- Research
- Open access
- Published:
Insights into cytomegalovirus-associated T cell receptors in recipients following allogeneic hematopoietic stem cell transplantation
Virology Journal volume 21, Article number: 236 (2024)
Abstract
Background
Cytomegalovirus (CMV) reactivation is a serious problem in recipients of allogeneic hematopoietic stem cell transplantation. Long-term latency depends on specific T cell immune reconstitution, which identifies various pathogens by T cell receptors (TCRs). However, the mechanisms underlying the selection of CMV-specific TCRs in recipients after transplantation remain unclear.
Methods
Using high-throughput sequencing and bioinformatics analysis, the T cell immune repertoire of seven CMV reactivated recipients (CRRs) were analyzed and compared to those of seven CMV non-activated recipients (CNRs) at an early stage after transplant.
Results
The counts of unique complementarity-determining region 3 (CDR3) were significantly higher in CNRs than in CRRs. The CDR3 clones in the CNRs exhibit higher homogeneity compared to the CRRs. With regard to T cell receptor β-chain variable region (TRBV) and joint region (TRBJ) genotypes, significant differences were observed in the frequencies of TRBV6, BV23, and BV7–8 between the two groups. In addition to TRBV29–1/BJ1–2, TRBV2/BJ2–2, and TRBV12–4/BJ1–5, 11 V-J combinations had significantly different expression levels between CRRs and CNRs.
Conclusions
The differences in TCR diversity, TRBV segments, and TRBV-BJ combinations observed between CNRs and CRRs might be associated with post-transplant CMV reactivation and could serve as a foundation for further research.
Background
Cytomegalovirus (CMV) is a ubiquitous pathogen worldwide and its global mean seroprevalence is 86% [1]. Although CMV infection is usually asymptomatic in immunocompetent individuals, reactivations are common among immunodeficient individuals (e.g., allograft recipients), which leads to CMV-related diseases, graft failure, and even life-threatening consequences [2,3,4]. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a key component of the treatment of many hematological diseases, such as myelodysplastic syndrome, myeloid leukemia, and aggressive T-cell lymphoma [5,6,7]. Therefore, it is necessary to identify the protective factors of CMV reactivation in allo-HSCT recipients.
T cells play important roles in the response against CMV reactivation in allo-HSCT recipients [8,9]. T cell receptors (TCRs) on the cell surface are responsible for the diverse T cell responses to pathogens. TCRs bind to peptide/major histocompatibility complex (MHC) via the complementarity-determining region 3 (CDR3) encoded by variable (V), diversity (D), and joint (J), as well as their rearrangement, splicing, and insertion, which forms the molecular basis of CMV reactivation suppression by CMV-specific T cells. Due to its high variability, CDR3 plays a crucial role in determining the specificity and diversity of TCRs in recognizing viral antigens, such as those presented by CMV. Studies have already demonstrated interactions between CDR3 and CMV, including specific T cell responses triggered by direct stimulation with CMV peptides, the development of antigen peptide vaccines, and analyses of the complementarity between CDR3 and CMV [10,11,12]. Therefore, investigating the role of CDR3 in CMV reactivation is both important and holds significant potential for further exploration. CMV reactivation and specific T cell clones influence the clonality of TCR repertoire after transplant [13]. Previous studies mostly focused on patients with specific human leukocyte antigen (HLA), and few selected particular patients and time points. The mechanism of CDR3 clones related to CMV reactivation after allo-HSCT is still unclear.
In the present study, we used high-throughput next-generation sequencing and bioinformatics analysis to characterize and compare the TCR β-chain repertoire of seven CMV reactivated recipients (CRRs) and seven CMV non-activated recipients (CNRs) at 1 month after the transplant. The results provide more data of CDR3 immune repertoire associated with CMV reactivation.
