scholarly journals Type I IFN Signaling Constrains IL-17A/F Secretion by γδ T Cells during Bacterial Infections

2010 ◽  
Vol 184 (7) ◽  
pp. 3755-3767 ◽  
Author(s):  
Thomas Henry ◽  
Girish S. Kirimanjeswara ◽  
Thomas Ruby ◽  
Jonathan W. Jones ◽  
Kaitian Peng ◽  
...  
Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2385-2385
Author(s):  
Anri Saito ◽  
Miwako Narita ◽  
Norihiro Watanabe ◽  
Ayumi Yokoyama ◽  
Asuka Sekiguchi ◽  
...  

Abstract In order to establish an efficient anti-tumor cellular immunotherapy using blood γδ T cells, we investigated the cytotoxic activity of γδ T cells expanded from patients with leukemia against autologous leukemia cells and explored the potent methods for enhancing the anti-tumor cytotoxic activity of γδ T cells. We clarified that γδ T cells generated from leukemia patients possess the cytotoxic activity against autologous leukemia cells. Besides, anti-tumor cytotoxic activity of expanded γδ T cells was enhanced by the short-term culture of γδ T cells with type I IFN (IFN-α and IFN-β). The sensitivity of target leukemia cells to γδ T cells was enhanced by the exposure of the target cells to bisphosphonate such as zoledronate, which is one of the antigens recognized by γδ T cells and elevates the content of potent antigen for γδ T cells, isoprenyl pyrophosphate (IPP), in tumor cells. Blood γδ T cells were expanded from anti-CD3 microbead-separated T cells or anti-γδ TCR microbead-separated γδ T cells in the patients with acute myelogenous leukemia by the culture with zoledronate and a low concentration of IL-2 for 1–2 weeks. For the activation of expanded γδ T cells, cultured γδ T cells were exposed with type I IFN for 1–3 days. The supernatant prepared from the culture of type I IFN-activated γδ T cells was assayed for cytokine (IFN-γ, TNF-α, IL-4, IL-5, IL-10) concentration by cytometric bead array. Anti-tumor cytotoxicity of γδ T cells was evaluated by 51Cr-release assay by using purified γδ T cells as effector cells and autologous leukemia cells as target cells. In most patients with acute leukemia, γδ T cells could be markedly expanded by the culture with zoledronate and IL-2 and almost all the expanded γδ T cells possessed Vδ2 TCR. Expanded and purified γδ T cells derived from the patients with leukemia were demonstrated to be cytotoxic against autologous leukemia cells. By the culture of expanded γδ T cells with type I IFN, the expression of the activation marker CD69 and the apoptosis molecule Trail was enhanced at the concentration dependent of type I IFN especially IFN-β. The expanded γδ T cells were shown to produce a remarkable amount of IFN-γ and a considerable amount of TNF-α and the cytokine production was increased by the addition of type I IFN. In addition, the cytotoxic activity of γδ T cells was enhanced by incubating target leukemia cells with zoledronate for 1–2 days. The present study demonstrated that γδ T cells expanded from patient’s blood are cytotoxic to patient’s leukemia cells. It is also demonstrated that there are two methods practically available for enhancing the cytotoxic activity of expanded γδ T cells against leukemia cells, one of which is activating γδ T cells by using type I IFN, and the other is elevating the sensitivity of target cells by using bisphosphonate. These findings implied the possibility that type I IFN-activated γδ T cells could be efficiently applied for cellular immunotherapy in the patients with hematological malignancies who is being administered with bisphosphonate. Moreover, in vivo administration of bisphosphpnate, a low dose of IL-2 and type I IFN could be effective for tumors as γδ T cell-based cellular immunotherapy.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3895-3895
Author(s):  
Anri Saito ◽  
Miwako Narita ◽  
Norihiro Watanabe ◽  
Nozomi Tochiki ◽  
Yumi Hiroi ◽  
...  

