scholarly journals Activation-induced Modification in the CD3 Complex of the γδ T Cell Receptor

2002 ◽  
Vol 196 (10) ◽  
pp. 1355-1361 ◽  
Author(s):  
Sandra M. Hayes ◽  
Karen Laky ◽  
Dalal El-Khoury ◽  
Dietmar J. Kappes ◽  
B.J. Fowlkes ◽  
...  

The T cell antigen receptor complexes expressed on αβ and γδ T cells differ not only in their respective clonotypic heterodimers but also in the subunit composition of their CD3 complexes. The γδ T cell receptors (TCRs) expressed on ex vivo γδ T cells lack CD3δ, whereas αβ TCRs contain CD3δ. While this result correlates with the phenotype of CD3δ−/− mice, in which γδ T cell development is unaffected, it is inconsistent with the results of previous studies reporting that CD3δ is a component of the γδ TCR. Since earlier studies examined the subunit composition of γδ TCRs expressed on activated and expanded peripheral γδ T cells or γδ TCR+ intestinal intraepithelial lymphocytes, we hypothesized that activation and expansion may lead to changes in the CD3 subunit composition of the γδ TCR. Here, we report that activation and expansion do in fact result in the inclusion of a protein, comparable in mass and mobility to CD3δ, in the γδ TCR. Further analyses revealed that this protein is not CD3δ, but instead is a differentially glycosylated form of CD3γ. These results provide further evidence for a major difference in the subunit composition of αβ- and γδ TCR complexes and raise the possibility that modification of CD3γ may have important functional consequences in activated γδ T cells.

2006 ◽  
Vol 74 (2) ◽  
pp. 1097-1105 ◽  
Author(s):  
Darren J. Newton ◽  
Elizabeth M. Andrew ◽  
Jane E. Dalton ◽  
Rainy Mears ◽  
Simon R. Carding

ABSTRACT Although γδ T cells are a common feature of many pathogen-induced immune responses, the factors that influence, promote, or regulate the response of individual γδ T-cell subsets to infection is unknown. Here we show that in the absence of Vγ1+ T cells, novel subsets of γδ T cells, expressing T-cell receptor (TCR)-Vγ chains that normally define TCRγδ+ dendritic epidermal T cells (DETCs) (Vγ5+), intestinal intraepithelial lymphocytes (iIELs) (Vγ7+), and lymphocytes associated with the vaginal epithelia (Vγ6+), are recruited to the spleen in response to bacterial infection in TCR-Vγ1−/− mice. By comparison of phenotype and structure of TCR-Vγ chains and/or -Vδ chains expressed by these novel subsets with those of their epithelium-associated counterparts, the Vγ6+ T cells elicited in infected Vγ1−/− mice were shown to be identical to those found in the reproductive tract, from where they are presumably recruited in the absence of Vγ1+ T cells. By contrast, Vγ5+ and Vγ7+ T cells found in infected Vγ1−/− mice were distinct from Vγ5+ DETCs and Vγ7+ iIELs. Functional analyses of the novel γδ T-cell subsets identified for infected Vγ1−/− mice showed that whereas the Vγ5+ and Vγ7+ subsets may compensate for the absence of Vγ1+ T cells by producing similar cytokines, they do not possess cytocidal activity and they cannot replace the macrophage homeostasis function of Vγ1+ T cells. Collectively, these findings identify novel subsets of γδ T cells, the recruitment and activity of which is under the control of Vγ1+ T cells.


2021 ◽  
Vol 11 (9) ◽  
pp. 923
Author(s):  
Josephine G. M. Strijker ◽  
Ronja Pscheid ◽  
Esther Drent ◽  
Jessica J. F. van der Hoek ◽  
Bianca Koopmans ◽  
...  

Currently ~50% of patients with a diagnosis of high-risk neuroblastoma will not survive due to relapsing or refractory disease. Recent innovations in immunotherapy for solid tumors are highly promising, but the low MHC-I expression of neuroblastoma represents a major challenge for T cell-mediated immunotherapy. Here, we propose a novel T cell-based immunotherapy approach for neuroblastoma, based on the use of TEG002, αβ-T cells engineered to express a defined γδ-T cell receptor, which can recognize and kill target cells independent of MHC-I. In a co-culture killing assay, we showed that 3 out of 6 neuroblastoma organoids could activate TEG002 as measured by IFNγ production. Transcriptional profiling showed this effect correlates with an increased activity of processes involved in interferon signaling and extracellular matrix organization. Analysis of the dynamics of organoid killing by TEG002 over time confirmed that organoids which induced TEG002 activation were efficiently killed independent of their MHC-I expression. Of note, efficacy of TEG002 treatment was superior to donor-matched untransduced αβ-T cells or endogenous γδ-T cells. Our data suggest that TEG002 may be a promising novel treatment option for a subset of neuroblastoma patients.