Methods
Participants and allo-HSCT protocols
Between July 2020 and March 2021, 179 allo-HSCT recipients were prospectively enrolled from the First Affiliated Hospital, College of Medicine, Zhejiang University, China. The study excluded individuals aged < 18 years and those with tumors, comorbid diseases, infusion of chimeric antigen receptor T cells, autologous HSCT, retransplantation, or history of infection with hepatitis B virus, treponema pallidum, mycobacterium tuberculosis, or human immunodeficiency virus. The grafts were from CMV-seropositive donors. Conditioning regimen, graft versus host disease (GVHD) prophylaxis, and antiviral treatment were administered according to protocols of the bone marrow transplant center.
The study participants were followed for at least 9 months and categorized into CRRs, i.e., CMV-seropositive recipients who experienced significant CMV DNAemia, (peak load > 1000 copies/mL) for > 3 weeks; CNRs, i.e., CMV-seropositive recipients with no evidence of CMV DNAemia during follow-up; and other recipients (ORs), i.e., those who had low CMV DNA load or short duration of CMV DNAemia.
Isolation and processing of peripheral blood mononuclear cells (PBMCs)
At day 30 post-HSCT, PBMCs were obtained from the participants using Ficoll density gradient centrifugation and cryopreserved in freezing medium (90% fetal bovine serum + 10% dimethyl sulfoxide). The process of PBMCs isolation was as follows. Whole blood was diluted with an equal volume of phosphate-buffered saline (PBS). The diluted blood was carefully layered over Lymphoprep (StemCell Technologies, Melbourne, Australia) in a 15 mL conical tube. The tubes were centrifuged at 2500 rpm for 20 min at room temperature with no brake applied. After centrifugation, the PBMC layer, located at the interface, was carefully collected. The collected PBMCs were washed twice with PBS and resuspended in the freezing medium for cryopreservation.
Monitoring of CMV DNA load
Ethylenediaminetetraacetic acid-anticoagulant peripheral blood samples were obtained every week during the first 3 months after transplantation and monthly or bi-monthly thereafter.
CMV DNA was extracted from fresh whole blood samples and detected using nuclei acid detection kits (Daan, Zhongshan, China) after grafting. According to the manufacturer’s instructions, load ≥ 500 copies/mL was considered CMV DNAemia.
RNA extraction and evaluation
Total RNA was extracted from cryopreserved PBMCs in accordance with the manufacturer’s instructions (Qiagen, Hilden, Germany). The RNA concentration was determined via spectrophotometry (Nano Drop 2000; ThermoFisher Scientific, Waltham, MA, USA) and the RNA integrity was evaluated via agarose gel electrophoresis.
Polymerase chain reaction (PCR)
The TCR profiling system was used for the construction of the sequencing library and TCR β chain sequencing (ImmuQuad Biotech, Hangzhou, China) as described previously [14,15,16,17]. Briefly, a 5’ rapid amplification of cDNA ends (RACE) unbiased amplification protocol was used. Two rounds of PCR were performed under suitable conditions. Final PCR products were purified using Agencourt AMPure XP beads and analyzed using Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA) to confirm the concentrations required for the sequencing samples.
High throughput sequencing (HTS) and bioinformatics analysis
Sequencing was performed using an Illumina NovaSeq® system with PE150 mode. Raw data were processed by basic filtration, screening, quality control, and merging. Then the sequence was compared to the international ImMunoGeneTics (IMGT) database (http://www.imgt.org).
Finally, gene annotation and bioinformatics analyses were performed. We calculated the quantities and frequencies of various CDR3 gene and amino acid sequences; corresponding formations of V, D, J, and constant (C) segments; the length distribution of the CDR3 region.
Statistical analysis
GraphPad Prism (version 7.0; GraphPad Software Inc., San Diego, CA, USA) and R (version 3.6.2; R Foundation for Statistical Computing, Vienna, Austria) were used to analyze and visualize the results. Continuous variables were compared using the t-test, corrected t’ test, or non-parametric Mann–Whitney U test based on data distribution and homogeneity. Categorical variables were compared using the chi-square test or Fisher’s exact test as appropriate. The level of statistical significance was set at p < 0.05.
Results
Detailed information about selected recipients
Seven CRRs and seven CNRs were selected based on the demographic and follow-up information. Table 1 and Table S1 present the characteristics of study participants involved in the sequencing analyses.
