scholarly journals The CCR4–NOT deadenylase complex safeguards thymic positive selection by down-regulating aberrant pro-apoptotic gene expression

2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Taku Ito-Kureha ◽  
Takahisa Miyao ◽  
Saori Nishijima ◽  
Toru Suzuki ◽  
Shin-ichi Koizumi ◽  
...  

AbstractA repertoire of T cells with diverse antigen receptors is selected in the thymus. However, detailed mechanisms underlying this thymic positive selection are not clear. Here we show that the CCR4-NOT complex limits expression of specific genes through deadenylation of mRNA poly(A) tails, enabling positive selection. Specifically, the CCR4-NOT complex is up-regulated in thymocytes before initiation of positive selection, where in turn, it inhibits up-regulation of pro-apoptotic Bbc3 and Dab2ip. Elimination of the CCR4-NOT complex permits up-regulation of Bbc3 during a later stage of positive selection, inducing thymocyte apoptosis. In addition, CCR4-NOT elimination up-regulates Dab2ip at an early stage of positive selection. Thus, CCR4-NOT might control thymocyte survival during two-distinct stages of positive selection by suppressing expression levels of pro-apoptotic molecules. Taken together, we propose a link between CCR4-NOT-mediated mRNA decay and T cell selection in the thymus.

2002 ◽  
Vol 195 (10) ◽  
pp. 1349-1358 ◽  
Author(s):  
Karen Honey ◽  
Terry Nakagawa ◽  
Christoph Peters ◽  
Alexander Rudensky

CD4+ T cells are positively selected in the thymus on peptides presented in the context of major histocompatibility complex class II molecules expressed on cortical thymic epithelial cells. Molecules regulating this peptide presentation play a role in determining the outcome of positive selection. Cathepsin L mediates invariant chain processing in cortical thymic epithelial cells, and animals of the I-Ab haplotype deficient in this enzyme exhibit impaired CD4+ T cell selection. To determine whether the selection defect is due solely to the block in invariant chain cleavage we analyzed cathepsin L–deficient mice expressing the I-Aq haplotype which has little dependence upon invariant chain processing for peptide presentation. Our data indicate the cathepsin L defect in CD4+ T cell selection is haplotype independent, and thus imply it is independent of invariant chain degradation. This was confirmed by analysis of I-Ab mice deficient in both cathepsin L and invariant chain. We show that the defect in positive selection in the cathepsin L−/− thymus is specific for CD4+ T cells that can be selected in a wild-type and provide evidence that the repertoire of T cells selected differs from that in wild-type mice, suggesting cortical thymic epithelial cells in cathepsin L knockout mice express an altered peptide repertoire. Thus, we propose a novel role for cathepsin L in regulating positive selection by generating the major histocompatibility complex class II bound peptide ligands presented by cortical thymic epithelial cells.


2020 ◽  
Vol 38 (17) ◽  
pp. 1938-1950 ◽  
Author(s):  
Nirali N. Shah ◽  
Steven L. Highfill ◽  
Haneen Shalabi ◽  
Bonnie Yates ◽  
Jianjian Jin ◽  
...  

PURPOSE Patients with B-cell acute lymphoblastic leukemia who experience relapse after or are resistant to CD19-targeted immunotherapies have limited treatment options. Targeting CD22, an alternative B-cell antigen, represents an alternate strategy. We report outcomes on the largest patient cohort treated with CD22 chimeric antigen receptor (CAR) T cells. PATIENTS AND METHODS We conducted a single-center, phase I, 3 + 3 dose-escalation trial with a large expansion cohort that tested CD22-targeted CAR T cells for children and young adults with relapsed/refractory CD22+ malignancies. Primary objectives were to assess the safety, toxicity, and feasibility. Secondary objectives included efficacy, CD22 CAR T-cell persistence, and cytokine profiling. RESULTS Fifty-eight participants were infused; 51 (87.9%) after prior CD19-targeted therapy. Cytokine release syndrome occurred in 50 participants (86.2%) and was grade 1-2 in 45 (90%). Symptoms of neurotoxicity were minimal and transient. Hemophagocytic lymphohistiocytosis–like manifestations were seen in 19/58 (32.8%) of subjects, prompting utilization of anakinra. CD4/CD8 T-cell selection of the apheresis product improved CAR T-cell manufacturing feasibility as well as heightened inflammatory toxicities, leading to dose de-escalation. The complete remission rate was 70%. The median overall survival was 13.4 months (95% CI, 7.7 to 20.3 months). Among those who achieved a complete response, the median relapse-free survival was 6.0 months (95% CI, 4.1 to 6.5 months). Thirteen participants proceeded to stem-cell transplantation. CONCLUSION In the largest experience of CD22 CAR T-cells to our knowledge, we provide novel information on the impact of manufacturing changes on clinical outcomes and report on unique CD22 CAR T-cell toxicities and toxicity mitigation strategies. The remission induction rate supports further development of CD22 CAR T cells as a therapeutic option in patients resistant to CD19-targeted immunotherapy.