2006 ◽  
Vol 74 (5) ◽  
pp. 2717-2725 ◽  
Author(s):  
Henri C. van der Heyde ◽  
Joan M. Batchelder ◽  
Matyas Sandor ◽  
William P. Weidanz

ABSTRACT Little is known about the function and regulation of splenic γδ T cells during chronic Plasmodium chabaudi malaria. The splenic γδ T-cell population continues to expand, reaching levels equal to 4 times the number of splenocytes in an uninfected mouse. Splenic γδ T cells from JH −/− mice with chronic malaria expressed Vγ1+ or Vδ4+ in the same ratio as uninfected controls with Vγ1 cells dominating, but the Vγ2 ratio declined about twofold. γδ T cells from G8 mice specific for the TL antigen increased only 2-fold in number, compared with 10-fold in BALB/c controls, but G8 γδ T cells failed to express the B220 activation marker. Elimination of the parasite by drug treatment caused a slow depletion in the number of splenic γδ, CD4+, and CD8+ T cells. Following challenge, drug-cured JH −/− mice exhibited nearly identical parasitemia time courses as naïve controls. Depletion of either CD4+ T cells or γδ T cells from chronically infected JH −/− mice by monoclonal antibody treatment resulted in an immediate and significant (P < 0.05) exacerbation of parasitemia coupled with a marked decrease in splenic γδ T-cell numbers. The number of CD4+ T cells, in contrast, did not decrease in mice after anti-T-cell receptor γδ treatment. The results indicate that cell-mediated immunity against blood-stage malarial parasites during chronic malaria (i) requires the continued presence of blood-stage parasites to remain functional, (ii) is dependent upon both γδ T cells and CD4+ T cells, and (iii) lacks immunological memory.


Blood ◽  
2009 ◽  
Vol 114 (3) ◽  
pp. 564-571 ◽  
Author(s):  
Qian Qi ◽  
Mingcan Xia ◽  
Jianfang Hu ◽  
Elizabeth Hicks ◽  
Archana Iyer ◽  
...  

AbstractThe Tec kinase Itk is critical for the development of αβ T cells as well as differentiation of CD4+ T cells into Th2 cells. Itk null mice have defects in the production of Th2 cytokines; however, they paradoxically have significant elevations in serum IgE. Here we show that Itk null mice have increased numbers of γδ T cells in the thymus and spleen. This includes elevated numbers of CD4+ γδ T cell, the majority of which carry the Vγ1.1 and Vδ6.2/3 γδ T-cell receptor with a distinct phenotype. The development of these CD4+ γδ T cells is T cell intrinsic, independent of either major histocompatibility complex class I or class II, and is favored during development in the absence of Itk. Itk null CD4+ γδ T cells secrete significant amounts of Th2 cytokines and can induce the secretion of IgE by wild-type B cells. Our data indicate that Itk plays important role in regulating γδ T-cell development and function. In addition, our data indicate that the elevated IgE observed in Itk-deficient mice is due in part to the enhanced development of CD4+ γδ T cells in the absence of Itk.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5771-5771
Author(s):  
Giulia Barbarito ◽  
Irma Airoldi ◽  
Alessia Zorzoli ◽  
Alice Bertaina ◽  
Andrea Petretto ◽  
...  

