scholarly journals Thymic selection can compensate for mutations affecting T cell activation and generate a normal T cell repertoire in mutant mice

2003 ◽  
Vol 101 (1) ◽  
pp. 210-214 ◽  
Author(s):  
S. Kissler ◽  
L. Lu ◽  
H. Cantor
Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4284-4284
Author(s):  
Pingping Zheng ◽  
Adrianne E Vasey ◽  
Jeanette Baker ◽  
Bettina Iliopoulou ◽  
Dennis B Leveson-Gower ◽  
...  

Abstract Graft-versus-host disease (GVHD) occurs when transplanted donors' T cells recognized the recipients' antigens and damaged host tissues and cells, particularly the skin, gut and liver in the acute setting. Although it is well known GVHD is more aggressive and manifests more quickly across major versus minor histocompatibility barrier, little is known comparatively about the donor T cell activation and T cell repertoire changes. To investigate temporal and spatial events of GHVD development, side-by-side transplants were conducted into major and minor-mismatched murine recipients (Balb.c and Balb.b) using hematopoietic cells from the same donor strain(B6). In both models, T cells home to nodal sites by day 3, proliferate, and exit to GVHD target tissues by day 6. Additionally, expression of homing and activation markers was equivalent for all markers examined on day 3. However, tissue migration and proliferation were reduced in the minor model. By day 6, minor-mismatched T cells had increased CD62L retention and reduced P-selectin and CD44 expression. We also found fewer MHC-matched T cells producing IFN-g and TNF-a. Our data show that early events of donor T cell activation are similar in both models, suggesting that the delayed onset and attenuated disease GVHD seen across minor barriers arise from temporal differences in the effector phase, rather than the initiation phase, of GVHD. To further understand the differences across major versus minor histocompatibility barriers on the T cell repertoire and patterns of T cell alloreactivity, we collected a sample of T cells from donor mice used for transplantation, and also sampled gut tissues from syngeneic, major and minor- mismatched transplanted mice on day 9, 9 and 30 respectively at times when the allogeneic groups of mice showed severe GVHD symptoms. To reduce the background of high percentage of TCR pseudogenes in mouse genome, 5'RACE starting from RNA samples and deep sequencing of TCRa and TCRb were applied to investigate whether TCR repertoire of the major and minor-mismatched mice were skewed with clonal expansion, and how among the major and minor-mismatched mice. While we hypothesized that the major MHC mismatched group would have lower diversity because of expanded clones associated with the GVHD, the shannon index of TCRa indicated gut TCR repertoire of major and minor mismatched mice have greater diversity than syngeneic group(P<0.05). We did not see differences in the shannon index of diversity on examination of the TCRb repertoire. Contrary to our expectation, that the TCR repertoire of major mismatch would be skewed towards representation of a few highly expanded clones in the gut, we in fact saw that the TCR repertoire in these mice appeared less skewed. The TCR repertoire of the gut was more highly related among individuals in the major mismatch groups than among or between the other groups. This strongly suggests a more reproducible repertoire structure. To examine if certain T cell clones were more likely to appear reproducibly in the major and/or minor mismatch setting, we compared the shared clones across the major-mismatched transplanted mice. Bhattacharyya coefficients showed that the major-mismatched mice shared more clones with the minor-mismatched group than the syngeneic groups(P<0.05). A total of eight shared TCRa clonotypes among major-mismatched mice are also detected. The common CDR3 clonotypes might be associated with GVHD. We found 7 of them are also in donor CD4 memory cells' repertoire; and one is present in both donor's CD4 and CD8 memory cells. One of the complementarity determining regions sequences is "AASYQGGRALI" which is also in common among minor-mismatched mice. We also measured the repertoire similarity between donor T cells and the gut TCR repertoires of three groups. Bhattacharyya coefficients of TCRa and TCRb between donor T cells and major-mismatched gut repertoire is greater than donor T cells with either syngeneic or minor-mismatched repertoire(P<0.05), which suggest that the major-mismatched mice has more T cells homing to gut which might be associated with GVHD. Our sequencing data showed that major and minor-mismatched transplantation might not cause the clonal expansion in the gut TCR repertoire, but their repertoire patterns are different from the syngeneic groups, which might be associated with T cell alloreactivity and GVHD. Disclosures No relevant conflicts of interest to declare.


2011 ◽  
Vol 94 (3) ◽  
pp. 245-264 ◽  
Author(s):  
Hugo A. Van Den Berg ◽  
Carmen Molina-París ◽  
Andrew K. Sewell

2018 ◽  
Vol 86 (11) ◽  
Author(s):  
Gillian J. Wilson ◽  
Stephen W. Tuffs ◽  
Bryan A. Wee ◽  
Keun Seok Seo ◽  
Nogi Park ◽  
...  