Blood ◽  
1999 ◽  
Vol 94 (12) ◽  
pp. 4358-4369 ◽  
Author(s):  
Barbara C. Godthelp ◽  
Maarten J.D. van Tol ◽  
Jaak M. Vossen ◽  
Peter J. van den Elsen

To evaluate the role of T-cell selection in the thymus and/or periphery in T-cell immune reconstitution after allogeneic bone marrow transplantation (allo-BMT), we have analyzed the overall and antigen-specific T-cell repertoires in pediatric allo-BMT recipients treated for leukemia. We observed a lack of overall T-cell receptor (TCR) diversity in the repopulating T cells at 3 months after allo-BMT, as was deduced from complementarity determining region 3 (CDR3) size distribution patterns displaying reduced complexity. This was noted particularly in recipients of a T-cell–depleted (TCD) graft and, to a lesser extent, also in recipients of unmanipulated grafts. At 1 year after allo-BMT, normalization was observed of TCR CDR3 size complexity in almost all recipients. Analysis of the antigen-specific T-cell repertoire at 1 year after BMT showed that the T cells responding to tetanus toxoid (TT) differed in TCR gene segment usage and in amino acid composition of the CDR3 region when comparing the recipient with the donor. Moreover, the TT-specific TCR repertoire was found to be stable within a given allo-BMT recipient, because TT-specific T cells with completely identical TCRs were found at 3 consecutive years after transplantation. These observations suggest an important role for T-cell selection processes in the complete restoration of the T-cell immune repertoire in children after allo-BMT.


1996 ◽  
Vol 183 (3) ◽  
pp. 1111-1118 ◽  
Author(s):  
J P DiSanto ◽  
D Guy-Grand ◽  
A Fisher ◽  
A Tarakhovsky

The common cytokine receptor gamma chain (gammac), which is a functional subunit of the receptors for interleukins (IL)-2, -4, -7, -9, and -15, plays an important role in lymphoid development. Inactivation of this molecule in mice leads to abnormal T cell lymphopoiesis characterized by thymic hypoplasia and reduced numbers of peripheral T cells. To determine whether T cell development in the absence of gammac is associated with alterations of intrathymic and peripheral T cell selection, we have analyzed gammac-deficient mice made transgenic for the male-specific T cell receptor (TCR) HY (HY/gammac- mice). In HY/gammac- male mice, negative selection of autoreactive thymocytes was not diminished; however, peripheral T cells expressing transgenic TCR-alpha and -beta chains (TCR-alphaT/betaT) were absent, and extrathymic T cell development was completely abrogated. In HY/gammac- female mice, the expression of the transgenic TCR partially reversed the profound thymic hypoplasia observed in nontransgenic gammac- mice, generating increased numbers of thymocytes in all subsets, particularly the TCR-alphaT/betaT CD8+ single-positive thymocytes. Despite efficient positive selection, however, naive CD8+ TCR-alphaT/betaT T cells were severely reduced in the peripheral lymphoid organs of HY/gammac- female mice. These results not only underscore the indispensible role of gammac in thymocyte development, but also demonstrate the critical role of gammac in the maintenance and/or expansion of peripheral T cell pools.


Cytokine ◽  
2009 ◽  
Vol 48 (1-2) ◽  
pp. 3
Author(s):  
D.C. Otero ◽  
H. Moro ◽  
Y. Tanabe ◽  
Michael David

2000 ◽  
Vol 12 (2) ◽  
pp. 215-222 ◽  
Author(s):  
Linda Fahlén ◽  
Linda Öberg ◽  
Thomas Brännström ◽  
Nelson K. S. Khoo ◽  
Urban Lendahl ◽  
...  

Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 809-809
Author(s):  
Nirali N Shah ◽  
Steven L Highfill ◽  
Haneen Shalabi ◽  
Bonnie Yates ◽  
Eli Kane ◽  
...  

