hypothesis’, where unique microbial peptide(s) presented by HLA-B*27, similar and cross-reactive to human ‘self’ peptide(s), prime CD8 T cells to targets endogenous antigens found at the site of disease. In other inflammatory and autoimmune conditions, such as rheumatoid arthritis or type-1 diabetes, Tcell/pHLA biology is critical for initiation and propagation of disease [35, 36]. Yet, this knowledge is missing in AS. Studying the TCR repertoire provides a snapshot of an individual’s response to prior antigen exposure and can inform on the dynamics of immune responses, with application to immune monitoring in inflammatory diseases such as multiple sclerosis [37], autoimmune diseases [38], viral infection [39] or cancer [40] as well as biomarkers of treatment response [41]. In a variety of immune mediated diseases including type 1 diabetes [42], vitiligo [43] and narcolepsy [44], identification of expanded TCR clonotypes has assisted in demonstrating antigenic drivers of disease. Herein, we review available methodologies used in immune repertoire studies, up-to-date developments on AS and future directions towards the identification of the antigenic trigger in AS and its potential impact in AS therapeutics.

Method for TCR Repertoire Studies

Overview of TCR Structure
T cells recognise pHLA complexes through their T cell receptor (TCR). TCRs are highly diverse heterodimeric proteins consisting of a disulphide-linked α- and β-chains (αβ TCR), in the majority of T cells, or γδ chains (γδ TCR), expressed by T cells abundant at mucosal sites. Each chain is formed of a constant and a variable domain, and each variable domain contains three hypervariable loops or complementarity-determining regions (CDRs), which determine pHLA specificity. The variable regions of TCR β and γ chains are encoded by a number of variable (V), diversity (D) and joining (J) genes, while TCRα and δ chains are encoded by V-J genes only. TCR diversity is generated by 3 processes: (1) somatic recombination of VDJ gene fragments, (2) non-templated/random nucleotides insertion/deletions at the junction sites between the gene segments [45] and (3) pairing of α-β or γ-δ chains to form a functional TCR [46]. CDRs 1 and 2 are entirely encoded in germline DNA segment, whereas the CDR3a and CDR3b loops are products of junctional diversity, consequently being the most variable. These mechanisms yield an immense variety of TCR repertoire [47], estimated at around 1011 different clonotypes, with only fractional overlap in the TCR repertoires of any two individuals [48].
Each naïve T cell expresses a unique receptor with a unique spectrum of pHLA affinities [49]. When a TCR engages a pHLA on the surface of APCs, it becomes activated, inducing T cell proliferation leading to clonal populations that share the same TCR. In instances of strong selective pressure to common pHLA ligands among individuals (e.g. same infection/disease), similar clonally expanded TCRs arise, termed ‘public clonotypes’[50]. Whilst the majority of TCRs are rare [48], sharing of expanded TCR clonotypes between multiple individuals with a disease thus strongly supports a common antigenic drive [46, 51]. Similarly, alterations of the repertoire of public clonotypes among the general population reflects an underlying immune alteration that can be measured by TCR repertoire profiling [52].
TCR Repertoire Profiling
The vast diversity of the immune repertoire creates major challenges for its analysis, both in terms of laboratory approaches to its characterisation and in the analysis of that data. In the past decades, different laboratory techniques have been developed to profile the TCR repertoire, which are reviewed in depth elsewhere [53]. Initial low-resolution studies enabled investigation of the diversity of V gene usage, including by employing monoclonal antibodies against V regions and flow cytometry, or by measurement of differences in the length of CDR3 sequences by quantitative PCR amplification of the region and spectratyping techniques. The development of next-generation sequencing (NGS) methods, however, has now enabled comprehensive repertoire profiling. Here, we will provide a brief description of the two main NGS approaches: bulk sequencing of pooled immune populations and single cell approaches.
Population-Based Bulk-NGS Sequencing
Bulk methods are used to study repertoire diversity in larger cohorts. One of the initial considerations in such studies is the choice of starting material, gDNA or RNA [54]. Whilst using gDNA benefits from higher stability and better quantification of clonotype frequencies (single template per cell) [55], it has proven to be less sensitive, does not consider allelic exclusion (overestimating diversity) and is susceptible to sequencing errors due to introns and residuals of VDJ rearrangements. The opposite applies to RNA-based methods, which also account for level of expression. Because of the uniqueness of TCRβ CDR3 (higher combinatorial potential compared to α chains [54]) and its key role in determining antigen specificity, this has been the main target of interest.
The three most common methods for library construction are multiplex PCR, target enrichment and 5'RACE cDNA synthesis and nested PCR. Multiplex PCR approaches have been the most commonly used and can be applied to either gDNA or RNA. They use a set of primers for the constant region and all known V alleles to amplify the CDR3 region. Such approaches are thus limited in the detection of novel V alleles and are susceptible to amplification biases [56], although this can be corrected for by adjusting primer concentrations and experimental conditions [57] or by molecular barcoding [58]. Target enrichment methods use custom-designed RNA baits to hybridize with the gDNA/cDNA target, followed by a further amplification step involving fewer PCR cycles than is typically used in multiplex PCR, thus reducing susceptibility to amplification bias [59]. The third method, 5′RACE [60], uses a primer against the constant region of the TCR mRNA transcript for cDNA synthesis of the complete 5′ end. The terminal transferase activity of the reverse transcriptase enzyme incorporates additional dCTP bases at the 3′ end of the cDNA, allowing for a template-switch with an oligoG, completing the second cDNA strand [61]. This is followed by a nested PCR amplification using a single pair of primers (3′-C region and 5′-common adaptor), thus minimising amplification bias related to primer differences. The method enriches for all TCR variants in the sample (known and novel), a significant advantage over other methods [62, 6364]. Since a specific TCR may differ from another by only a single nucleotide and every method is susceptible to errors, to distinguish between errors and low-frequency clonotypes, the introduction of unique molecular identifiers (UMIs) during cDNA synthesis allows application of correction algorithms and for absolute counts to be obtained [65, 66]. Once the library is prepared, sequencing depth requirements depend on the aim of the experiment. For a disease-oriented analysis looking for clonally expanded TCRs, a low-coverage screening may even be enough, whilst deep sequencing would be required for a more complete and complex repertoire and to identify rare clones [64].
Primary analysis involves the recovery of TCR sequences from raw data, annotation and clonotype clustering and quantification, as summarized in previous reviews [54, 67, 68]. In brief, reads are initially processed to eliminate those with a high error rate, remove primer sequences and build consensus sequences (contigs) from aligning paired-end reads and multiple reads from the same cDNA molecule with matching UMIs. For each contig, the germline V(D)J alleles most likely involved in gene rearrangement are inferred from a reference set and annotated. The frequency of each identified rearranged sequence (clonotype) is then identified. From that point, output data (generally in the form of table) is used for secondary analysis and visualization. Of note, immune repertoire sequences can be also extrapolated from untargeted transcriptome sequencing [69]. Whilst this approach may be limited by sequencing depth (revealing only a fraction of the TCR diversity) and is susceptible to errors from short read sequence assembly, given the availability of transcriptomic datasets, it can be a useful tool to screen for highly prevalent clonotypes.
Secondary repertoire analysis uses descriptive statistical indices of diversity and homology and visualization methods that can be approached with the numerous omics tools developed by the scientific community over recent years [70, 7172] (https://​omictools.​com/​repseq-category). While no framework is ubiquitous, the AIRR Community of the Antibody Society has established some standards for data representation (https://​docs.​airr-community.​org/​en/​stable). Mathematical indexes used to measure diversity and convergence of immune repertoires [71] derive from ecology, to quantify ecosystem biodiversity based on the information theory. Diversity relates to the level of uncertainty that a TCR sequence, sorted from a repertoire, would belong to a certain T cell clone, which depends on the number of unique TCR sequences (richness) and their relative abundance (evenness). The different parameters used include the Shannon, Inverse-Simpson and Gini indices [73].
Differences between immune repertoire profiles from different samples/datasets, such as biased VDJ gene usage [74], suggest association of TCR clonotypes with disease pathogenesis. By providing a fingerprint of adaptive immunity, immune repertoire analysis can capture alterations of the immune fitness. Further analysis options address the antigen specificity of the TCR receptors. By evaluating patterns in sequence [75], structure [76] and physicochemical properties [77], clustering algorithms link TCRs to antigen specificity or to clinical outcomes. However, the prediction of a T cell epitome remains one of the biggest challenges of cellular and computational immunology, which requires additional experimental input [47]. Such input will probably derive from single-cell analysis, providing the full TCRαβ pair required for pHLA interaction, and subsequent functional studies identifying the complementary epitome and range of affinities.
Single-Cell Transcriptomics
Single-cell TCR profiling has emerged in recent years, linking αβ chains with phenotyping by transcriptional profiling. scRNAseq was originally developed to obtain paired TCRαβ sequences [78]. Whilst it still can lead to straightforward antigen identification [79], further development of the technique now allows for surface marker phenotyping using DNA-barcoded antibodies, epigenetic profiling and even screening of a small number of peptide using MHC tetramers, thus expanding the potential to draw biological in vivo function of the clones. Among the biggest limitations are the high cost, limited number of covered cells and requirement of fresh material for the isolation and sorting of live cells (not always available). The different methodologies are reviewed elsewhere [80], but it is worth highlighting the major improvement in scTCR-Seq with emulsion-based approaches, which use water-in-oil emulsion droplets that trap single cells with small volumes of reagents, including the barcodes required for multiplexing.