Abstract In order to establish an efficient anti-tumor cellular immunotherapy using blood In order to establish an efficient anti-tumor cellular immunotherapy using blood γδ T cells, we investigated the in vitro expansion of γδ T cells in the patients with myeloma and lymphoma by the culture of PB-MNC with bisphosphonate and a low dose of IL-2 and we demonstrated the cytotoxic activity of the expanded γδ T cells against myeloma/lymphoma cells. Simultaneously we explored the potent methods for enhancing the anti-tumor cytotoxic activity of γδ T cells by both directions of activating the expanded γδ T cells and making target tumor cells sensitive to γδ T cells. For the activation of γδ T cells, expanded γδ T cells were exposed with type I IFN, monocyte-derived dendritic cells (mo-DC), or plasmacytoid dendritic cell like cell line PMDC05 (leukemia cell line established from CD4+ CD56+ acute leukemia in our laboratory) for 2 days. For the enhancement of sensitivity of target tumor cell to γδ T cells, we aimed to increase the content of IPP (the potent pyrophosphate antigen for γδ T cells) in tumor cells by decreasing the metabolic downstream of IPP. For decreasing the downstream of IPP, we tried to suppress FPP synthetase, which is involved in downstream metabolism of IPP, by using nitrogen-containing bisphosphonate. In addition, the expression of stress-induced molecules such as MICA/B on target tumor cells was evaluated in association with the level of cytotoxicity of γδ T cells against the tumor cells. Compared with normal control, the patients with myeloma (n=8) demonstrated decreased percentage and counts of PB γδ T cells. Patients with lymphoma (n=7) showed a wide range of values in PB γδ T cells, covering a normal range. Amplification rate of PB γδ T cells by culture with zoledronate and IL-2 varied markedly from patient to patient up to 120 times in myeloma and 90 times in lymphoma. Expanded γδ T cells generated in patients with myeloma/lymphoma were demonstrated to possess the cytotoxic activity against myeloma/lymphoma cells by 51Cr-release assay and CFSE-labeled target cell. The cytotoxic activity of expanded γδ T cells was enhanced by the exposure of γδ T cells with type I IFN (IFN-α and IFN-β). The activation of γδ T cells, which was evaluated by the elevation of CD69 expression, was observed by the exposure of γδ T cells with type I IFN, mo-DC, or PMDC05 for 2 days. The sensitivity of target myeloma/lymphoma cells to γδ T cells was enhanced by the exposure of the target cells to bisphosphonate such as zoledronate. The expression level of MICA/B on target tumor cells was demonstrated to be associated with the potency of cytotoxicity of γδ T cells against the tumor cells. The present study demonstrated that γδ T cells expanded from myeloma/lymphoma patient’s blood are cytotoxic to myeloma/lymphoma cells. There are two methods practically available for enhancing the cytotoxic activity of expanded γδ T cells against myeloma/lymphoma cells, one of which is activating γδ T cells and the other is elevating the sensitivity of target cells by using bisphosphonate.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4762-4762
Author(s):  
Anri Saito ◽  
Miwako Narita ◽  
Norihiro Watanabe ◽  
Nozomi Tochiki ◽  
Noriyuki Satoh ◽  
...  