Abstract Background: We previously demonstrated that non-T cells in apheresis products collected from patients with cancer, particularly in those with circulating leukemic cells or low CD3 counts, can substantially affect T cell expansion and transduction and, potentially, in vivo efficacy. Flask adhesion and elutriation methods to remove monocytes and granulocytes, and separation using CD3/CD28 activation beads are inefficient at removing tumor and other inhibitory cells. In the context of an ongoing CD22 CAR clinical trial, we introduced magnetic CD4/CD8 selection prior to activation and lentiviral transduction which resulted in enhanced in vivo CAR T-cell potency compared to products generated prior to the introduction of CD4/CD8 T cell selection. Design: Children and young adults with relapsed/refractory CD22+ hematologic malignancies eligible for our phase I dose escalation anti-CD22 CAR protocol were enrolled on study (NCT02315612). All had bone marrow evaluations at baseline, prior to lympho-depleting chemotherapy (Fludarabine 25 mg/m2 x 3 days and Cyclophosphamide 900 mg/m2 x 1 day) and again at day 28 (+/- 4 days) post-CAR infusion. Three dose levels were explored during dose-escalation (3 x 10e5; 1 x 10e6 and 3 x 10e6 transduced CAR T-cells/kg) prior to treating in the expansion phase at the 1 x 10e6 dose level, which is the focus of this report. As a result of a manufacturing failure in an enrolled patient, as of January 2017, the CAR-T manufacturing process was modified to include CD4/CD8 bead selection (TCS) for all subsequent patients. CAR T cell activation, transduction and expansion processes were otherwise unchanged. Results: From December 2014 to July 2017, 30 patients with ALL were treated, of which 22 (73%) were treated at 1 x 10e6 transduced CAR T cells/kg. All patients had active bone marrow involvement at baseline and 18 (82%) had an M2 marrow (>5% blasts) or higher disease burden. Eighteen had a prior transplant and 14 were previously treated with CAR. The first 15 patients were treated using an unselected apheresis product and 7 patients were treated using CD4/CD8 TCS. The first patient treating using TCS initially had a product failure when CAR-T cells were manufactured without TCS, due in part to a low CD3 at the time of collection resulting in substantial numbers of non-T cells in the apheresis product. Following CD4/CD8 TCS, the CD3 T-cell content in the starting product increased from 47% to 90%, T cell proliferation increased from 1.97-fold to 24.2-fold, transduction increased from 2.57% to 33.4% and an MRD negative complete remission was achieved. Following TCS, median transduction efficiencies were significantly higher (33.4% (pre-TCS) vs 40.7%, p=0.02). Cytokine release syndrome (CRS) occurred in 19 of 22 patients and was grade 1 CRS in 12 (4 in the TCS arm); grade 2 in 6 (2 in the TCS arm) and grade 4 in one patient. Amongst those developing CRS, tocilizumab was utilized in 2 of 13 (15%) patients pre-TCS and in 3 of 6 (50%) patients in the TCS arm to prevent higher grade CRS. Steroids were administered to 1 of 13 patients pre-TCS versus 3 of 6 (50%) patients in the TCS arm. Clinical parameters suggestive of a more robust inflammatory response in TCS patients included statistically significantly higher peak IL-6, IL-8, IL-10 and IL-15 levels as well as higher ferritin (Figure), and higher peak CAR-T% (75% vs 82%, p=0.03) with a trend towards higher absolute CAR values. Furthermore, 3 of the 5 responders in the TCS arm had evidence for a secondary expansion with a late rise in ferritin, WBC and CAR-T% with 2 patients having confirmed hemophagocytosis on the bone marrow at day 28, which was not seen in patients enrolled pre-TCS. For the entire cohort, 16 of 21 (76%) patients evaluable for response attained a complete remission (11 of 15 enrolled pre-TCS and 5 of 7 post-TCS). Amongst the TCS group, 5 of 5 who were CD22 CAR naïve attained MRD negative remission; two patients pre-treated with CD22 CAR T at an outside institution experienced poor in vivo CAR expansion possibly due to immune rejection. Conclusion: Ongoing experience on our expansion cohort confirms activity of CD22 CAR T cells resulting in high remission induction rates in relapsed refractory ALL, including responses in patients previously treated with CD19 CAR T cells. T cell selection using CD4/CD8 positive selection led to enhanced in vivo CAR-T expansion resulting in a 100% MRD negative complete remission rate (5 of 5 patients) in CD22 CAR naïve patients. Figure Figure. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1238-1238 ◽  
Author(s):  
Kohei Hosokawa ◽  
Takumi Nishiuchi ◽  
Takamasa Katagiri ◽  
Chizuru Saito ◽  
Hiroyuki Maruyama ◽  
...  