Samples sequencing data
The read counts and CDR3 clone counts from study participants' samples are presented in Table S2. By analyzing these parameters, the study aims to assess differences in the overall abundance of TCR β sequences, the breadth of CDR3 region diversity, and the functional variation in T cell responses. These comparisons provide insights into the potential differences in immune response capabilities, particularly in relation to CMV recognition and reactivity, between the patient groups.
Notably, the counts of unique CDR3 genes and amino acid sequences were significantly higher in CNRs compared to CRRs, with p-values of 0.0307 and 0.0298, respectively (Fig. 1). Additionally, the overlap of shared CDR3 sequences between the two groups was minimal (Fig. S1).
Clonal overlap rate of TCR CDR3 between CRRs and CNRs
The clonal overlap rate between the two groups was calculated using the Baroni–Urbani and Buser (BUB) index, as described in the Supplementary Method. By evaluating the clonal overlap, the study aims to determine whether there are similarities or differences in the T cell repertoires of CRRs and CNRs, which could provide insights into the commonality or uniqueness of their immune responses, particularly in the context of CMV reactivation. The resulting heatmap visually illustrates these overlap rates, highlighting significant differences in clonal overlap between CRRs and CNRs (Fig. 2).
Frequency of T cell receptor β-chain variable region (TRBV) and joint region (TRBJ) use
By analyzing the usage patterns of TRBV and TRBJ regions, the study aims to identify whether certain segments are more commonly associated with either CRRs or CNRs. This could provide insights into how these segments contribute to the immune response, particularly in relation to CMV reactivation and the effectiveness of T cell-mediated immunity in these patient groups.
A total of 61 TRBV segments were identified and grouped into 27 families. Among the 26 TRBV families shared between the two groups, TRBV6 and TRBV23 were significantly more frequently used in CRRs compared to CNRs (p = 0.0212 and 0.0262, respectively; Fig. 3A). Additionally, we analyzed the usage of individual TRBV segments and found that the TRBV7–8 segment was significantly more prevalent in CRRs than in CNRs (p = 0.0023; Fig. S2). In contrast, the analysis of TRBJ segments identified 13 segments (6 from the TRBJ1 family and 7 from the TRBJ2 family), with no significant differences in usage between the two groups, except for TRBJ2–2P, which was present in one CNR (Fig. 3B).
Differences in V-J combinations
V and J segments are the most variable components of TCRs and play a crucial role in targeting a wide range of pathogenic microorganisms. As a result, the combination of V and J segments has been the primary focus of many TCR-related studies. This comparison could reveal how particular V-J pairings contribute to the recognition and response to CMV, and help explain differences in clinical outcomes or immune effectiveness between the groups.
The preferred V/J genes and their combinations are illustrated as Circos plots in Fig. S3. The stacked bar graph below displays the frequencies of differentially selected V-J combinations between the two subject groups (Fig. 4), with detailed data provided in Table S3. Notably, the TRBV16/BJ1–6 and TRBV11–3/BJ1–5 combinations were absent in CRRs but present in most or all CNRs, leading to a significant difference in expression between the groups according to Fisher’s exact tests (p = 0.021 and 0.001, respectively; Table 2). Additionally, nine V-J combinations showed significantly different expression levels between the CRR and CNR groups (Table 2). Specifically, the frequencies of TRBV2/BJ2–2, TRBV14/BJ2–2, TRBV29–1/BJ1–2, TRBV12–4/BJ1–5, and TRBV5–4/BJ1–4 were significantly higher in CNRs than in CRRs. However, this difference did not account for the higher frequency of certain CDR3 gene sequences in the CNR group; instead, it reflected the significantly greater diversity of CDR3 gene sequences in CNRs compared to CRRs (Fig. S4).
CDR3 length distribution
CDR3 length is a critical factor in determining the specificity and binding properties of TCRs. Analysis revealed that the CDR3 length distribution pattern deviated from a Gaussian distribution to varying degrees in both CRRs and CNRs (Fig. S5). The majority of CDR3 lengths ranged from 30 to 60 base pairs (bp) in both groups. There was no significant difference in the average CDR3 length between CRRs and CNRs.