Abstract A new method of graft manipulation based on physical removal of αβ+ T cells and CD19+ B cells, leaving mature NK cells and γδ T cells in the graft, has been recently developed for HLA-haploidentical HSCT. We demonstrated that γδ T cells collected from transplanted patients are endowed with capacity of killing leukemia cells after ex vivo treatment with zoledronic acid (ZOL). Thus, we hypothesized that infusion of ZOL in patients receiving this type of graft, may boost γδ T cell cytotoxic activity against leukemia cells. Thirty-three patients were treated with ZOL every 28 days at least twice. γδ T cells before and after ZOL treatments were studied till at least 7 months after HSCT by high-resolution mass spectrometry, flow-cytometry, and degranulation assay. Proteomic analysis of γd T cells purified from patients showed that, starting from the first infusion, ZOL caused up-regulation of proteins involved in activation processes and immune response, paralleled by down-regulation of proteins involved in proliferation. These findings are consistent with an induction of Vδ2 cell differentiation, paralleled by increased cytotoxicity of both Vδ1 and Vδ2 cells against primary leukemia blasts. Furthermore, a proteomic signature was identified for each individual ZOL treatment. Patients given 3 or more ZOL infusions had a better probability of survival in comparison to those given 1 or 2 treatments. In conclusion,ZOL influences Vδ2 cell activity, determines a specific proteomic signature and enhances anti-leukemia activity, this potentially resulting into an increased anti-tumor effect. Disclosures No relevant conflicts of interest to declare.


Viruses ◽  
2021 ◽  
Vol 13 (10) ◽  
pp. 1987
Author(s):  
Jessica Tuengel ◽  
Sanya Ranchal ◽  
Alexandra Maslova ◽  
Gurpreet Aulakh ◽  
Maria Papadopoulou ◽  
...  

Gamma-delta (γδ) T cells are unconventional T cells that help control cytomegalovirus (CMV) infection in adults. γδ T cells develop early in gestation, and a fetal public γδ T cell receptor (TCR) clonotype is detected in congenital CMV infections. However, age-dependent γδ T cell responses to primary CMV infection are not well-understood. Flow cytometry and TCR sequencing was used to comprehensively characterize γδ T cell responses to CMV infection in a cohort of 32 infants followed prospectively from birth. Peripheral blood γδ T cell frequencies increased during infancy, and were higher among CMV-infected infants relative to uninfected. Clustering analyses revealed associations between CMV infection and activation marker expression on adaptive-like Vδ1 and Vδ3, but not innate-like Vγ9Vδ2 γδ T cell subsets. Frequencies of NKG2C+CD57+ γδ T cells were temporally associated with the quantity of CMV shed in saliva by infants with primary infection. The public γδ TCR clonotype was only detected in CMV-infected infants <120 days old and at lower frequencies than previously described in fetal infections. Our findings support the notion that CMV infection drives age-dependent expansions of specific γδ T cell populations, and provide insight for novel strategies to prevent CMV transmission and disease.


2021 ◽  
Vol 13 (577) ◽  
pp. eabb0192
Author(s):  
Emelie Foord ◽  
Lucas C. M. Arruda ◽  
Ahmed Gaballa ◽  
Charlotte Klynning ◽  
Michael Uhlin

The role of γδ T cells in antitumor immunity has been under investigation for the past two decades, but little is known about their contribution to clinical outcomes in patients. Here, we set out to define the clonotypic, phenotypic, and functional features of γδ T cells in peripheral blood, ascites, and metastatic tumor tissue from patients with advanced epithelial ovarian cancer. T cell receptor (TCR) sequencing of the γ chain revealed that tumor-infiltrating γδ T cells have a unique and skewed repertoire with high TCR diversity and low clonality. In contrast, ascites-derived γδ T cells presented a lower TCR diversity and higher clonality, suggesting a TCR-dependent clonal focusing at this site. Further investigation showed that tumor samples had abundant γδ T cells with a tissue-resident, activation-associated phenotype, less usage of Vγ9 and an impaired response to adaptive-associated stimuli, implying an innate-like activation pathway, rather than an adaptive TCR-engaging pathway, at these tumor sites. Furthermore, high γδ T cell cytokine responsiveness upon stimulation was associated with a favorable outcome for patients in terms of both overall survival and reduced residual tumor burden after primary surgery. Last, the functionality of γδ T cells and patient survival were negatively affected by the proportions of CD39-expressing T cells, highlighting the potential of CD39 as a target to improve γδ T cell responses and unleash their antitumor capabilities.