Abstract Background Although T-cell cytokines are believed to play an essential role in the development of acquired aplastic anemia (AA), it remains unclear which cytokines or cytokine signals trigger hematopoietic stem cell (HSC) suppression in AA. Gene expression profiling of bone marrow (BM) T cells may be useful for identifying inciting molecules, but the BM T cells in patients with severe AA are inappropriate subjects for such studies, because their gene expression profile suffers changes due to the T-cell exposure to various cytokines. Some patients with moderate AA present with profound thrombocytopenia and an increase in glycosylphophatidylinositol-anchored protein (GPI-AP)-deficient cells without significant neutropenia and anemia, and eventually progress to pancytopenia that fulfills the criteria of AA. They usually show a rapid and complete response to cyclosporine (CsA) if treated early after the thrombocytopenia is manifested. Such patients with the “pre-AA” state may serve as ideal subjects to clarify which genes are involved in the development of AA. Objectives/methods To identify the genes closely associated with the development of AA responsive to CsA, we carried out gene expression profiling of the BM mononuclear cells (BMMCs) obtained from 6 un-transfused patients with BM failure with increased GPI-AP-deficient cells (two with the pre-AA state and four with moderate AA) and three healthy individuals using a microarray analysis with Agilent Microarrays, and identified genes that were highly expressed in patients' BMMCs at the diagnosis. All patients responded to CsA monotherapy and achieved platelet recovery≥50 x 109/L within one year. Results A total of 1119 genes, including IL17C, CD40LG and NR4A1, were upregulated in the BMMCs of the six patients at onset by at least two-fold the level of healthy individuals. The extent of gene overexpression was more pronounced in two patients with the pre-AA state than in those with MAA (Fig. 1A). A comparison of the gene expression levels in BMMCs at onset with those in BMMCs obtained after achieving the platelet count≥100 x 109/L in two rapid responders to CsA revealed 1858 genes, including TNFSF9, NFATC2 and CCL4, that were upregulated. Of note, all three Nr4A family members, including Nr4a1 (Nur77), Nr4a2 (Nurr1) and Nr4a3 (NOR-1), were upregulated at onset (17.4-, 46.5- and 106.4-fold increases compared to those after CsA therapy; 37.7-, 17.9- and 72.9-fold increases compared to healthy controls, respectively) in patients who obtained the good platelet response within three months of CsA therapy (rapid responders). The expression levels of Nr4A genes in four AA patients who showed a slow response to CsA (slow responders) were comparable to those of healthy individuals (Fig. 1B). Conclusions Genes in the Nr4a subfamily are immediate early genes that can be induced by a wide array of stimuli, including growth factors and inflammatory signals. Nr4a2 is reportedly upregulated in T cells of patients with multiple sclerosis via the calcineurin–NFAT pathway, and plays a pivotal role in mediating cytokine production, such as the production of IL-17 from pathogenic T cells. In patients with early stage AA, some inflammatory stimuli may induce the overexpression of Nr4A, leading to the development of BM failure. Quantification of the Nr4A expression of BMMCs may therefore be useful for diagnosing thrombocytopenia that can progress to AA and also predicting a rapid response to CsA therapy in patients with the early stage AA. Disclosures: Nakao: Alexion: Honoraria, Research Funding.


Blood ◽  
1999 ◽  
Vol 94 (12) ◽  
pp. 4358-4369 ◽  
Author(s):  
Barbara C. Godthelp ◽  
Maarten J.D. van Tol ◽  
Jaak M. Vossen ◽  
Peter J. van den Elsen

Abstract To evaluate the role of T-cell selection in the thymus and/or periphery in T-cell immune reconstitution after allogeneic bone marrow transplantation (allo-BMT), we have analyzed the overall and antigen-specific T-cell repertoires in pediatric allo-BMT recipients treated for leukemia. We observed a lack of overall T-cell receptor (TCR) diversity in the repopulating T cells at 3 months after allo-BMT, as was deduced from complementarity determining region 3 (CDR3) size distribution patterns displaying reduced complexity. This was noted particularly in recipients of a T-cell–depleted (TCD) graft and, to a lesser extent, also in recipients of unmanipulated grafts. At 1 year after allo-BMT, normalization was observed of TCR CDR3 size complexity in almost all recipients. Analysis of the antigen-specific T-cell repertoire at 1 year after BMT showed that the T cells responding to tetanus toxoid (TT) differed in TCR gene segment usage and in amino acid composition of the CDR3 region when comparing the recipient with the donor. Moreover, the TT-specific TCR repertoire was found to be stable within a given allo-BMT recipient, because TT-specific T cells with completely identical TCRs were found at 3 consecutive years after transplantation. These observations suggest an important role for T-cell selection processes in the complete restoration of the T-cell immune repertoire in children after allo-BMT.


Sign in / Sign up

Export Citation Format

Share Document