Discussion
CMV reactivation is a serious problem in allo-HSCT recipients [18], which necessitates antiviral treatment with the associated side effects and limitations [19,20]. Therefore, the mechanism underlying anti-CMV immune responses should be elucidated to improve the diagnosis and treatment of CMV reactivation. T cells are the main protection against CMV. The CDR3 region of the β chain of TCRs is formed by rearrangement, insertion, mutation, and deletion of the V, D, and J gene segments, which generated T cells targeting specific pathogens. HTS can determine the CDR3 sequences of TCRs at the molecular level with higher resolution. In the present study, we compared TCR sequences between allo-HSCT recipients with disparate CMV outcomes at an early stage after allo-HSCT and characterized the TCR repertoire, which will provide more meaningful data contributing to the diagnosis and treatment of CMV reactivation.
The diversity of the overall TCR repertoire is positively correlated with the reservoir capacity and homeostatic stability of the immune system, which resists the invasion of diverse pathogens [21,22]. The absolute numbers of clones of unique CDR3 genes and AAs were significantly higher in the CNR group than in the CRR group, consistent with previous studies [23,24].
To analyze the difference in the CDR3 immune repertoire between CRRs and CNRs, we compared the BUB index of CRRs and CNRs. The clone overlap rate was significantly higher in the CNR group than in the CRR group; therefore, individuals with resistance against CMV reactivation had high CDR3 homology.
Previous studies have reported the skewed preferences of BV and BJ in CMV-specific T cells, which are influenced by antigen specificity [25,26]. In the present study, TRBV6, TRBV23, and TRBV7–8 had higher expression levels in CRRs than in CNRs, which suggests that the BV segments were associated with subsequent CMV outcomes and may be promising biomarkers for the early identification of recipients at risk of CMV reactivation. However, additional studies are needed to verify these results.
The V-J gene combination, which contributes to CDR3 diversity and resistance against various pathogens, is one of the most important properties of the TCR repertoire. In a large cohort of 666 individuals with different HLA alleles, Emerson et al. [27] found that 164 clones were significantly enriched in CMV-positive individuals compared to CMV-negative individuals. Consistent with this finding, three V-J combinations (TRBV29–1/BJ1–2, TRBV2/BJ2–2, and TRBV12–4/BJ1–5) were highly expressed in CNRs compared to CRRs. These combinations were also found in pp65 peptides of HLA-A02-restricted CMV-specific T cells in previous studies [28,29]. In addition, TRBV14/BJ2–2 and TRBV5–4/BJ1–4 expression levels were higher in CNRs compared to CRRs, which has not previously been reported. We further investigated the gene sequences of these five V-J combinations and found higher diversity in CNRs than in CRRs. Moreover, the TRBV16/BJ1–6 and TRBV11–3/BJ1–5 gene combinations were detected in most or all CNRs (albeit at low frequencies), but they were observed in few or no CRRs. Our results provide more evidence that TRBV29–1/BJ1–2, TRBV2/BJ2–2, and TRBV12–4/BJ1–5 probably play vital roles in protecting against CMV infection and reactivation, and the newly discovered V-J gene combinations warrant further investigations. Meanwhile, future research needs to pay more attention to the diversity of these V-J combination genes.
Considering the influence of HLA alleles on antigen presentation and TCR recognition, previous studies [30,31] have mainly focused on individuals with one or several types of HLA allele and corresponding restricted peptides. However, CMV reactivation occur in recipients with different HLA alleles. In addition, the previously identified CDR3 sequences are usually observed in specific MHC-restricted populations. These findings suggest that the MHC-restricted CDR3 sequences do not indicate the underlying immune mechanism of anti-CMV T cells in all recipients. In the present study, we identified three V-J gene combinations with significant differences between the two groups. Although these combinations have previously been reported, the CDR3 gene sequences identified in the present study differ from those reported previously. Meanwhile, the V-J gene combinations were identified in an overwhelming majority of recipients, irrespective of the MHC. Therefore, we need further analysis of the TCR CDR3 AA or gene sequences related to CMV reactivation in the future.