Blood ◽  
2018 ◽  
Vol 131 (10) ◽  
pp. 1063-1072 ◽  
Author(s):  
Rupert Handgretinger ◽  
Karin Schilbach

Abstract Allogeneic hematopoetic stem cell transplantation (HCT) offers an option for patients with hematologic malignancies, in whom conventional standard therapies failed or are not effective enough to cure the disease. Successful HCT can restore functional hematopoiesis and immune function, and the new donor-derived immune system can exert a graft-versus-leukemia (GVL) effect. However, allogenic HCT can also be associated with serious risks for transplantation-related morbidities or mortalities such as graft-versus-host disease (GVHD) or life-threatening infectious complications. GVHD is caused by alloreactive T lymphocytes, which express the αβ T-cell receptor, whereas lymphocytes expressing the γδ T-cell receptor are not alloreactive and do not induce GVHD but can exhibit potent antileukemia and anti-infectious activities. Therefore, γδ T cells are becoming increasingly interesting in allogeneic HCT, and clinical strategies to exploit the full function of these lymphocytes have been and are being developed. Such strategies comprise the in vivo activation of γδ T cells or subsets after HCT by certain drugs or antibodies or the ex vivo expansion and manipulation of either patient-derived or donor-derived γδ T cells and their subsets and the adoptive transfer of the ex vivo–activated lymphocytes. On the basis of the absence of dysregulated alloreactivity, such approaches could induce potent GVL effects in the absence of GVHD. The introduction of large-scale clinical methods to enrich, isolate, expand, and manipulate γδ T cells will facilitate future clinical studies that aim to exploit the full function of these beneficial nonalloreactive lymphocytes.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1530-1530
Author(s):  
Helene Sicard ◽  
Jean-Francois Rossi ◽  
Philippe Rousselot ◽  
Alexis Collette ◽  
Christine Paiva ◽  
...  

Abstract Background: IPH1101, a chemically-synthesized, structural analogue of γδ T lymphocyte natural phosphoantigens, combined with low doses of IL-2 induces a highly selective proliferation of γδ T cells, non conventional lymphocytes bearing potent effector and regulatory immune functions. Today, IPH1101 is tested in Phase II clinical trials in different indications and settings (alone or in combination), as the first specific γδ T cell-mediated immunotherapy. Whereas γδT cells from healthy donors always proliferate in response to IPH1101 + IL-2, cells from cancer patients often present moderate to very strong impaired proliferative capacity. The reasons for this defect are still unclear, but might result from a suppressive effect induced by the tumour itself. Thus, defective γδ T cell responses might be partly disease-dependent. In some cases, the decreased ability to respond to IPH1101 might also correlate with the stage of the disease or the nature of previous or ongoing treatments. In order to explore in which cancer indications or clinical settings γδ T cell pharmacology is impaired or fully maintained, we have set up a quantitative standardized in vitro “IPH1101 sensitivity test” that requires only a small sample of patient’s PBMC. The objective of this ongoing ex-vivo observational study is to identify types of cancers and settings for which γδ T cell immunotherapy using IPH1101 treatment may be beneficial. Here are reported results from 3 hematological indications: multiple myeloma (MM), follicular lymphoma (FL) and chronic myeloid leukaemia (CML) receiving long term imatinib treatment. Method: Patients with MM, FL and CML have been enrolled at 2 French sites. A small sample of blood (20 mL) is sufficient to prepare PBMCs and culture them in the presence of IPH1101 and IL-2. Results on the extent of in vitro amplification of cells by IPH1101 are available within 8 days and are expressed as % of γδ T cells in the culture and total amplification rate of γδ T cells. Results: Nineteen patients with MM, 31 with FL and 19 with CML receiving imatinib were evaluable in this study. One patient by indication was found strictly not sensitive to IPH1101 stimulation ex vivo, meaning that their γδT cells showed no signs of proliferation in culture. Samples from patients with MM and, to a lesser extent, FL showed a clear impairment of their proliferative response as compared to samples from healthy individuals, with 32% and 10% of MM and FL samples, respectively, demonstrating an impaired response. Finally, in imatinib-treated CML patients, 94% PBMC samples had γδ T cells with full proliferative capacity in response to IPH1101. Conclusion: Ex vivo, γδ T cells from multiple myeloma and follicular lymphoma patients show a low proportion of moderate to strong impairment of their proliferative capacity in response to their specific stimulus IPH1101. For future γδ T cell based immunotherapy trials in these indications, it might be beneficial to consider selecting patients based on such a criteria. In CML patients undergoing long-term treatment with imatinib, γδ T cells have maintained full ability to respond to their specific stimulus (despite the well-known immunosuppressive effects of imatinib). Thus, these results confirm the biological feasibility of combining imatinib and IPH1101 in a clinical trial in CML.


Sign in / Sign up

Export Citation Format

Share Document