Findings to Date in AS (Implications for Aetiopathogenesis)

Multiple studies have now investigated T cell repertoire variation in AS and ReA, with consistent evidence of expansion of T cell clonotypes. Prior to the development of NGS, Marker-Hermann and colleagues performed seminal research in this field, following from their demonstration of CTL in ReA patients capable of reacting to the bacteria involved in triggering the ReA, and also self-reactive CTL [81]. CDR3 spectratyping and sequencing were then used to study TCR usage in patients with ReA, in comparison with healthy controls and rheumatoid arthritis patients [82]. Expansion of two TCRBV1-bearing T cells was demonstrated in synovial fluid of ReA but not healthy controls. TCRBV1 was defined by antibody staining (clone BL37.2) and is now known as TCRBV9 as defined by DNA sequence. In two patients, expansions involving two TCR clonotypes, bearing either the CDR3 sequence CAS-SVGLYSTDTQ or CAS-SPGLYSTDTQ, were identified [82]. A database search subsequently identified 148 HLA-B27/SpA-derived TCRB CDR3 sequences, from which a canonical TCRB CDR3 sequence, TCRBV1/23-CASSVG(V/I/L)(Y/F)STDTQYF-J2S3, was identified [83]. The database-derived HLA-B27/SpA sequences did not match those of CDR3 sequences from HLA-B27-responding clonotypes from healthy subjects, indicating a disease-association rather than simply being an HLA-B27-restricted motif. This TCR-CDR3 sequence matched 43/148 (29%) published ReA-derived CDR3 sequences from 12 different patients, but only 19/3799 (0.5%) other human TCRB sequences present in public sequence databases (odds ratio = 60, P < 10−100)[83].
The development of NGS enabled TCR profiling, as described above, encouraged further studies in this field. Using TCRB VDJ sequencing, Faham et al. studied 234 AS patient (192 HLA-B27 positive), and 227 controls (10 HLA-B27 positive), and confirmed increased carriage of a subset of six of the TCR clonotypes reported previously, and in total demonstrated 15 motifs enriched in the B27-positive AS patients, as compared to B27-positive healthy individuals (P = 0.001 and P = 0.049, respectively) [84]. The study involved only 10 HLA-B27 controls, and the findings for novel motifs that had not previously been reported were not analysed separately and thus require further validation. The previously reported SVGLYSTDTQ and TRBV9-SVGLYST-TRBVJ2-3 motifs were found in 30–39% of two sets of AS cases, and 0% of the overall control set (P = 4.6 × 10−12).
Using 5′-RACE, Komech et al. studied TCRB usage in blood and synovial fluid from 25 AS patients (24 HLA-B27 positive) and blood from 107 healthy controls (7 HLA-B27 positive, 8 HLA-B27 negative, 92 unknown HLA-B27 status). They also demonstrated expansion of TRBV9-CASSVGLYSTDTQYF-TRVBJ2-3. In total, eight TCR-CDR3 motifs were found to be expanded including three (CASSVGLFSTDTQYF, CASSVGLYSTDTQYF and CASSVGVYSTDTQYF) previously reported and confirmed by Faham et al. [84]. Seven of the eight expanded clonotypes were found amongst CD8-positive lymphocytes, consistent with their interaction with HLA-I alleles. Each of the eight AS-associated clonotypes were found in synovial fluid from four HLA-B*27-positive AS patients but not in synovial fluid from an HLA-B*27-negative patient.
Subsequent to this, Hanson et al. reported findings using bulk TCR repertoire profiling of sorted CD8 + and CD4 + PBMCs from 47 AS patients (37 HLA-B27-positive) and 38 healthy controls (20 HLA-B27-positive) [85••]. This study demonstrated expansion of both CD8 + and CD4 + TCR clonotypes in AS patients compared with HLA-B27-matched healthy controls. Ten closely related CD8 TCR clones were shown to be strongly associated with AS, found in all of 37 HLA-B27-positive AS patients, but only 4/19 HLA-B27-positive controls (P = 2.6 × 10−6). The previously reported CASSVGLFSTDTQYF motif was found in CD8 lymphocytes from 12 cases and no controls (P = 1.6 × 10−5).
Lastly, Zheng et al. used multiplex PCR and RNA-seq to profile TCR usage in peripheral blood and synovial joint fluid from axial spondyloarthritis patients (with either AS or non-radiographic disease) and healthy controls of unreported HLA-B27 status [86••]. This study suggested that expanded TCR clonotypes were seen amongst both CD4- and CD8-positive T cells, with identical expanded TCRB sequences in both CD4 and CD8 T cells observed. Comparison with previous findings confirmed expansion of the CAS***STDTQYF CDR3 motif in synovial joint fluid CD8 and CD4/8 T cells but not in CD4 + T cells. Additionally, expansion was observed with two other CDR3 motifs (CAS***SPLHF in SJF CD4/8 T cells, and CAS***GANVLTF in all CD4, CD8 and CD4/8 T cell populations from SJF). Whether these expansions are disease or HLA-B27 specific is not yet clear, but if they are truly disease-associated, then this is a particularly exciting confirmation, occurring in a completely different ancestral group to those previously studied, and likely involving patients with different HLA-B27 subtypes.
Overall, these findings provide strong evidence of expansion of a restricted set of CDR3 sequence–defined clonotypes particularly amongst CD8 lymphocytes in AS cases. This provides strong evidence supporting the arthritogenic-peptide model of how HLA-B27 induces AS.

Immune Repertoire — Epitome Linking Approaches and Future Directions

Having identified and characterised an expanded clonotype, identifying the peptide(s) to which it is responding is a key translational step. The approaches taken to achieve this involve either TCR or antigen screening studies (Fig. 3).
TCR Screening
TCR screening studies have been historically performed by co-culturing of mixed lymphocyte populations with antigen-pulsed APCs. Upon recognition, T cell activation can be evaluated by changes in surface markers, proliferation, cytokine production or cytolytic activity [87]. Activated clones can then be characterized, including by TCR sequencing. Increased throughput can be achieved with novel methods using fluorescently labelled soluble MHC tetramers loaded with the antigen, identifying antigen-specific T cells among a heterogenous population using flow cytometry [88]. Using tetramers labelled with different fluorochromes, a few different antigens can be studied per experiment, including the direct assessment of their phenotype with antibody panels, and by sorting and sequencing. Different variations of this approach have been developed. The use of nanoparticles harbouring > 104 tetramers per particle increases the method’s sensitivity [89]. CyTOF detection with metal tags instead of fluorescence improves the phenotypic characterization, but at the cost of TCR definition by sequencing, as the cell is destroyed during detection [90]. DNA-tagged tetramers increase the number of epitopes that can be simultaneously evaluated [91] in combination with single-cell sequencing. Microfluidic devices have been developed enabling higher throughputs with reduced cell input requirements [89, 92]. These methods could be useful for the identification of high-avidity interactions, such as those of some neoantigens or microbe-specific T cells [93]. However, it is worth mentioning that the TCR-pHLA binding is of relatively low affinity and degenerate [48], so that many TCRs could recognize the same antigen, and many antigens are recognized by the same TCR. As such, more precise methods are required to investigate low-affinity repertoires, as are methods to overcome the difficulty of production, and lack of stability, of the multimers. Nevertheless, these technics are still in their infancy, so they hold promise as further developments overcome some of these limitations, such as their reliance in MHC tetramer technology.
Antigen Screening
Antigen screening is a more suitable approach to follow-up for immune repertoire analysis that identifies clonal expansions associated to a particular condition, as in most cases, the antigen involved is unknown. This approach requires large numbers of cells expressing the TCRs of interest, which are typically obtained either from a natural source by immortalization or, more typically, by cloning into a cell line. When the origin of the epitope is suspected, the TCRs can be tested in vitro against a library of peptides or whole genome libraries of the suspected organism displayed on the surface of baculoviruses, yeast or mammalian cells [93, 94, 9596]. In a similar fashion to pHLA tetramers, soluble fluorescently tagged TCR tetramers can be used to screen these libraries, with the specific antigen being confirmed by sorting and sequencing of the host-display system [97]. However, this method entails the same challenges of generating the tetramers and the avidity of the TCR/pHLA physical interaction [47]. Cell-based strategies that evaluate triggered signals, rather than binding itself, can overcome these issues. Some recent developments use chimeric receptors that couple extracellular pHLA and intracellular TCR signalling domains in reporter systems, such as the SABRs (‘signalling and antigen-presenting bifunctional receptors’)[98] or pHLA/eTCRs[99]. However, the challenges of adapting those systems to the particular requirements of the study, such as specific HLA alleles (risk or protective), unconventional peptides or various TCRs variants, may limit their applicability and widespread adoption.
The most direct experimental method for antigen screening is based on the use of combinatorial peptide libraries (CPLs) [100]. These are highly complex mixtures of peptides systematically arranged so that each pool keeps one position defined whilst the others vary (termed as “positional scanning library”) [101]. Whilst complex, time consuming and costly, these studies allow the unbiased elucidation of T cell ligands and the functional characterization at each position, defining peptide variants that also induce activity [102]. Their use led to the discovery of TCR degeneracy [103] and to the identification of multiple ligands of different affinities for both MHC class I [104] and class II molecules [105]. CPLs have the advantages that they are more independent of the experimental system, applicable to different APC lines harbouring different sets of HLA molecules and can be used with a wide variety of readouts.
The sensitivity of the readout system may be a particularly relevant factor for a successful experimental detection of a TCR epitome, given than many TCR-pHLA interactions are of low affinity. Many studies use classical readouts, such as clonal expansion, surface marker upregulation or cytokines/granzyme B release [49], whist others have exploited “extravagant/unusual” outcomes of TCR-pHLA interaction, such as the transfer of membranous materials between T cells and APC (‘trogocytosis’) [106]. The employment of transcriptional reporter systems conveniently allows for unmanipulated straightforward quantification [107], an attractive option for large-scale studies.