ABSTRACTSuperantigens (SAgs) represent a diverse family of bacterial toxins that induce Vβ-specific T cell proliferation associated with an array of important diseases in humans and animals, including mastitis of dairy cows. However, an understanding of the diversity and distribution of SAg genes among bovineStaphylococcus aureusstrains and their role in the pathogenesis of mastitis is lacking. Population genomic analysis of 195 bovineS. aureusisolates representing 57 unique sequence types revealed that strains encode 2 to 13 distinct SAgs and that the majority of isolates contain 5 or more SAg genes. A genome-scale analysis of bovine reference strain RF122 revealed a complement of 11 predicted SAg genes, which were all expressedin vitro. Detection of specific antibodies in convalescent cows suggests expression of 7 of 11 SAgs during naturalS. aureusinfection. We determined the Vβ T cell activation profile for all functional SAgs encoded by RF122, revealing evidence for bovine host-specific activity among the recently identified RF122-encoded SAgs SElY and SElZ. Remarkably, we discovered that some strains have evolved the capacity to stimulate the entire T cell repertoire of cattle through an array of diverse SAgs, suggesting a key role in bovine immune evasion.


2021 ◽  
Vol 12 ◽  
Author(s):  
Carolin T. Turner ◽  
James Brown ◽  
Emily Shaw ◽  
Imran Uddin ◽  
Evdokia Tsaliki ◽  
...  

ObjectiveIn people living with HIV (PLHIV), we sought to test the hypothesis that long term anti-retroviral therapy restores the normal T cell repertoire, and investigate the functional relationship of residual repertoire abnormalities to persistent immune system dysregulation.MethodsWe conducted a case-control study in PLHIV and HIV-negative volunteers, of circulating T cell receptor repertoires and whole blood transcriptomes by RNA sequencing, complemented by metadata from routinely collected health care records.ResultsT cell receptor sequencing revealed persistent abnormalities in the clonal T cell repertoire of PLHIV, characterized by reduced repertoire diversity and oligoclonal T cell expansion correlated with elevated CD8 T cell counts. We found no evidence that these expansions were driven by cytomegalovirus or another common antigen. Increased frequency of long CDR3 sequences and reduced frequency of public sequences among the expanded clones implicated abnormal thymic selection as a contributing factor. These abnormalities in the repertoire correlated with systems level evidence of persistent T cell activation in genome-wide blood transcriptomes.ConclusionsThe diversity of T cell receptor repertoires in PLHIV on long term anti-retroviral therapy remains significantly depleted, and skewed by idiosyncratic clones, partly attributable to altered thymic output and associated with T cell mediated chronic immune activation. Further investigation of thymic function and the antigenic drivers of T cell clonal selection in PLHIV are critical to efforts to fully re-establish normal immune function.


2019 ◽  
Vol 37 (7_suppl) ◽  
pp. 22-22 ◽  
Author(s):  
Scott Williams ◽  
Simon Keam ◽  
Heloise Halse ◽  
Catherine Mitchell ◽  
Franco Caramia ◽  
...  

22 Background: We characterised the immune response in human prostate cancer (PC) to high dose rate brachytherapy (HDRBT) using serial pre- and post-HDRBT biopsies. Methods: Prostate biopsies and peripheral blood samples were obtained from 9 men with clinically localised PC at baseline and again 14 days following a 10 Gy fraction of HDRBT. Immune infiltrates were characterised using multiplex immunohistology (T cell and pan-immune panels) and targeted gene expression profiling (Nanostring pan-cancer immune panel). T cell repertoire was assessed using T cell receptor (TCR) Vbeta CDR3 deep sequencing. Results: HDRBT induced profound but heterogenous immune responses. At the extremes, one patient had an extensive tumor-infiltrating lymphocyte (TIL) response post-HDRBT, comprising T cells (CD4+ > CD8+ > Treg), B cells, and PDL1+ antigen presenting cells (macrophages and CD11c+ dendritic cells); another had a substantial baseline immune infiltrate eliminated by HDRBT. TIL-hi cases (either before or after HDRBT) had a conserved signature of T cell activation, differentiation, and trafficking, with upregulation of checkpoint molecules PD1, IDO1, BTLA, CTLA4, ICOS, B7-H3 and FoxP3. Those TIL-hi specifically in response to HDRBT also had down-regulation of genes regulating myeloid differentiation and granulocyte chemotaxis pre-HDBRT. Conversely, a monocyte-like myeloid-derived suppressor cell signature was seen in the TIL-lo responders. The median number of dominant clones ( > 1% prevalence) determined by TCR sequencing was 24.5 and 26.5 pre- and post-HDRBT respectively. Only 2 clones (in 1 patient) were present at both time points with the T cell repertoire otherwise being completely new following HDRBT. A subset of PC-associated T cell dominant clones was also present in the PB in all patients, suggesting HDRBT can induce systemic immunity. Conclusions: We have shown that treatment of localized prostate cancer with HDRBT induces a marked clonal TIL infiltrate dominated by T cells likely to be operating under the control of multiple checkpoints. These first-in-human data may be able to inform the rational selection of immunotherapy to combine with HDRBT.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A589-A589
Author(s):  
Alex Histed ◽  
Natasha Girgis ◽  
Miguel Moreta ◽  
Jonathan Soriano ◽  
BS Luke Witt ◽  
...  