The activity of TCRs against specific antigens is not solely determined by CDR3 genes but is also influenced by other physical and chemical properties (e.g., CDR3 length) [32]. In the present study, we respectively analyzed the CDR3 length distribution and mean length in CNRs and CRRs, which can impact the folding of the TCR loop and its affinity to the peptide–MHC complex, and even result in the changes in TCRs [33]. Previous studies have indicated that CDR3 length is normally distributed in the healthy population [32,34]. However, in our study, we observed varying degrees of skewness in CDR3 length, which could be attributed to immune reconstitution and CMV reactivation. Nakasone et al. reported that the preferred AA length in CMV-specific cytotoxic T cells was almost 15 [35]. Consistent with this finding, the most frequently observed CDR3 lengths in our study participants were 45 bp (equivalent to 15 amino acids), which significantly differed from those in influenza A-specific cytotoxic T cells [25]. Additionally, certain diseases are characterized by short CDR3 lengths [36,37]. The mean CDR3 lengths were not different between the two groups, suggesting that recipients in both groups of the present study were exposed to a similar CMV level.
There were several noteworthy features in our study. Firstly, sequencing was performed on a small number of participants. Although we prospectively studied hundreds of recipients, only 14 were included to minimize potential factors that could disrupt the TCR repertoire, such as drug application and lymphocyte infusion. The CMV serostatus of both donors and recipients also affects TCR diversity; however, in this study, all donors and recipients were positive, as CMV infection rates are high in the Chinese population. Secondly, we obtained the sequencing data of total lymphocytes to comprehensively evaluate the data from all T cells, instead of specific subsets, such as cytotoxic CD8 + T cells or CMV antigen tetramer + T cells restricted by HLA alleles [35,38]. Each sequencing method has its advantages. The former method may be more directly related to the immune response against CMV, while it has limitations in cell sorting and the results may not be comprehensive. Additionally, previous studies have reported the significance of T cells in preventing CMV reactivation and infection, regardless of specific HLA types [27,39]. Despite the limitations in sequencing PBMCs, three V-J combinations described in this study were consistent with those from previous reports on CMV-specific T cells, which further support the credibility and value of our results. Thirdly, we focused solely on the β chain of TCRs. However, the α chain also play an important role in the recognition of antigen-MHC complexes, and the binding affinity of TCR α and β chains may also influence the immune responses [40]. Hence, future studies should analyze the TCR α chain as well. Finally, although the TCR repertoire after allo-HSCT is associated with risks of relapse and GVHD [41,42], no relapse was observed in our study participants and only one recipient had acute GVHD.
Conclusion
In conclusion, the early characteristics of the post-transplant TCR repertoire of recipients correlate with CMV outcomes. Our results enhance understanding of the mechanisms underlying the T cell resistance against CMV reactivation.
Availability of data and materials
The datasets used and analysed during the current study are available from the corresponding author on reasonable request.
Abbreviations
- CMV:
-
Cytomegalovirus
- TCR:
-
T cell receptor
- CRR:
-
CMV reactivated recipient
- CNR:
-
CMV non-activated recipient
- OR:
-
Other recipient
- allo-HSCT:
-
Allogeneic hematopoietic stem cell transplantation
- MHC:
-
Major histocompatibility complex
- CDR3:
-
Complementarity-determining region 3
- TRBV:
-
T cell receptor β-chain variable region
- D:
-
Diversity
- TRBJ:
-
T cell receptor β-chain joint region
- C:
-
Constant
- HLA:
-
Human leukocyte antigen
- GVHD:
-
Graft versus host disease
- 5’ RACE:
-
5’ Rapid amplification of cDNA ends
- HTS:
-
High throughput sequencing
- BUB:
-
Baroni–Urbani and Buser
- PBS:
-
Phosphate-buffered saline
References
Manicklal S, Emery VC, Lazzarotto T, Boppana SB, Gupta RK. The, “silent” global burden of congenital cytomegalovirus. Clin Microbiol Rev. 2013;26:86–102. https://doi.org/10.1128/CMR.00062-12.