Repertoire Studies and the Development of AS Therapeutics

Repertoire analyses and the identification of TCR-pHLA associations involved in AS could not only help resolve the pathogenesis of AS, but would also be informative about multiple other HLA-I immune-mediated inflammatory diseases (IMIDs) such as psoriasis (HLA-Cw6), Behcet’s disease (HLA-B51) and birdshot retinopathy (HLA-A29). It is very likely that in these diseases, which are each strongly associated with different HLA Class I antigens and with ERAP1, the mechanism by which these associations operate to cause disease is similar. Identifying expanded TCR clonotypes and the antigen(s) driving these expansions have obvious potential therapeutic significance. Most directly, if these clonotypes are involved in driving inflammation in AS, then deleting them may be therapeutic. Phase 1 trials of an anti-TRBV9 monoclonal antibody, BCD-180, are underway in Russia on the bases of clonal expansions frequently carrying TRBV9 [108] (https://​biocad.​ru/​pipelines/​). Whilst this individual TRBV gene may be expanded in AS, there is substantial risk of targeting a large range of non-pathogenic TRBV9 TCRs, as well as missing AS-associated clones with a similar CDR3 but from a range of TRBV families. It is also not yet clear whether the expanded TRBV9-bearing cells are causing AS-associated inflammation or are involved in some unsuccessful disease-protective response. Thus, such approaches could lead to broader immunological effects and treatment-associated toxicities.
A more precise characterization of CD8 T cell clones that are uniquely expanded in HLA-B27 + AS patients could help to identify cellular biomarkers for improved diagnostics. Most importantly, this new knowledge would be critical for the development of targeted therapies informed by underlying disease mechanisms. The use of AS-expanded T cells to identify the antigenic driver(s) and its source, potentially cross-reactive with a bacterial antigen as suggested by the model of ReA and gut microbiome studies discussed above, raises the possibility of specific antimicrobial therapies to manage or prevent the disease. In rheumatoid arthritis, knowledge of immunopathogenic peptides, which are present 15 years before the onset of clinical symptoms, has revolutionised diagnosis [109] and opened the door for development of peptide-specific therapies [110]. In AS, microbiome studies [20•] and peptide elution studies from HLA-B27 [111, 112] could help to narrow down the number of peptides that require testing. Whilst an efficient approach, this could hinder the discovery of novel peptides of potential application to modulate the response of these pathogenic clones. For example, the antigen-specific response of AS-associated clones could be therapeutically inactivated by targeting the antigen to steady-state dendritic cells, which in turn regulate Ag-specific memory and effector T cell populations [113, 114]. Or as shown in ovalbumin-induced arthritis in mice [115], by TCR-gene transfer, the target of primary regulatory T cells could be redirected and adoptive therapy used to induce antigen-specific suppression of the pathology. These strategies would be specific to the condition, thus eluding off-target toxicity. In conclusion, the strategies informed by repertoire studies enable the prospect of preventative/early treatment for AS with potential for prolonged disease remissions [116].

Declarations

Human and Animal Rights and Informed Consent.

This article does not contain any studies with human or animal subjects performed by any of the authors.

Competing Interests

The authors declare no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
1.
Zurück zum Zitat Dean LE, Jones GT, MacDonald AG, Downham C, Sturrock RD, Macfarlane GJ. Global prevalence of ankylosing spondylitis. Rheumatology (Oxford). 2014;53(4):650–7.CrossRef Dean LE, Jones GT, MacDonald AG, Downham C, Sturrock RD, Macfarlane GJ. Global prevalence of ankylosing spondylitis. Rheumatology (Oxford). 2014;53(4):650–7.CrossRef
2.
Zurück zum Zitat Healey EL, Haywood KL, Jordan KP, Garratt A, Packham JC. Impact of ankylosing spondylitis on work in patients across the UK. Scand J Rheumatol. 2011;40(1):34–40.PubMedCrossRef Healey EL, Haywood KL, Jordan KP, Garratt A, Packham JC. Impact of ankylosing spondylitis on work in patients across the UK. Scand J Rheumatol. 2011;40(1):34–40.PubMedCrossRef
3.
Zurück zum Zitat Bakland G, Gran JT, Nossent JC. Increased mortality in ankylosing spondylitis is related to disease activity. Ann Rheum Dis. 2011;70(11):1921–5.PubMedCrossRef Bakland G, Gran JT, Nossent JC. Increased mortality in ankylosing spondylitis is related to disease activity. Ann Rheum Dis. 2011;70(11):1921–5.PubMedCrossRef
4.
Zurück zum Zitat Brown MA, Laval SH, Brophy S, Calin A. Recurrence risk modelling of the genetic susceptibility to ankylosing spondylitis. Ann Rheum Dis. 2000;59(11):883–6.PubMedPubMedCentralCrossRef Brown MA, Laval SH, Brophy S, Calin A. Recurrence risk modelling of the genetic susceptibility to ankylosing spondylitis. Ann Rheum Dis. 2000;59(11):883–6.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Brown MA, Kennedy LG, MacGregor AJ, Darke C, Duncan E, Shatford JL, et al. Susceptibility to ankylosing spondylitis in twins: the role of genes, HLA, and the environment. Arthritis Rheum. 1997;40(10):1823–8.PubMedCrossRef Brown MA, Kennedy LG, MacGregor AJ, Darke C, Duncan E, Shatford JL, et al. Susceptibility to ankylosing spondylitis in twins: the role of genes, HLA, and the environment. Arthritis Rheum. 1997;40(10):1823–8.PubMedCrossRef
6.
Zurück zum Zitat Pedersen OB, Svendsen AJ, Ejstrup L, Skytthe A, Harris JR, Junker P. Ankylosing spondylitis in Danish and Norwegian twins: occurrence and the relative importance of genetic vs. environmental effectors in disease causation. Scand J Rheumatol. 2008;37(2):120–6.PubMedCrossRef Pedersen OB, Svendsen AJ, Ejstrup L, Skytthe A, Harris JR, Junker P. Ankylosing spondylitis in Danish and Norwegian twins: occurrence and the relative importance of genetic vs. environmental effectors in disease causation. Scand J Rheumatol. 2008;37(2):120–6.PubMedCrossRef
7.
Zurück zum Zitat Burton PR, Clayton DG, Cardon LR, Craddock N, Deloukas P, Duncanson A, et al. Association scan of 14,500 nonsynonymous SNPs in four diseases identifies autoimmunity variants. Nat Genet. 2007;39(11):1329–37.PubMedCrossRef Burton PR, Clayton DG, Cardon LR, Craddock N, Deloukas P, Duncanson A, et al. Association scan of 14,500 nonsynonymous SNPs in four diseases identifies autoimmunity variants. Nat Genet. 2007;39(11):1329–37.PubMedCrossRef
8.
Zurück zum Zitat Cortes A, Pulit SL, Leo PJ, Pointon JJ, Robinson PC, Weisman MH, et al. Major histocompatibility complex associations of ankylosing spondylitis are complex and involve further epistasis with ERAP1. Nat Commun. 2015;21(6):7146.CrossRef Cortes A, Pulit SL, Leo PJ, Pointon JJ, Robinson PC, Weisman MH, et al. Major histocompatibility complex associations of ankylosing spondylitis are complex and involve further epistasis with ERAP1. Nat Commun. 2015;21(6):7146.CrossRef
9.
Zurück zum Zitat Ellinghaus D, Jostins L, Spain SL, Cortes A, Bethune J, Han B, et al. Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nat Genet. 2016;48(5):510–8.PubMedPubMedCentralCrossRef Ellinghaus D, Jostins L, Spain SL, Cortes A, Bethune J, Han B, et al. Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nat Genet. 2016;48(5):510–8.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat International Genetics of Ankylosing Spondylitis Consortium, Cortes A, Hadler J, Pointon JP, Robinson PC, Karaderi T, et al. Identification of multiple risk variants for ankylosing spondylitis through high-density genotyping of immune-related loci. Nat Genet. 2013;45(7):730–8.CrossRef International Genetics of Ankylosing Spondylitis Consortium, Cortes A, Hadler J, Pointon JP, Robinson PC, Karaderi T, et al. Identification of multiple risk variants for ankylosing spondylitis through high-density genotyping of immune-related loci. Nat Genet. 2013;45(7):730–8.CrossRef
11.
Zurück zum Zitat Reveille J, Sims A, Maksymowych W, Ward M, Stone MPR, et al. Genomewide association study in ankylosing spondylitis identifies major non-MHC genetic determinants of disease susceptibility. Arthritis Rheum. 2008;Suppl 1:1186. Reveille J, Sims A, Maksymowych W, Ward M, Stone MPR, et al. Genomewide association study in ankylosing spondylitis identifies major non-MHC genetic determinants of disease susceptibility. Arthritis Rheum. 2008;Suppl 1:1186.
12.•
Zurück zum Zitat Li Z, Akar S, Yarkan H, Lee SK, Cetin P, Can G, et al. Genome-wide association study in Turkish and Iranian populations identify rare familial Mediterranean fever gene (MEFV) polymorphisms associated with ankylosing spondylitis. PLoS Genet. 2019;15(4):e1008038. Demonstrated strong association of variants of MEFV which is known to cause familial Mediterranean fever with AS, including HLA-B27 negative cases.PubMedPubMedCentralCrossRef Li Z, Akar S, Yarkan H, Lee SK, Cetin P, Can G, et al. Genome-wide association study in Turkish and Iranian populations identify rare familial Mediterranean fever gene (MEFV) polymorphisms associated with ankylosing spondylitis. PLoS Genet. 2019;15(4):e1008038. Demonstrated strong association of variants of MEFV which is known to cause familial Mediterranean fever with AS, including HLA-B27 negative cases.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Brewerton DA, Cafrey M, Hart FD, James DCO, Nicholls A, Sturrock RD. Ankylosing spondylitis and HL-A 27 Lancet. 1973;904–907 Brewerton DA, Cafrey M, Hart FD, James DCO, Nicholls A, Sturrock RD. Ankylosing spondylitis and HL-A 27 Lancet. 1973;904–907
14.
Zurück zum Zitat Brown MA, Kenna T, Wordsworth BP. Genetics of ankylosing spondylitis-insights into pathogenesis. Nat Rev Rheumatol. 2016;12(2):81–91.PubMedCrossRef Brown MA, Kenna T, Wordsworth BP. Genetics of ankylosing spondylitis-insights into pathogenesis. Nat Rev Rheumatol. 2016;12(2):81–91.PubMedCrossRef
15.