BackgroundActivation of T cells requires a specific peptide/HLA (human leukocyte antigen) signal presented by an interacting immune or target cell along with engagement of co-stimulatory molecules or cytokine receptors. Cue Biopharma has developed a proprietary biologics platform, termed Immuno-STAT™ (Selective Targeting and Alteration of T cells), wherein a singular protein framework incorporates peptide/HLA complexes and co-stimulatory or cytokine signals. The CUE-100 series Immuno-STATs selectively deliver rationally engineered IL-2 molecules to antigen-specific T cells. The IL-2 molecules in the CUE-100 series Immuno-STATs contain mutations that attenuate binding to IL-2 receptors alpha and beta, which minimizes activation of regulatory T cells (Tregs) and the irrelevant non-antigen-specific T cell repertoire. We have demonstrated that CUE-100 series Immuno-STATs specific for different antigenic peptides (from HPV16, WT1, MART-1, CMV, Flu, and HIV) induce expansion of functional, oligoclonal, antigen-specific repertoires from human PBMCs. The lead clinical candidate CUE-101, presenting the E711-20 peptide from HPV-16 in the context of HLA-A*02:01, is currently being tested in a Phase 1 clinical trial in recurrent/metastatic head and neck cancer patients with evidence of dose-proportional PK, early pharmacodynamic effects and signals of clinical activity.MethodsCUE-100 series Immuno-STATs were tested with human PBMCs to demonstrate specific T cell activation and expansion. Expanded T cell clonality was assessed by single cell TCR sequencing. Responses of T cells to peptide-presenting targets was evaluated by cytokine staining and by assessing their ability to kill target cells. In vivo activity of CUE-100 series Immuno-STATs was assessed in HLA-A2 transgenic mice.ResultsData demonstrate that the CUE-100 series Immuno-STATs selectively activate antigen-specific CD8+ T cells. Signaling, cell-based assays and cytokine release studies confirmed functional attenuation of the IL-2 components of the CUE-100 series, which allows for a favorable safety and selectivity profile. Immuno-STATs demonstrated robust expansion of CD8+ T cells after primary stimulation of unprimed hPBMCs, or re-stimulation of hPBMCs after initial cognate peptide stimulation. In addition, CUE-100 series Immuno-STATs expanded CD8+ T cells in naïve HLA-A*02 transgenic mice. In both cases the expanded T cells exhibited a polyfunctional response upon challenge with peptide-presenting target cells.ConclusionsThe presented data suggests that CUE-100 series Immuno-STATs have the potential to enhance anti-tumor immune responses by inducing a robust antigen-specific, oligoclonal, polyfunctional T cell repertoire. Early validation of CUE-100 series Immuno-STATs is obtained through the emerging signs of pharmacodynamic and clinical activity in the ongoing Phase I trial with CUE-101.ReferenceQuayle SN, Girgis N, Thapa DR, et al. CUE-101, a Novel HPV16 E7-pHLA-IL-2-Fc Fusion protein, enhances tumor antigen specific T cell activation for the treatment of HPV16-driven malignancies. Clin Cancer Res 2020;26:1953–64.


1988 ◽  
Vol 15 (1-3) ◽  
pp. 169-174 ◽  
Author(s):  
U. Hämmerling ◽  
M. Toulon ◽  
R. G. E. Palfree ◽  
M. K. Hoffmann

Science ◽  
2021 ◽  
Vol 372 (6546) ◽  
pp. eabe9124
Author(s):  
Pirooz Zareie ◽  
Christopher Szeto ◽  
Carine Farenc ◽  
Sachith D. Gunasinghe ◽  
Elizabeth M. Kolawole ◽  
...  

T cell receptor (TCR) recognition of peptide–major histocompatibility complexes (pMHCs) is characterized by a highly conserved docking polarity. Whether this polarity is driven by recognition or signaling constraints remains unclear. Using “reversed-docking” TCRβ-variable (TRBV) 17+ TCRs from the naïve mouse CD8+ T cell repertoire that recognizes the H-2Db–NP366 epitope, we demonstrate that their inability to support T cell activation and in vivo recruitment is a direct consequence of reversed docking polarity and not TCR–pMHCI binding or clustering characteristics. Canonical TCR–pMHCI docking optimally localizes CD8/Lck to the CD3 complex, which is prevented by reversed TCR–pMHCI polarity. The requirement for canonical docking was circumvented by dissociating Lck from CD8. Thus, the consensus TCR–pMHC docking topology is mandated by T cell signaling constraints.


Sign in / Sign up

Export Citation Format

Share Document