Green ML, et al. CMV reactivation after allogeneic HCT and relapse risk: evidence for early protection in acute myeloid leukemia. Blood. 2013;122:1316–24. https://doi.org/10.1182/blood-2013-02-487074.
Teira P, et al. Early cytomegalovirus reactivation remains associated with increased transplant-related mortality in the current era: a CIBMTR analysis. Blood. 2016;127:2427–38. https://doi.org/10.1182/blood-2015-11-679639.
Cantoni N, et al. Evidence for a bidirectional relationship between cytomegalovirus replication and acute graft-versus-host disease. Biol Blood Marrow Transplant. 2010;16:1309–14. https://doi.org/10.1016/j.bbmt.2010.03.020.
Estey EH. Acute myeloid leukemia: 2019 update on risk-stratification and management. Am J Hematol. 2018;93:1267–91. https://doi.org/10.1002/ajh.25214.
Zain JM, Hanona P. Aggressive T-cell lymphomas: 2021 Updates on diagnosis, risk stratification and management. Am J Hematol. 2021;96:1027–46. https://doi.org/10.1002/ajh.26270.
Garcia-Manero G, Chien KS, Montalban-Bravo G. Myelodysplastic syndromes: 2021 update on diagnosis, risk stratification and management. Am J Hematol. 2020;95:1399–420. https://doi.org/10.1002/ajh.25950.
Pei XY, et al. Cytomegalovirus-Specific T-Cell Transfer for Refractory Cytomegalovirus Infection After Haploidentical Stem Cell Transplantation: The Quantitative and Qualitative Immune Recovery for Cytomegalovirus. J Infect Dis. 2017;216:945–56. https://doi.org/10.1093/infdis/jix357.
Camargo JF, et al. Deep functional immunophenotyping predicts risk of cytomegalovirus reactivation after hematopoietic cell transplantation. Blood. 2019;133:867–77. https://doi.org/10.1182/blood-2018-10-878918.
Alkassab L, et al. TCR CDR3-CMV antigen chemical complementaries are associated with a worse outcome for renal cell carcinoma. Anticancer Res. 2024;44:1505–11. https://doi.org/10.21873/anticanres.16947.
Bunse L, et al. Common T-cell-receptor motifs and features in patients with cytomegalovirus (CMV)-seronegative end-stage renal disease receiving a peptide vaccination against CMV. Int J Mol Sci. 2022. https://doi.org/10.3390/ijms23031029.
Lee MN, Meyerson M. Antigen identification for HLA class I- and HLA class II-restricted T cell receptors using cytokine-capturing antigen-presenting cells. Sci Immunol. 2021. https://doi.org/10.1126/sciimmunol.abf4001.
Buhler S, et al. Genetic T-cell receptor diversity at 1 year following allogeneic hematopoietic stem cell transplantation. Leukemia. 2020;34:1422–32. https://doi.org/10.1038/s41375-019-0654-y.
Sheng J, et al. Deep sequencing of T-cell receptors for monitoring peripheral CD8(+) T cells in chinese advanced non-small-cell lung cancer patients treated with the anti-PD-L1 antibody. Front Mol Biosci. 2021;8:679130. https://doi.org/10.3389/fmolb.2021.679130.
Jiang H, et al. Efficacy and safety of sintilimab in combination with chemotherapy in previously untreated advanced or metastatic nonsquamous or squamous NSCLC: two cohorts of an open-label, phase 1b study. Cancer Immunol Immunother. 2021;70:857–68. https://doi.org/10.1007/s00262-020-02738-x.
Gao X, et al. Quantitative analysis of thymus-independent donor-derived T cell expansion in transplant patients. Biol Blood Marrow Transplant. 2020;26:242–53. https://doi.org/10.1016/j.bbmt.2019.10.026.
Wang X, et al. Quantitative characterization of T-cell repertoire alteration in Chinese patients with B-cell acute lymphocyte leukemia after CAR-T therapy. Bone Marrow Transplant. 2019;54:2072–80. https://doi.org/10.1038/s41409-019-0625-y.