Zurück zum Zitat Genetic Analysis of Psoriasis C, the Wellcome Trust Case Control C, Strange A, Capon F, Spencer CC, Knight J, et al. A genome-wide association study identifies new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat Genet. 2010;42(11):985–90.CrossRef Genetic Analysis of Psoriasis C, the Wellcome Trust Case Control C, Strange A, Capon F, Spencer CC, Knight J, et al. A genome-wide association study identifies new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat Genet. 2010;42(11):985–90.CrossRef
16.
Zurück zum Zitat Kirino Y, Bertsias G, Ishigatsubo Y, Mizuki N, Tugal-Tutkun I, Seyahi E, et al. Genome-wide association analysis identifies new susceptibility loci for Behcet’s disease and epistasis between HLA-B*51 and ERAP1. Nat Genet. 2013;45(2):202–7.PubMedPubMedCentralCrossRef Kirino Y, Bertsias G, Ishigatsubo Y, Mizuki N, Tugal-Tutkun I, Seyahi E, et al. Genome-wide association analysis identifies new susceptibility loci for Behcet’s disease and epistasis between HLA-B*51 and ERAP1. Nat Genet. 2013;45(2):202–7.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Tito RY, Cypers H, Joossens M, Varkas G, Van Praet L, Glorieus E, et al. Brief report: dialister as a microbial marker of disease activity in spondyloarthritis. Arthritis Rheumatol. 2017;69(1):114–21.PubMedCrossRef Tito RY, Cypers H, Joossens M, Varkas G, Van Praet L, Glorieus E, et al. Brief report: dialister as a microbial marker of disease activity in spondyloarthritis. Arthritis Rheumatol. 2017;69(1):114–21.PubMedCrossRef
19.
Zurück zum Zitat Wen C, Zheng Z, Shao T, Liu L, Xie Z, Le Chatelier E, et al. Quantitative metagenomics reveals unique gut microbiome biomarkers in ankylosing spondylitis. Genome Biol. 2017;18(1):142.PubMedPubMedCentralCrossRef Wen C, Zheng Z, Shao T, Liu L, Xie Z, Le Chatelier E, et al. Quantitative metagenomics reveals unique gut microbiome biomarkers in ankylosing spondylitis. Genome Biol. 2017;18(1):142.PubMedPubMedCentralCrossRef
20.•
Zurück zum Zitat Yin J, Sternes PR, Wang M, Song J, Morrison M, Li T, et al. Shotgun metagenomics reveals an enrichment of potentially cross-reactive bacterial epitopes in ankylosing spondylitis patients, as well as the effects of TNFi therapy upon microbiome composition. Ann Rheum Dis. 2020;79(1):132–40. Confirms in a non-European setting that the gut microbiome is different in AS patients, and that peptides derived from microbiome components are recognised by T-cells in HLA-B27 positive but not negative subjects.PubMedCrossRef Yin J, Sternes PR, Wang M, Song J, Morrison M, Li T, et al. Shotgun metagenomics reveals an enrichment of potentially cross-reactive bacterial epitopes in ankylosing spondylitis patients, as well as the effects of TNFi therapy upon microbiome composition. Ann Rheum Dis. 2020;79(1):132–40. Confirms in a non-European setting that the gut microbiome is different in AS patients, and that peptides derived from microbiome components are recognised by T-cells in HLA-B27 positive but not negative subjects.PubMedCrossRef
21.
Zurück zum Zitat Klingberg E, Magnusson MK, Strid H, Deminger A, Stahl A, Sundin J, et al. A distinct gut microbiota composition in patients with ankylosing spondylitis is associated with increased levels of fecal calprotectin. Arthritis Res Ther. 2019;21(1):248.PubMedPubMedCentralCrossRef Klingberg E, Magnusson MK, Strid H, Deminger A, Stahl A, Sundin J, et al. A distinct gut microbiota composition in patients with ankylosing spondylitis is associated with increased levels of fecal calprotectin. Arthritis Res Ther. 2019;21(1):248.PubMedPubMedCentralCrossRef
22.••
Zurück zum Zitat Asquith M, Sternes PR, Costello ME, Karstens L, Diamond S, Martin TM, et al. HLA alleles associated with risk of ankylosing spondylitis and rheumatoid arthritis influence the gut microbiome. Arthritis Rheumatol. 2019;71(10):1642–50. Demonstrates that HLA-B27 in healthy subjects influences the gut microbiome.PubMedCrossRef Asquith M, Sternes PR, Costello ME, Karstens L, Diamond S, Martin TM, et al. HLA alleles associated with risk of ankylosing spondylitis and rheumatoid arthritis influence the gut microbiome. Arthritis Rheumatol. 2019;71(10):1642–50. Demonstrates that HLA-B27 in healthy subjects influences the gut microbiome.PubMedCrossRef
24.
Zurück zum Zitat Robinson S, Panayi GS. Deficient control of in vitro Epstein-Barr virus infection in patients with ankylosing spondylitis. Ann Rheum Dis. 1986;45:974–7.PubMedPubMedCentralCrossRef Robinson S, Panayi GS. Deficient control of in vitro Epstein-Barr virus infection in patients with ankylosing spondylitis. Ann Rheum Dis. 1986;45:974–7.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Smith JA, Barnes MD, Hong D, DeLay ML, Inman RD, Colbert RA. Gene expression analysis of macrophages derived from ankylosing spondylitis patients reveals interferon-gamma dysregulation. Arthritis Rheum. 2008;58(6):1640–9.PubMedPubMedCentralCrossRef Smith JA, Barnes MD, Hong D, DeLay ML, Inman RD, Colbert RA. Gene expression analysis of macrophages derived from ankylosing spondylitis patients reveals interferon-gamma dysregulation. Arthritis Rheum. 2008;58(6):1640–9.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Fert I, Cagnard N, Glatigny S, Letourneur F, Jacques S, Smith JA, et al. Reverse interferon signature is characteristic of antigen-presenting cells in human and rat spondyloarthritis. Arthritis Rheumatol. 2014;66(4):841–51.PubMedPubMedCentralCrossRef Fert I, Cagnard N, Glatigny S, Letourneur F, Jacques S, Smith JA, et al. Reverse interferon signature is characteristic of antigen-presenting cells in human and rat spondyloarthritis. Arthritis Rheumatol. 2014;66(4):841–51.PubMedPubMedCentralCrossRef
29.••
Zurück zum Zitat Huang H, Sikora MJ, Islam S, Chowdhury RR, Chien YH, Scriba TJ, et al. Select sequencing of clonally expanded CD8(+) T cells reveals limits to clonal expansion. Proc Natl Acad Sci U S A. 2019;116(18):8995. Provides evidence of clonal expansions of CD8 lymphocytes in AS patients.PubMedPubMedCentralCrossRef Huang H, Sikora MJ, Islam S, Chowdhury RR, Chien YH, Scriba TJ, et al. Select sequencing of clonally expanded CD8(+) T cells reveals limits to clonal expansion. Proc Natl Acad Sci U S A. 2019;116(18):8995. Provides evidence of clonal expansions of CD8 lymphocytes in AS patients.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Ciccia F, Guggino G, Rizzo A, Alessandro R, Luchetti MM, Milling S, et al. Dysbiosis and zonulin upregulation alter gut epithelial and vascular barriers in patients with ankylosing spondylitis. Ann Rheum Dis. 2017;76(6):1123–32.PubMedCrossRef Ciccia F, Guggino G, Rizzo A, Alessandro R, Luchetti MM, Milling S, et al. Dysbiosis and zonulin upregulation alter gut epithelial and vascular barriers in patients with ankylosing spondylitis. Ann Rheum Dis. 2017;76(6):1123–32.PubMedCrossRef
31.
Zurück zum Zitat Smith MD, Gibson RA, Brooks PM. Abnormal bowel permeability in ankylosing spondylitis and rheumatoid arthritis. J Rheumatol. 1985;12:299–305.PubMed Smith MD, Gibson RA, Brooks PM. Abnormal bowel permeability in ankylosing spondylitis and rheumatoid arthritis. J Rheumatol. 1985;12:299–305.PubMed
32.
Zurück zum Zitat Martinez-Gonzalez O, Cantero-Hinojosa J, Paule-Sastre P, Gomez-Magan JC, Salcatierra-Rios D. Intestinal permeability in patients with ankylosing spondylitis and their healthy relatives. Br J Rheumatol. 1994;33:644–7.PubMedCrossRef Martinez-Gonzalez O, Cantero-Hinojosa J, Paule-Sastre P, Gomez-Magan JC, Salcatierra-Rios D. Intestinal permeability in patients with ankylosing spondylitis and their healthy relatives. Br J Rheumatol. 1994;33:644–7.PubMedCrossRef
33.
Zurück zum Zitat Chen L, Fischer R, Peng Y, Reeves E, McHugh K, Ternette N, et al. Critical role of endoplasmic reticulum aminopeptidase 1 in determining the length and sequence of peptides bound and presented by HLA-B27. Arthritis & Rheumatology. 2014;66(2):284–94.CrossRef Chen L, Fischer R, Peng Y, Reeves E, McHugh K, Ternette N, et al. Critical role of endoplasmic reticulum aminopeptidase 1 in determining the length and sequence of peptides bound and presented by HLA-B27. Arthritis & Rheumatology. 2014;66(2):284–94.CrossRef
34.
Zurück zum Zitat Martin-Esteban A, Sanz-Bravo A, Guasp P, Barnea E, Admon A, de Castro JAL. Separate effects of the ankylosing spondylitis associated ERAP1 and ERAP2 aminopeptidases determine the influence of their combined phenotype on the HLA-B*27 peptidome. J Autoimmun. 2017;May:28–38.CrossRef Martin-Esteban A, Sanz-Bravo A, Guasp P, Barnea E, Admon A, de Castro JAL. Separate effects of the ankylosing spondylitis associated ERAP1 and ERAP2 aminopeptidases determine the influence of their combined phenotype on the HLA-B*27 peptidome. J Autoimmun. 2017;May:28–38.CrossRef
35.