Abstract In order to establish an efficient gd T cell-mediated immunotherapy for hematological malignancies, we tried to clarify whether γδ T cells could be expanded from blood cells of patients with myeloma, lymphoma and acute leukemia by culture with zoledronate and a low dose of IL-2 and whether the expanded patients’ γδ T cells could kill tumor cells including self tumor cells with sparing normal clone cells. In addition, we explored the methods to enhance the anti-tumor cytotoxicity of the expanded γδ T cells by activating them with type I IFN, monocyte-derived dendritic cells (mo-DCs), or ab T cells. Although γδ T cells could be expanded in patients with myeloma, lymphoma and leukemia as well as normal persons, the amplification rates of gd T cells before and after the culture were varied from patient to patient in the patients with hematological malignancies. γδ T cells generated in patients with myeloma and lymphoma showed a potent cytotoxic ability against myeloma/lymphoma cell lines (RPMI8226, Daudi) as shown in γδ T cells generated in normal persons. In addition, γδ T cells generated in a patient with myeloma and acute leukemia showed a cytotoxic ability against self myeloma or leukemia cells freshly prepared from bone marrow. However, the same γδ T cells were not cytotoxic to normal lymphocytes of the patients. Then the expanded γδ T cells were stimulated with type I IFN, mo-DCs, or αβ T cells and the activation (CD69 expression) and cytotoxicity against tumor cells were examined. By the stimulation with type I IFN, the expression of CD69 and Trail of γδ T cells was increased and the cytotoxic ability of γδ T cells was enhanced at dose-dependent manner of type I IFN. CD69 expression on γδ T cells was enhanced by co-culture with both immature and mature mo-DCs in a cell-number-dependent fashion. CD69 expression was enhanced after the addition of mo-DCs of either autologous or allogeneic origin. Activation of γδ T cells with mo-DCs enhanced anti-tumor cytotoxicity of γδ T cells against RPMI8226 and CML blastic crisis cell line (C2F8) in an effector-to-target ratio-dependent manner. Although CD69 expression of γδ T cells was enhanced by the co-culture with allogeneic ab T cells, autologous ab T cells couldn’t activate γδ T cells. However, autologous ab T cells stimulated with IL-2 or PHA could induce the activation of γδ T cells. The activation of γδ T cells with stimulated αβ T cells required cell-to-cell interaction. These findings suggested that αβ T cells stimulated by allogeneic γδ T cells could activate the same allogeneic γδ T cells. The present data demonstrated that γδ T cells, which could be expanded in vitro from blood cells of the patients with myeloma, lymphoma and leukemia by culture with zoledronate and IL-2, possess an enough cytotoxic ability against tumor cells including self tumor cells with sparing normal cells. These findings suggested that in vitro generated patients’ γδ T cells could be applied to γδ T cell-mediated immunotherapy for hematological malignancies. Besides, potent γδ T cells activated by type I IFN, mo-DCs or activated αβ T cells were considered to be applicable for γδ T cell-mediated immunotherapy.


Cytotherapy ◽  
2006 ◽  
Vol 8 (2) ◽  
pp. 118-129 ◽  
Author(s):  
N. Watanabe ◽  
M. Narita ◽  
A. Yokoyama ◽  
A. Sekiguchi ◽  
A. Saito ◽  
...  

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Norzawani Buang ◽  
Lunnathaya Tapeng ◽  
Victor Gray ◽  
Alessandro Sardini ◽  
Chad Whilding ◽  
...  

AbstractThe majority of patients with systemic lupus erythematosus (SLE) have high expression of type I IFN-stimulated genes. Mitochondrial abnormalities have also been reported, but the contribution of type I IFN exposure to these changes is unknown. Here, we show downregulation of mitochondria-derived genes and mitochondria-associated metabolic pathways in IFN-High patients from transcriptomic analysis of CD4+ and CD8+ T cells. CD8+ T cells from these patients have enlarged mitochondria and lower spare respiratory capacity associated with increased cell death upon rechallenge with TCR stimulation. These mitochondrial abnormalities can be phenocopied by exposing CD8+ T cells from healthy volunteers to type I IFN and TCR stimulation. Mechanistically these ‘SLE-like’ conditions increase CD8+ T cell NAD+ consumption resulting in impaired mitochondrial respiration and reduced cell viability, both of which can be rectified by NAD+ supplementation. Our data suggest that type I IFN exposure contributes to SLE pathogenesis by promoting CD8+ T cell death via metabolic rewiring.


2019 ◽  
Vol 18 (1) ◽  
Author(s):  
Yuanyuan Zhu ◽  
Xiang An ◽  
Xiao Zhang ◽  
Yu Qiao ◽  
Tongsen Zheng ◽  
...  

Abstract The aberrant appearance of DNA in the cytoplasm triggers the activation of cGAS-cGAMP-STING signaling and induces the production of type I interferons, which play critical roles in activating both innate and adaptive immune responses. Recently, numerous studies have shown that the activation of STING and the stimulation of type I IFN production are critical for the anticancer immune response. However, emerging evidence suggests that STING also regulates anticancer immunity in a type I IFN-independent manner. For instance, STING has been shown to induce cell death and facilitate the release of cancer cell antigens. Moreover, STING activation has been demonstrated to enhance cancer antigen presentation, contribute to the priming and activation of T cells, facilitate the trafficking and infiltration of T cells into tumors and promote the recognition and killing of cancer cells by T cells. In this review, we focus on STING and the cancer immune response, with particular attention to the roles of STING activation in the cancer-immunity cycle. Additionally, the negative effects of STING activation on the cancer immune response and non-immune roles of STING in cancer have also been discussed.