Kaito S, et al. Heterogeneous impact of cytomegalovirus reactivation on nonrelapse mortality in hematopoietic stem cell transplantation. Blood Adv. 2020;4:1051–61. https://doi.org/10.1182/bloodadvances.2019000814.
Hodowanec AC, Pikis A, Singer ME. The development of therapeutics for the treatment and prevention of CMV disease in the transplant population: a regulatory perspective. J Infect Dis. 2020;221:S109–12. https://doi.org/10.1093/infdis/jiz389.
Marty FM, et al. Outcomes of patients with detectable CMV DNA at randomization in the phase III trial of letermovir for the prevention of CMV infection in allogeneic hematopoietic cell transplantation. Am J Transplant. 2020;20:1703–11. https://doi.org/10.1111/ajt.15764.
Britanova OV, et al. Age-related decrease in TCR repertoire diversity measured with deep and normalized sequence profiling. J Immunol. 2014;192:2689–98. https://doi.org/10.4049/jimmunol.1302064.
Hou X, et al. Analysis of the repertoire features of TCR beta chain CDR3 in human by high-throughput sequencing. Cell Physiol Biochem. 2016;39:651–67. https://doi.org/10.1159/000445656.
Suessmuth Y, et al. CMV reactivation drives posttransplant T-cell reconstitution and results in defects in the underlying TCRbeta repertoire. Blood. 2015;125:3835–50. https://doi.org/10.1182/blood-2015-03-631853.
Kuzich JA, et al. T cell receptor beta locus sequencing early post-allogeneic stem cell transplant identifies patients at risk of initial and recurrent cytomegalovirus infection. Bone Marrow Transplant. 2021;56:2582–90. https://doi.org/10.1038/s41409-021-01354-2.
Chen G, et al. Sequence and structural analyses reveal distinct and highly diverse human CD8(+) TCR repertoires to immunodominant viral antigens. Cell Rep. 2017;19:569–83. https://doi.org/10.1016/j.celrep.2017.03.072.
Ishihara Y, et al. A unique T-cell receptor amino acid sequence selected by human T-cell lymphotropic virus type 1 Tax301–309-specific cytotoxic T cells in HLA-A24:02-positive asymptomatic carriers and adult T-Cell leukemia/lymphoma patients. J Virol. 2017. https://doi.org/10.1128/JVI.00974-17.
Emerson RO, et al. Immunosequencing identifies signatures of cytomegalovirus exposure history and HLA-mediated effects on the T cell repertoire. Nat Genet. 2017;49:659–65. https://doi.org/10.1038/ng.3822.
Hanley PJ, et al. CMV-specific T cells generated from naive T cells recognize atypical epitopes and may be protective in vivo. Sci Transl Med. 2015;7:285ra263. https://doi.org/10.1126/scitranslmed.aaa2546.
Nguyen TH, et al. Recognition of distinct cross-reactive virus-specific CD8+ T cells reveals a unique TCR signature in a clinical setting. J Immunol. 2014;192:5039–49. https://doi.org/10.4049/jimmunol.1303147.
Klarenbeek PL, et al. Deep sequencing of antiviral T-cell responses to HCMV and EBV in humans reveals a stable repertoire that is maintained for many years. PLoS Pathog. 2012;8:e1002889. https://doi.org/10.1371/journal.ppat.1002889.
Nakasone H, et al. Single-cell T-cell receptor-beta analysis of HLA-A*2402-restricted CMV- pp65-specific cytotoxic T-cells in allogeneic hematopoietic SCT. Bone Marrow Transplant. 2014;49:87–94. https://doi.org/10.1038/bmt.2013.122.
Miqueu P, et al. Statistical analysis of CDR3 length distributions for the assessment of T and B cell repertoire biases. Mol Immunol. 2007;44:1057–64. https://doi.org/10.1016/j.molimm.2006.06.026.
Werner L, et al. Altered T cell receptor beta repertoire patterns in pediatric ulcerative colitis. Clin Exp Immunol. 2019;196:1–11. https://doi.org/10.1111/cei.13247.
Robins HS, et al. Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. Blood. 2009;114:4099–107. https://doi.org/10.1182/blood-2009-04-217604.