Zurück zum Zitat Chemin K, Gerstner C, Malmström V. Effector functions of CD4+ T cells at the site of local autoimmune inflammation-lessons from rheumatoid arthritis. Front Immunol. 2019;10:353.PubMedPubMedCentralCrossRef Chemin K, Gerstner C, Malmström V. Effector functions of CD4+ T cells at the site of local autoimmune inflammation-lessons from rheumatoid arthritis. Front Immunol. 2019;10:353.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Alves Sousa AP, Johnson KR, Ohayon J, Zhu J, Muraro PA, Jacobson S. Comprehensive analysis of TCR-β repertoire in patients with neurological immune-mediated disorders. Sci Rep. 2019;9(1):344.PubMedPubMedCentralCrossRef Alves Sousa AP, Johnson KR, Ohayon J, Zhu J, Muraro PA, Jacobson S. Comprehensive analysis of TCR-β repertoire in patients with neurological immune-mediated disorders. Sci Rep. 2019;9(1):344.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Chang CM, Hsu YW, Wong HS, Wei JC, Liu X, Liao HT, et al. Characterization of T-Cell receptor repertoire in patients with rheumatoid arthritis receiving biologic therapies. Dis Markers. 2019;2019:2364943.PubMedPubMedCentralCrossRef Chang CM, Hsu YW, Wong HS, Wei JC, Liu X, Liao HT, et al. Characterization of T-Cell receptor repertoire in patients with rheumatoid arthritis receiving biologic therapies. Dis Markers. 2019;2019:2364943.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Schober K, Voit F, Grassmann S, Müller TR, Eggert J, Jarosch S, et al. Reverse TCR repertoire evolution toward dominant low-affinity clones during chronic CMV infection. Nat Immunol. 2020;21(4):434–41.PubMedCrossRef Schober K, Voit F, Grassmann S, Müller TR, Eggert J, Jarosch S, et al. Reverse TCR repertoire evolution toward dominant low-affinity clones during chronic CMV infection. Nat Immunol. 2020;21(4):434–41.PubMedCrossRef
40.
Zurück zum Zitat Wu TD, Madireddi S, de Almeida PE, Banchereau R, Chen YJ, Chitre AS, et al. Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature. 2020;579(7798):274–8.PubMedCrossRef Wu TD, Madireddi S, de Almeida PE, Banchereau R, Chen YJ, Chitre AS, et al. Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature. 2020;579(7798):274–8.PubMedCrossRef
41.
Zurück zum Zitat Valpione S, Galvani E, Tweedy J, Mundra PA, Banyard A, Middlehurst P, et al. Immune-awakening revealed by peripheral T cell dynamics after one cycle of immunotherapy. Nat Cancer. 2020;1(2):210–21.PubMedPubMedCentralCrossRef Valpione S, Galvani E, Tweedy J, Mundra PA, Banyard A, Middlehurst P, et al. Immune-awakening revealed by peripheral T cell dynamics after one cycle of immunotherapy. Nat Cancer. 2020;1(2):210–21.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Skowera A, Ellis RJ, Varela-Calviño R, Arif S, Huang GC, Van-Krinks C, et al. CTLs are targeted to kill beta cells in patients with type 1 diabetes through recognition of a glucose-regulated preproinsulin epitope. J Clin Invest. 2008;118(10):3390–402.PubMedPubMedCentral Skowera A, Ellis RJ, Varela-Calviño R, Arif S, Huang GC, Van-Krinks C, et al. CTLs are targeted to kill beta cells in patients with type 1 diabetes through recognition of a glucose-regulated preproinsulin epitope. J Clin Invest. 2008;118(10):3390–402.PubMedPubMedCentral
43.
Zurück zum Zitat Ogg GS, Rod Dunbar P, Romero P, Chen JL, Cerundolo V. High frequency of skin-homing melanocyte-specific cytotoxic T lymphocytes in autoimmune vitiligo. J Exp Med. 1998;188(6):1203–8.PubMedPubMedCentralCrossRef Ogg GS, Rod Dunbar P, Romero P, Chen JL, Cerundolo V. High frequency of skin-homing melanocyte-specific cytotoxic T lymphocytes in autoimmune vitiligo. J Exp Med. 1998;188(6):1203–8.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Latorre D, Kallweit U, Armentani E, Foglierini M, Mele F, Cassotta A, et al. T cells in patients with narcolepsy target self-antigens of hypocretin neurons. Nature. 2018;562(7725):63–8.PubMedCrossRef Latorre D, Kallweit U, Armentani E, Foglierini M, Mele F, Cassotta A, et al. T cells in patients with narcolepsy target self-antigens of hypocretin neurons. Nature. 2018;562(7725):63–8.PubMedCrossRef
45.
Zurück zum Zitat Bassing CH, Swat W, Alt FW. The mechanism and regulation of chromosomal V(D)J recombination. Cell. 2002;109(Suppl):S45-55.PubMedCrossRef Bassing CH, Swat W, Alt FW. The mechanism and regulation of chromosomal V(D)J recombination. Cell. 2002;109(Suppl):S45-55.PubMedCrossRef
46.
Zurück zum Zitat Turner SJ, Doherty PC, McCluskey J, Rossjohn J. Structural determinants of T-cell receptor bias in immunity. Nat Rev Immunol. 2006;6(12):883–94.PubMedCrossRef Turner SJ, Doherty PC, McCluskey J, Rossjohn J. Structural determinants of T-cell receptor bias in immunity. Nat Rev Immunol. 2006;6(12):883–94.PubMedCrossRef
47.
48.
Zurück zum Zitat Warren RL, Freeman JD, Zeng T, Choe G, Munro S, Moore R, 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. 2011;21(5):790–7.PubMedPubMedCentralCrossRef Warren RL, Freeman JD, Zeng T, Choe G, Munro S, Moore R, 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. 2011;21(5):790–7.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Wooldridge L, Ekeruche-Makinde J, van den Berg HA, Skowera A, Miles JJ, Tan MP, et al. A single autoimmune T cell receptor recognizes more than a million different peptides. J Biol Chem. 2012;287(2):1168–77.PubMedCrossRef Wooldridge L, Ekeruche-Makinde J, van den Berg HA, Skowera A, Miles JJ, Tan MP, et al. A single autoimmune T cell receptor recognizes more than a million different peptides. J Biol Chem. 2012;287(2):1168–77.PubMedCrossRef
51.
Zurück zum Zitat Gate D, Saligrama N, Leventhal O, Yang AC, Unger MS, Middeldorp J, et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease. Nature. 2020;577(7790):399–404.PubMedPubMedCentralCrossRef Gate D, Saligrama N, Leventhal O, Yang AC, Unger MS, Middeldorp J, et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease. Nature. 2020;577(7790):399–404.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Emerson RO, DeWitt WS, Vignali M, Gravley J, Hu JK, Osborne EJ, et al. Immunosequencing identifies signatures of cytomegalovirus exposure history and HLA-mediated effects on the T cell repertoire. Nat Genet. 2017. Emerson RO, DeWitt WS, Vignali M, Gravley J, Hu JK, Osborne EJ, et al. Immunosequencing identifies signatures of cytomegalovirus exposure history and HLA-mediated effects on the T cell repertoire. Nat Genet. 2017.
53.
Zurück zum Zitat Wardemann H, Busse CE. Novel approaches to analyze immunoglobulin repertoires. Trends Immunol. 2017;38(7):471–82.PubMedCrossRef Wardemann H, Busse CE. Novel approaches to analyze immunoglobulin repertoires. Trends Immunol. 2017;38(7):471–82.PubMedCrossRef
55.
Zurück zum Zitat Dziubianau M, Hecht J, Kuchenbecker L, Sattler A, Stervbo U, Rödelsperger C, et al. TCR repertoire analysis by next generation sequencing allows complex differential diagnosis of T cell-related pathology. Am J Transplant. 2013;13(11):2842–54.PubMedCrossRef Dziubianau M, Hecht J, Kuchenbecker L, Sattler A, Stervbo U, Rödelsperger C, et al. TCR repertoire analysis by next generation sequencing allows complex differential diagnosis of T cell-related pathology. Am J Transplant. 2013;13(11):2842–54.PubMedCrossRef
56.
Zurück zum Zitat Okino ST, Kong M, Sarras H, Wang Y. Evaluation of bias associated with high-multiplex, target-specific pre-amplification. Biomol Detect Quantif. 2016;6:13–21.PubMedCrossRef Okino ST, Kong M, Sarras H, Wang Y. Evaluation of bias associated with high-multiplex, target-specific pre-amplification. Biomol Detect Quantif. 2016;6:13–21.PubMedCrossRef
57.
Zurück zum Zitat Carlson CS, Emerson RO, Sherwood AM, Desmarais C, Chung MW, Parsons JM, et al. Using synthetic templates to design an unbiased multiplex PCR assay. Nat Commun. 2013;4:2680.PubMedCrossRef Carlson CS, Emerson RO, Sherwood AM, Desmarais C, Chung MW, Parsons JM, et al. Using synthetic templates to design an unbiased multiplex PCR assay. Nat Commun. 2013;4:2680.PubMedCrossRef
58.
Zurück zum Zitat Peng Q, Vijaya Satya R, Lewis M, Randad P, Wang Y. Reducing amplification artifacts in high multiplex amplicon sequencing by using molecular barcodes. BMC Genomics. 2015;16(1):589.PubMedPubMedCentralCrossRef Peng Q, Vijaya Satya R, Lewis M, Randad P, Wang Y. Reducing amplification artifacts in high multiplex amplicon sequencing by using molecular barcodes. BMC Genomics. 2015;16(1):589.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Linnemann C, Heemskerk B, Kvistborg P, Kluin RJ, Bolotin DA, Chen X, et al. High-throughput identification of antigen-specific TCRs by TCR gene capture. Nat Med. 2013;19(11):1534–41.PubMedCrossRef Linnemann C, Heemskerk B, Kvistborg P, Kluin RJ, Bolotin DA, Chen X, et al. High-throughput identification of antigen-specific TCRs by TCR gene capture. Nat Med. 2013;19(11):1534–41.PubMedCrossRef
60.