2015 ◽  
Vol 195 (10) ◽  
pp. 4650-4659 ◽  
Author(s):  
Dennis Ng ◽  
Blandine Maître ◽  
Derek Cummings ◽  
Albert Lin ◽  
Lesley A. Ward ◽  
...  
Keyword(s):  
T Cells ◽  
Type I ◽  

Blood ◽  
2000 ◽  
Vol 95 (6) ◽  
pp. 2024-2030 ◽  
Author(s):  
Giovanna Schiavoni ◽  
Fabrizio Mattei ◽  
Tiziana Di Pucchio ◽  
Stefano M. Santini ◽  
Laura Bracci ◽  
...  

Abstract In a previous study, we reported that a single injection of cyclophosphamide (CTX) in tumor-bearing mice resulted in tumor eradication when the animals were subsequently injected with tumor-sensitized lymphocytes. Notably, CTX acted by inducing bystander effects on T cells, and the response to the combined CTX/adoptive immunotherapy regimen was inhibited in mice treated with antibodies to mouse interferon (IFN)–/β. In the present study, we have investigated whether CTX induced the expression of type I IFN, and we have characterized the CTX effects on the phenotype of T cells in normal mice. CTX injection resulted in an accumulation of type I IFN messenger RNA in the spleen of inoculated mice, at 24 to 48 hours, that was associated with IFN detection in the majority of the animals. CTX also enhanced the expression of the Ly-6C on spleen lymphocytes. This enhancement was inhibited in mice treated with anti–type I IFN antibodies. Moreover, CTX induced a long-lasting increase in in vivo lymphocyte proliferation and in the percentage of CD44hiCD4+ and CD44hiCD8+T lymphocytes. These results demonstrate that CTX is an inducer of type I IFN in vivo and enhances the number of T cells exhibiting the CD44hi memory phenotype. Since type I IFN has been recently recognized as the important cytokine for the in vivo expansion and long-term survival of memory T cells, we suggest that induction of this cytokine may explain at least part of the immunomodulatory effects observed after CTX treatment. Finally, these findings provide a new rationale for combined treatments with CTX and adoptive immunotherapy in cancer patients.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3797-3797
Author(s):  
Olivier Detournay ◽  
Naima Mazouz ◽  
Michel Goldman ◽  
Michel Toungouz

Abstract The dendritic cell family is composed of different subsets able to differentially govern the immune response. Their potent antigen presenting properties make them an attractive candidate for immunization against pathogens or cancer. In that setting, the recently characterized type I IFN DCs present interesting features including a higher expression of molecules involved in antigen presentation and the ability to trigger both the cellular and humoral arms of the immune responses. In view of the pivotal role of regulatory T cells in limiting the effectiveness of effector cells, we analyzed the interactions between these cells and type I IFN DC. DC generated from monocytes in the presence of IFN-β and IL-3 (DCI3) were activated by the maturation agent poly I:C and compared with the classical myeloid DC generated in the presence of GM-CSF and IL-4 (DCG4). Despite the release of lower amounts of IL-12 after maturation, DCI3 were able to induce a higher IFN-γ production by T lymphocytes during the MLR. Analysis at the mRNA level disclosed that DCI3 over transcribed the IL-6 gene leading to the release of high amounts of the protein both after the maturation process and during the MLR itself. Neutralization of IL-6 revealed that this cytokine specifically contributed to the IFN-γ release induced by DCI3. Finally, depletion of CD25+ T cells prior to the MLR identified these cells as a target for IL-6. We conclude from these results that DCI3 are endowed with the unique property of blocking the suppressive effect of regulatory T cells through high IL-6 production during the MLR. This novel mechanism of T cell control is relevant for the use of this DC type in vaccination strategies.


Sign in / Sign up

Export Citation Format

Share Document