Nakasone H, et al. Features of repertoire diversity and gene expression in human cytotoxic T cells following allogeneic hematopoietic cell transplantation. Commun Biol. 2021;4:1177. https://doi.org/10.1038/s42003-021-02709-7.
Laplaud DA, et al. Blood T-cell receptor beta chain transcriptome in multiple sclerosis. Characterization of the T cells with altered CDR3 length distribution. Brain. 2004;127:981–95. https://doi.org/10.1093/brain/awh119.
Tong Y, et al. T cell repertoire diversity is decreased in type 1 diabetes patients. Genom Proteom Bioinf. 2016;14:338–48. https://doi.org/10.1016/j.gpb.2016.10.003.
Toya T, et al. T-cell receptor repertoire of cytomegalovirus-specific cytotoxic T-cells after allogeneic stem cell transplantation. Sci Rep. 2020;10:22218. https://doi.org/10.1038/s41598-020-79363-2.
Liu A, et al. Preliminary exploration of HLA-A 1101-restricted human cytomegalovirus glycoprotein B-specific CD8(+) T cells in allogeneic stem-cell transplant recipients. Virus Res. 2014;188:38–44. https://doi.org/10.1016/j.virusres.2014.03.027.
Yu K, Shi J, Lu D, Yang Q. Comparative analysis of CDR3 regions in paired human alphabeta CD8 T cells. FEBS Open Bio. 2019;9:1450–9. https://doi.org/10.1002/2211-5463.12690.
Yew PY, et al. Quantitative characterization of T-cell repertoire in allogeneic hematopoietic stem cell transplant recipients. Bone Marrow Transplant. 2015;50:1227–34. https://doi.org/10.1038/bmt.2015.133.
Leick M, et al. t cell clonal dynamics determined by high-resolution TCR-beta sequencing in recipients after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2020;26:1567–74. https://doi.org/10.1016/j.bbmt.2020.04.026.
Acknowledgements
Not applicable.
Funding
This study was supported by the National Natural Science Foundation of China (Grant Number 30872239), Huadong Medicine Joint Funds of the Zhejiang Provincial Natural Science Foundation of China (Grant Number LHDMZ24H160002), Zhejiang Provincial Natural Science Foundation of China (Grant Number LY14H190002) and Technology Team for Major Infectious Diseases Prevention Control, Warning and Treatment (Grant Number 2009R50041).
Author information
Authors and Affiliations
Contributions
JTX, YJX and JF contributed to the conception and design of the study, SNG and GYG performed the CMV DNA detection. JTX, HQW and XJL performed the remaining assays. RY and JTX performed the statistical analyses. JTX wrote the first draft of the manuscript and JF revised the manuscript. All authors reviewed the manuscript and approved the final version for publication.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Ethics Committee of the First Affiliated Hospital, College of Medicine, Zhejiang University under protocol number 2019-484 on 25 February 2019. Consent was obtained in writing after providing participants with detailed information about the study’s purpose, procedures, and potential risks.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
12985_2024_2511_MOESM1_ESM.docx
The online version contains supplementary material available at https://doi.org/10.1186/s12985-024-02511-x. Table S1. Comprehensive information about the study subjects. Table S2. Overview of samples sequencing data. Table S3. Frequencies of differentially selected V-J combinations between CRR and CNR. Fig. S1. Shared CDR3 genes and amino acids between the CRR and CNR groups. Fig. S2. The percentages of TRBV segment usage in each subject within the CRR and CNR groups. Fig. S3. Usage of V-J combinations by each subject. Fig. S4. Comparison of the gene sequences of five V-J combinations. Fig. S5. Distribution of CDR3 lengths for each subject. Supplementary Method: Introduction to the BUB index.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.
About this article
Cite this article
Xia, J., Xiao, Y., Gui, G. et al. Insights into cytomegalovirus-associated T cell receptors in recipients following allogeneic hematopoietic stem cell transplantation. Virol J 21, 236 (2024). https://doi.org/10.1186/s12985-024-02511-x
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s12985-024-02511-x