Zurück zum Zitat Rapid amplification of 5' complementary DNA ends (5' RACE). Nat Methods. 2005; 2(8):629–630. Rapid amplification of 5' complementary DNA ends (5' RACE). Nat Methods. 2005; 2(8):629–630.
61.
Zurück zum Zitat Zhu YY, Machleder EM, Chenchik A, Li R, Siebert PD. Reverse transcriptase template switching: a SMART approach for full-length cDNA library construction. Biotechniques. 2001;30(4):892–7.PubMedCrossRef Zhu YY, Machleder EM, Chenchik A, Li R, Siebert PD. Reverse transcriptase template switching: a SMART approach for full-length cDNA library construction. Biotechniques. 2001;30(4):892–7.PubMedCrossRef
62.
Zurück zum Zitat Matz M, Shagin D, Bogdanova E, Britanova O, Lukyanov S, Diatchenko L, et al. Amplification of cDNA ends based on template-switching effect and step-out PCR. Nucleic Acids Res. 1999;27(6):1558–60.PubMedPubMedCentralCrossRef Matz M, Shagin D, Bogdanova E, Britanova O, Lukyanov S, Diatchenko L, et al. Amplification of cDNA ends based on template-switching effect and step-out PCR. Nucleic Acids Res. 1999;27(6):1558–60.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Mamedov IZ, Britanova OV, Zvyagin IV, Turchaninova MA, Bolotin DA, Putintseva EV, et al. Preparing unbiased T-cell receptor and antibody cDNA libraries for the deep next generation sequencing profiling. Front Immunol. 2013;4:456.PubMedPubMedCentralCrossRef Mamedov IZ, Britanova OV, Zvyagin IV, Turchaninova MA, Bolotin DA, Putintseva EV, et al. Preparing unbiased T-cell receptor and antibody cDNA libraries for the deep next generation sequencing profiling. Front Immunol. 2013;4:456.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Shugay M, Britanova OV, Merzlyak EM, Turchaninova MA, Mamedov IZ, Tuganbaev TR, et al. Towards error-free profiling of immune repertoires. Nat Methods. 2014;11(6):653–5.PubMedCrossRef Shugay M, Britanova OV, Merzlyak EM, Turchaninova MA, Mamedov IZ, Tuganbaev TR, et al. Towards error-free profiling of immune repertoires. Nat Methods. 2014;11(6):653–5.PubMedCrossRef
65.
Zurück zum Zitat Bolotin DA, Mamedov IZ, Britanova OV, Zvyagin IV, Shagin D, Ustyugova SV, et al. Next generation sequencing for TCR repertoire profiling: platform-specific features and correction algorithms. Eur J Immunol. 2012;42(11):3073–83.PubMedCrossRef Bolotin DA, Mamedov IZ, Britanova OV, Zvyagin IV, Shagin D, Ustyugova SV, et al. Next generation sequencing for TCR repertoire profiling: platform-specific features and correction algorithms. Eur J Immunol. 2012;42(11):3073–83.PubMedCrossRef
66.
Zurück zum Zitat Kivioja T, Vähärautio A, Karlsson K, Bonke M, Enge M, Linnarsson S, et al. Counting absolute numbers of molecules using unique molecular identifiers. Nat Methods. 2011;9(1):72–4.PubMedCrossRef Kivioja T, Vähärautio A, Karlsson K, Bonke M, Enge M, Linnarsson S, et al. Counting absolute numbers of molecules using unique molecular identifiers. Nat Methods. 2011;9(1):72–4.PubMedCrossRef
67.
Zurück zum Zitat Calis JJ, Rosenberg BR. Characterizing immune repertoires by high throughput sequencing: strategies and applications. Trends Immunol. 2014;35(12):581–90.PubMedPubMedCentralCrossRef Calis JJ, Rosenberg BR. Characterizing immune repertoires by high throughput sequencing: strategies and applications. Trends Immunol. 2014;35(12):581–90.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Li S, Lefranc MP, Miles JJ, Alamyar E, Giudicelli V, Duroux P, et al. IMGT/HighV QUEST paradigm for T cell receptor IMGT clonotype diversity and next generation repertoire immunoprofiling. Nat Commun. 2013;4:2333.PubMedCrossRef Li S, Lefranc MP, Miles JJ, Alamyar E, Giudicelli V, Duroux P, et al. IMGT/HighV QUEST paradigm for T cell receptor IMGT clonotype diversity and next generation repertoire immunoprofiling. Nat Commun. 2013;4:2333.PubMedCrossRef
69.
Zurück zum Zitat Brown SD, Raeburn LA, Holt RA. Profiling tissue-resident T cell repertoires by RNA sequencing. Genome Med. 2015;30(7):125.CrossRef Brown SD, Raeburn LA, Holt RA. Profiling tissue-resident T cell repertoires by RNA sequencing. Genome Med. 2015;30(7):125.CrossRef
70.
Zurück zum Zitat Greiff V, Miho E, Menzel U, Reddy ST. Bioinformatic and statistical analysis of adaptive immune repertoires. Trends Immunol. 2015;36(11):738–49.PubMedCrossRef Greiff V, Miho E, Menzel U, Reddy ST. Bioinformatic and statistical analysis of adaptive immune repertoires. Trends Immunol. 2015;36(11):738–49.PubMedCrossRef
71.
Zurück zum Zitat Miho E, Yermanos A, Weber CR, Berger CT, Reddy ST, Greiff V. Computational strategies for dissecting the high-dimensional complexity of adaptive immune repertoires. Front Immunol. 2018;9:224.PubMedPubMedCentralCrossRef Miho E, Yermanos A, Weber CR, Berger CT, Reddy ST, Greiff V. Computational strategies for dissecting the high-dimensional complexity of adaptive immune repertoires. Front Immunol. 2018;9:224.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Henry VJ, Bandrowski AE, Pepin AS, Gonzalez BJ, Desfeux A. OMICtools: an informative directory for multi-omic data analysis. Database (Oxford). 2014; 2014. Henry VJ, Bandrowski AE, Pepin AS, Gonzalez BJ, Desfeux A. OMICtools: an informative directory for multi-omic data analysis. Database (Oxford). 2014; 2014.
74.
Zurück zum Zitat Miles JJ, Douek DC, Price DA. Bias in the αβ T-cell repertoire: implications for disease pathogenesis and vaccination. Immunol Cell Biol. 2011;89(3):375–87.PubMedCrossRef Miles JJ, Douek DC, Price DA. Bias in the αβ T-cell repertoire: implications for disease pathogenesis and vaccination. Immunol Cell Biol. 2011;89(3):375–87.PubMedCrossRef
75.
Zurück zum Zitat Dash P, Fiore-Gartland AJ, Hertz T, Wang GC, Sharma S, Souquette A, et al. Quantifiable predictive features define epitope-specific T cell receptor repertoires. Nature. 2017;547(7661):89–93.PubMedPubMedCentralCrossRef Dash P, Fiore-Gartland AJ, Hertz T, Wang GC, Sharma S, Souquette A, et al. Quantifiable predictive features define epitope-specific T cell receptor repertoires. Nature. 2017;547(7661):89–93.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Glanville J, Huang H, Nau A, Hatton O, Wagar LE, Rubelt F, et al. Identifying specificity groups in the T cell receptor repertoire. Nature. 2017;547(7661):94–8.PubMedPubMedCentralCrossRef Glanville J, Huang H, Nau A, Hatton O, Wagar LE, Rubelt F, et al. Identifying specificity groups in the T cell receptor repertoire. Nature. 2017;547(7661):94–8.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Ostmeyer J, Christley S, Toby IT, Cowell LG. Biophysicochemical motifs in T-cell receptor sequences distinguish repertoires from tumor-infiltrating lymphocyte and adjacent healthy tissue. Cancer Res. 2019;79(7):1671–80.PubMedPubMedCentralCrossRef Ostmeyer J, Christley S, Toby IT, Cowell LG. Biophysicochemical motifs in T-cell receptor sequences distinguish repertoires from tumor-infiltrating lymphocyte and adjacent healthy tissue. Cancer Res. 2019;79(7):1671–80.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Kim SM, Bhonsle L, Besgen P, Nickel J, Backes A, Held K, et al. Analysis of the paired TCR α- and β-chains of single human T cells. PLoS ONE. 2012;7(5):e37338.PubMedPubMedCentralCrossRef Kim SM, Bhonsle L, Besgen P, Nickel J, Backes A, Held K, et al. Analysis of the paired TCR α- and β-chains of single human T cells. PLoS ONE. 2012;7(5):e37338.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Stubbington MJT, Rozenblatt-Rosen O, Regev A, Teichmann SA. Single-cell transcriptomics to explore the immune system in health and disease. Science. 2017;358(6359):58–63.PubMedPubMedCentralCrossRef Stubbington MJT, Rozenblatt-Rosen O, Regev A, Teichmann SA. Single-cell transcriptomics to explore the immune system in health and disease. Science. 2017;358(6359):58–63.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Proserpio V, Mahata B. Single-cell technologies to study the immune system. Immunology. 2016;147(2):133–40.PubMedCrossRef Proserpio V, Mahata B. Single-cell technologies to study the immune system. Immunology. 2016;147(2):133–40.PubMedCrossRef
81.
Zurück zum Zitat Hermann E, Yu DT, Meyer zumBüschenfelde KH, Fleischer B. HLA-B27-restricted CD8 T cells derived from synovial fluids of patients with reactive arthritis and ankylosing spondylitis. Lancet. 1993;342(8872):646–50.PubMedCrossRef Hermann E, Yu DT, Meyer zumBüschenfelde KH, Fleischer B. HLA-B27-restricted CD8 T cells derived from synovial fluids of patients with reactive arthritis and ankylosing spondylitis. Lancet. 1993;342(8872):646–50.PubMedCrossRef
82.
Zurück zum Zitat Dulphy N, Peyrat MA, Tieng V, Douay C, Rabian C, Tamouza R, et al. Common intra-articular T cell expansions in patients with reactive arthritis: identical beta-chain junctional sequences and cytotoxicity toward HLA-B27. J Immunol. 1999;162(7):3830–9.PubMed Dulphy N, Peyrat MA, Tieng V, Douay C, Rabian C, Tamouza R, et al. Common intra-articular T cell expansions in patients with reactive arthritis: identical beta-chain junctional sequences and cytotoxicity toward HLA-B27. J Immunol. 1999;162(7):3830–9.PubMed
83.
Zurück zum Zitat May E, Dulphy N, Frauendorf E, Duchmann R, Bowness P, de Castro JAL, et al. Conserved TCR beta chain usage in reactive arthritis; evidence for selection by a putative HLA-B27-associated autoantigen. Tissue Antigens. 2002;60(4):299–308.PubMedCrossRef May E, Dulphy N, Frauendorf E, Duchmann R, Bowness P, de Castro JAL, et al. Conserved TCR beta chain usage in reactive arthritis; evidence for selection by a putative HLA-B27-associated autoantigen. Tissue Antigens. 2002;60(4):299–308.PubMedCrossRef
84.
Zurück zum Zitat Faham M, Carlton V, Moorhead M, Zheng J, Klinger M, Pepin F, et al. Discovery of T cell receptor beta motifs specific to HLA-B27-positive ankylosing spondylitis by deep repertoire sequence analysis. Arthritis Rheumatol. 2017;69(4):774–84.PubMedCrossRef Faham M, Carlton V, Moorhead M, Zheng J, Klinger M, Pepin F, et al. Discovery of T cell receptor beta motifs specific to HLA-B27-positive ankylosing spondylitis by deep repertoire sequence analysis. Arthritis Rheumatol. 2017;69(4):774–84.PubMedCrossRef
85.••
Zurück zum Zitat Hanson AL, Nel HJ, Bradbury L, Phipps J, Thomas R, Le Cao KA, et al. T-cell receptor immunosequencing reveals altered repertoire diversity and disease-associated clonal expansions in ankylosing spondylitis patients. Arthritis Rheumatol. 2020;72:1289–302. Demonstrates differences in CD4 and CD8 diversity in AS patients compared with HLA-B27 matched controls, and of CD8 T-cell expansions in AS patients.PubMedCrossRef Hanson AL, Nel HJ, Bradbury L, Phipps J, Thomas R, Le Cao KA, et al. T-cell receptor immunosequencing reveals altered repertoire diversity and disease-associated clonal expansions in ankylosing spondylitis patients. Arthritis Rheumatol. 2020;72:1289–302. Demonstrates differences in CD4 and CD8 diversity in AS patients compared with HLA-B27 matched controls, and of CD8 T-cell expansions in AS patients.PubMedCrossRef
86.••
Zurück zum Zitat Zheng M, Zhang X, Zhou Y, Tang J, Han Q, Zhang Y, et al. TCR repertoire and CDR3 motif analyses depict the role of αβ T cells in ankylosing spondylitis. EBioMedicine. 2019;47:414–26. (Provides evidence of CD8 expansions in AS patients from a different ancestral background and environment.PubMedPubMedCentralCrossRef Zheng M, Zhang X, Zhou Y, Tang J, Han Q, Zhang Y, et al. TCR repertoire and CDR3 motif analyses depict the role of αβ T cells in ankylosing spondylitis. EBioMedicine. 2019;47:414–26. (Provides evidence of CD8 expansions in AS patients from a different ancestral background and environment.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat McCutcheon M, Wehner N, Wensky A, Kushner M, Doan S, Hsiao L, et al. A sensitive ELISPOT assay to detect low-frequency human T lymphocytes. J Immunol Methods. 1997;210(2):149–66.PubMedCrossRef McCutcheon M, Wehner N, Wensky A, Kushner M, Doan S, Hsiao L, et al. A sensitive ELISPOT assay to detect low-frequency human T lymphocytes. J Immunol Methods. 1997;210(2):149–66.PubMedCrossRef
88.
Zurück zum Zitat Altman JD, Moss PA, Goulder PJ, Barouch DH, McHeyzer-Williams MG, Bell JI, et al. Phenotypic analysis of antigen-specific T lymphocytes. Science. 1996;274(5284):94–6.PubMedCrossRef Altman JD, Moss PA, Goulder PJ, Barouch DH, McHeyzer-Williams MG, Bell JI, et al. Phenotypic analysis of antigen-specific T lymphocytes. Science. 1996;274(5284):94–6.PubMedCrossRef
89.
Zurück zum Zitat Peng S, Zaretsky JM, Ng AHC, Chour W, Bethune MT, Choi J, et al. Sensitive detection and analysis of neoantigen-specific T cell populations from tumors and blood. Cell Rep. 2019;28(10):2728-2738.e2727.PubMedPubMedCentralCrossRef Peng S, Zaretsky JM, Ng AHC, Chour W, Bethune MT, Choi J, et al. Sensitive detection and analysis of neoantigen-specific T cell populations from tumors and blood. Cell Rep. 2019;28(10):2728-2738.e2727.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Ornatsky O, Baranov VI, Bandura DR, Tanner SD, Dick J. Multiple cellular antigen detection by ICP-MS. J Immunol Methods. 2006;308(1–2):68–76.PubMedCrossRef Ornatsky O, Baranov VI, Bandura DR, Tanner SD, Dick J. Multiple cellular antigen detection by ICP-MS. J Immunol Methods. 2006;308(1–2):68–76.PubMedCrossRef
91.
Zurück zum Zitat Bentzen AK, Marquard AM, Lyngaa R, Saini SK, Ramskov S, Donia M, et al. Large-scale detection of antigen-specific T cells using peptide-MHC-I multimers labeled with DNA barcodes. Nat Biotechnol. 2016;34(10):1037–45.PubMedCrossRef Bentzen AK, Marquard AM, Lyngaa R, Saini SK, Ramskov S, Donia M, et al. Large-scale detection of antigen-specific T cells using peptide-MHC-I multimers labeled with DNA barcodes. Nat Biotechnol. 2016;34(10):1037–45.PubMedCrossRef
92.
Zurück zum Zitat Ng AHC, Peng S, Xu AM, Noh WJ, Guo K, Bethune MT, et al. MATE-Seq: microfluidic antigen-TCR engagement sequencing. Lab Chip. 2019;19(18):3011–21.PubMedCrossRef Ng AHC, Peng S, Xu AM, Noh WJ, Guo K, Bethune MT, et al. MATE-Seq: microfluidic antigen-TCR engagement sequencing. Lab Chip. 2019;19(18):3011–21.PubMedCrossRef
93.
Zurück zum Zitat Ansaldo E, Slayden LC, Ching KL, Koch MA, Wolf NK, Plichta DR, et al. Akkermansia muciniphila induces intestinal adaptive immune responses during homeostasis. Science. 2019;364(6446):1179–84.PubMedPubMedCentralCrossRef Ansaldo E, Slayden LC, Ching KL, Koch MA, Wolf NK, Plichta DR, et al. Akkermansia muciniphila induces intestinal adaptive immune responses during homeostasis. Science. 2019;364(6446):1179–84.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Wang Y, Rubtsov A, Heiser R, White J, Crawford F, Marrack P, et al. Using a baculovirus display library to identify MHC class I mimotopes. Proc Natl Acad Sci U S A. 2005;102(7):2476–81.PubMedPubMedCentralCrossRef Wang Y, Rubtsov A, Heiser R, White J, Crawford F, Marrack P, et al. Using a baculovirus display library to identify MHC class I mimotopes. Proc Natl Acad Sci U S A. 2005;102(7):2476–81.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Rappazzo CG, Huisman BD, Birnbaum ME. Repertoire-scale determination of class II MHC peptide binding via yeast display improves antigen prediction. Nat Commun. 2020;11(1):4414.PubMedPubMedCentralCrossRef Rappazzo CG, Huisman BD, Birnbaum ME. Repertoire-scale determination of class II MHC peptide binding via yeast display improves antigen prediction. Nat Commun. 2020;11(1):4414.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Gee MH, Han A, Lofgren SM, Beausang JF, Mendoza JL, Birnbaum ME, et al. Antigen identification for orphan T cell receptors expressed on tumor-infiltrating lymphocytes. Cell. 2018;172(3):549-563.e516.PubMedCrossRef Gee MH, Han A, Lofgren SM, Beausang JF, Mendoza JL, Birnbaum ME, et al. Antigen identification for orphan T cell receptors expressed on tumor-infiltrating lymphocytes. Cell. 2018;172(3):549-563.e516.PubMedCrossRef
97.
Zurück zum Zitat Birnbaum ME, Mendoza JL, Sethi DK, Dong S, Glanville J, Dobbins J, et al. Deconstructing the peptide-MHC specificity of T cell recognition. Cell. 2014;157(5):1073–87.PubMedPubMedCentralCrossRef Birnbaum ME, Mendoza JL, Sethi DK, Dong S, Glanville J, Dobbins J, et al. Deconstructing the peptide-MHC specificity of T cell recognition. Cell. 2014;157(5):1073–87.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Joglekar AV, Leonard MT, Jeppson JD, Swift M, Li G, Wong S, et al. T cell antigen discovery via signaling and antigen-presenting bifunctional receptors. Nat Methods. 2019;16(2):191–8.PubMedPubMedCentralCrossRef Joglekar AV, Leonard MT, Jeppson JD, Swift M, Li G, Wong S, et al. T cell antigen discovery via signaling and antigen-presenting bifunctional receptors. Nat Methods. 2019;16(2):191–8.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Kisielow J, Obermair FJ, Kopf M. Deciphering CD4(+) T cell specificity using novel MHC-TCR chimeric receptors. Nat Immunol. 2019;20(5):652–62.PubMedCrossRef Kisielow J, Obermair FJ, Kopf M. Deciphering CD4(+) T cell specificity using novel MHC-TCR chimeric receptors. Nat Immunol. 2019;20(5):652–62.PubMedCrossRef
100.
Zurück zum Zitat Houghten RA, Pinilla C, Blondelle SE, Appel JR, Dooley CT, Cuervo JH. Generation and use of synthetic peptide combinatorial libraries for basic research and drug discovery. Nature. 1991;354(6348):84–6.PubMedCrossRef Houghten RA, Pinilla C, Blondelle SE, Appel JR, Dooley CT, Cuervo JH. Generation and use of synthetic peptide combinatorial libraries for basic research and drug discovery. Nature. 1991;354(6348):84–6.PubMedCrossRef
101.
Zurück zum Zitat Pinilla C, Appel JR, Blanc P, Houghten RA. Rapid identification of high affinity peptide ligands using positional scanning synthetic peptide combinatorial libraries. Biotechniques. 1992;13(6):901–5.PubMed Pinilla C, Appel JR, Blanc P, Houghten RA. Rapid identification of high affinity peptide ligands using positional scanning synthetic peptide combinatorial libraries. Biotechniques. 1992;13(6):901–5.PubMed
102.
Zurück zum Zitat Nino-Vasquez JJ, Allicotti G, Borras E, Wilson DB, Valmori D, Simon R, et al. A powerful combination: the use of positional scanning libraries and biometrical analysis to identify cross-reactive T cell epitopes. Mol Immunol. 2004;40(14–15):1063–74.PubMedCrossRef Nino-Vasquez JJ, Allicotti G, Borras E, Wilson DB, Valmori D, Simon R, et al. A powerful combination: the use of positional scanning libraries and biometrical analysis to identify cross-reactive T cell epitopes. Mol Immunol. 2004;40(14–15):1063–74.PubMedCrossRef
103.
Zurück zum Zitat Gundlach BR, Wiesmüller KH, Junt T, Kienle S, Jung G, Walden P. Specificity and degeneracy of minor histocompatibility antigen-specific MHC-restricted CTL. J Immunol. 1996;156(10):3645–51.PubMed Gundlach BR, Wiesmüller KH, Junt T, Kienle S, Jung G, Walden P. Specificity and degeneracy of minor histocompatibility antigen-specific MHC-restricted CTL. J Immunol. 1996;156(10):3645–51.PubMed
104.
Zurück zum Zitat Udaka K, Wiesmüller KH, Kienle S, Jung G, Walden P. Tolerance to amino acid variations in peptides binding to the major histocompatibility complex class I protein H-2Kb. J Biol Chem. 1995;270(41):24130–4.PubMedCrossRef Udaka K, Wiesmüller KH, Kienle S, Jung G, Walden P. Tolerance to amino acid variations in peptides binding to the major histocompatibility complex class I protein H-2Kb. J Biol Chem. 1995;270(41):24130–4.PubMedCrossRef
105.
Zurück zum Zitat Fleckenstein B, Kalbacher H, Muller CP, Stoll D, Halder T, Jung G, et al. New ligands binding to the human leukocyte antigen class II molecule DRB1*0101 based on the activity pattern of an undecapeptide library. Eur J Biochem. 1996;240(1):71–7.PubMedCrossRef Fleckenstein B, Kalbacher H, Muller CP, Stoll D, Halder T, Jung G, et al. New ligands binding to the human leukocyte antigen class II molecule DRB1*0101 based on the activity pattern of an undecapeptide library. Eur J Biochem. 1996;240(1):71–7.PubMedCrossRef
106.
107.
Zurück zum Zitat Jutz S, Leitner J, Schmetterer K, Doel-Perez I, Majdic O, Grabmeier-Pfistershammer K, et al. Assessment of costimulation and coinhibition in a triple parameter T cell reporter line: simultaneous measurement of NF-κB, NFAT and AP-1. J Immunol Methods. 2016;430:10–20.PubMedCrossRef Jutz S, Leitner J, Schmetterer K, Doel-Perez I, Majdic O, Grabmeier-Pfistershammer K, et al. Assessment of costimulation and coinhibition in a triple parameter T cell reporter line: simultaneous measurement of NF-κB, NFAT and AP-1. J Immunol Methods. 2016;430:10–20.PubMedCrossRef
108.
Zurück zum Zitat Israelson M, Stepanov A, Staroverov D, Shagina I, Misorin A, Evstratieva A, et al. Testing of monoclonal antibodies against the T-cell receptor associated with ankylosing spondylitis. Bull Russian State Med J. 2018;2018(5):71–80. Israelson M, Stepanov A, Staroverov D, Shagina I, Misorin A, Evstratieva A, et al. Testing of monoclonal antibodies against the T-cell receptor associated with ankylosing spondylitis. Bull Russian State Med J. 2018;2018(5):71–80.
109.
Zurück zum Zitat Pruijn GJ, Wiik A, van Venrooij WJ. The use of citrullinated peptides and proteins for the diagnosis of rheumatoid arthritis. Arthritis Res Ther. 2010;12(1):203.PubMedPubMedCentralCrossRef Pruijn GJ, Wiik A, van Venrooij WJ. The use of citrullinated peptides and proteins for the diagnosis of rheumatoid arthritis. Arthritis Res Ther. 2010;12(1):203.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Bell GM, Anderson AE, Diboll J, Reece R, Eltherington O, Harry RA, et al. Autologous tolerogenic dendritic cells for rheumatoid and inflammatory arthritis. Ann Rheum Dis. 2017;76(1):227–34.PubMedCrossRef Bell GM, Anderson AE, Diboll J, Reece R, Eltherington O, Harry RA, et al. Autologous tolerogenic dendritic cells for rheumatoid and inflammatory arthritis. Ann Rheum Dis. 2017;76(1):227–34.PubMedCrossRef
111.
Zurück zum Zitat Schittenhelm RB, Sivaneswaran S, Lim Kam Sian TC, Croft NP, Purcell AW. Human leukocyte antigen (HLA) B27 allotype-specific binding and candidate arthritogenic peptides revealed through heuristic clustering of data-independent acquisition mass spectrometry (DIA-MS) data. Mol Cell Proteomics. 2016;15(6):1867–76.PubMedPubMedCentralCrossRef Schittenhelm RB, Sivaneswaran S, Lim Kam Sian TC, Croft NP, Purcell AW. Human leukocyte antigen (HLA) B27 allotype-specific binding and candidate arthritogenic peptides revealed through heuristic clustering of data-independent acquisition mass spectrometry (DIA-MS) data. Mol Cell Proteomics. 2016;15(6):1867–76.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Schittenhelm RB, Sian TC, Wilmann PG, Dudek NL, Purcell AW. Revisiting the arthritogenic peptide theory: quantitative not qualitative changes in the peptide repertoire of HLA-B27 allotypes. Arthritis Rheumatol. 2015;67(3):702–13.PubMedCrossRef Schittenhelm RB, Sian TC, Wilmann PG, Dudek NL, Purcell AW. Revisiting the arthritogenic peptide theory: quantitative not qualitative changes in the peptide repertoire of HLA-B27 allotypes. Arthritis Rheumatol. 2015;67(3):702–13.PubMedCrossRef
113.
Zurück zum Zitat Kenna TJ, Thomas R, Steptoe RJ. Steady-state dendritic cells expressing cognate antigen terminate memory CD8+ T-cell responses. Blood. 2008;111(4):2091–100.PubMedCrossRef Kenna TJ, Thomas R, Steptoe RJ. Steady-state dendritic cells expressing cognate antigen terminate memory CD8+ T-cell responses. Blood. 2008;111(4):2091–100.PubMedCrossRef
114.
Zurück zum Zitat Kenna TJ, Waldie T, McNally A, Thomson M, Yagita H, Thomas R, et al. Targeting antigen to diverse APCs inactivates memory CD8+ T cells without eliciting tissue-destructive effector function. J Immunol. 2010;184(2):598–606.PubMedCrossRef Kenna TJ, Waldie T, McNally A, Thomson M, Yagita H, Thomas R, et al. Targeting antigen to diverse APCs inactivates memory CD8+ T cells without eliciting tissue-destructive effector function. J Immunol. 2010;184(2):598–606.PubMedCrossRef
115.
Zurück zum Zitat Wright GP, Notley CA, Xue SA, Bendle GM, Holler A, Schumacher TN, et al. Adoptive therapy with redirected primary regulatory T cells results in antigen-specific suppression of arthritis. Proc Natl Acad Sci U S A. 2009;106(45):19078–83.PubMedPubMedCentralCrossRef Wright GP, Notley CA, Xue SA, Bendle GM, Holler A, Schumacher TN, et al. Adoptive therapy with redirected primary regulatory T cells results in antigen-specific suppression of arthritis. Proc Natl Acad Sci U S A. 2009;106(45):19078–83.PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat Tseng HW, Glant TT, Brown MA, Kenna TJ, Thomas GP, Pettit AR. Early anti-inflammatory intervention ameliorates axial disease in the proteoglycan-induced spondylitis mouse model of ankylosing spondylitis. BMC Musculoskelet Disord. 2017;18(1):228.PubMedPubMedCentralCrossRef Tseng HW, Glant TT, Brown MA, Kenna TJ, Thomas GP, Pettit AR. Early anti-inflammatory intervention ameliorates axial disease in the proteoglycan-induced spondylitis mouse model of ankylosing spondylitis. BMC Musculoskelet Disord. 2017;18(1):228.PubMedPubMedCentralCrossRef
Metadaten
Titel
T cell Repertoire Profiling and the Mechanism by which HLA-B27 Causes Ankylosing Spondylitis
verfasst von
Jose Garrido-Mesa
Matthew A. Brown
Publikationsdatum
05.10.2022
Verlag
Springer US
Erschienen in
Current Rheumatology Reports / Ausgabe 12/2022
Print ISSN: 1523-3774
Elektronische ISSN: 1534-6307
DOI
https://doi.org/10.1007/s11926-022-01090-6

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.