scholarly journals IFN Regulatory Factor 8 Represses GM-CSF Expression in T Cells To Affect Myeloid Cell Lineage Differentiation

2015 ◽  
Vol 194 (5) ◽  
pp. 2369-2379 ◽  
Author(s):  
Amy V. Paschall ◽  
Ruihua Zhang ◽  
Chen-Feng Qi ◽  
Kankana Bardhan ◽  
Liang Peng ◽  
...  
Blood ◽  
1993 ◽  
Vol 82 (11) ◽  
pp. 3401-3414 ◽  
Author(s):  
A Barcena ◽  
MO Muench ◽  
AH Galy ◽  
J Cupp ◽  
MG Roncarolo ◽  
...  

Abstract It has been proposed that the CD7 molecule is the first antigen expressed on the membrane of cells committed to the T-cell lineage during human fetal T-cell ontogeny. To further identify the pre-T cell subpopulation that migrates to the thymus early in ontogeny, we analyzed the phenotypic and functional characteristics of the fetal liver populations separated on the basis of CD7 expression. Three populations expressing different levels of CD7 were observed: CD7bright, CD7dull, and CD7-. A CD7bright population depleted of mature T, B, and myeloid cells (lineage negative, lin-) and mostly composed of CD56+ CD34- natural killer cells did not mature into T cells in a fetal thymic organ culture (FTOC) assay and was devoid of myeloid progenitors in a clonal colony-forming cell assay. In contrast, the CD7-/dull CD34+ lin- populations were capable of differentiating into phenotypically mature T cells after injection into FTOC and contained early myeloid progenitors. Here we phenotypically compared the fetal liver CD7 populations with the most immature fetal thymic subset that differentiated in the FTOC assay, namely the triple negative (TN, CD3- CD4-CD8-) thymocytes. Fetal TN lin- expressed high levels of CD34 marker and were further subdivided by their expression of CD1 antigen, because CD1- TN thymocytes express higher levels of CD34 antigen compared with CD1+ TN cells. CD1- lin -TN thymocytes are characterized by expressing high levels of CD2, CD7, and CD34 markers and dull levels of CD5, CD10, and CD28 molecules. We could not find fetal liver pre-T cells with a phenotype equivalent to that of TN thymocytes. Our data show that CD7 does not necessarily identify T-cell precursors during fetal T-cell development and strongly support the hypothesis that the acquisition of early T-cell markers as CD2, CD28, and CD5 molecules on the cell surface of T-cell progenitors takes place intrathymically.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi100-vi101
Author(s):  
Stephen J Bagley ◽  
Cecile Alanio ◽  
Jacob Till ◽  
Aseel Abdalla ◽  
Zev Binder ◽  
...  

Abstract BACKGROUND We have previously demonstrated that high baseline plasma cfDNA concentration is associated with poor survival in patients with newly diagnosed GBM. The mechanism of this association remains unknown. To explore whether differences in the immune landscape between high- vs. low-cfDNA patients may play a role in their divergent clinical outcomes, we phenotyped tumors from patients with high vs. low cfDNA using mass cytometry by time of flight (CyTOF). METHODS We performed CyTOF on frozen tumor infiltrate suspension from a pilot cohort of patients with previously untreated GBM with known baseline plasma cfDNA concentration (Bagley, Clin Cancer Res 2020). CyTOF was used to simultaneously measure expression of 39 molecules related to immune cell lineage, differentiation state, and function. Differences in immune cell infiltrates between high- and low-cfDNA patients were assessed using Mann-Whitney U tests. RESULTS Four patients with high cfDNA (median 57, range 33-90 ng/mL) were compared to six patients with low cfDNA (median 12, range 7-16 ng/mL). Immune cell infiltrates with increased adaptive cells (high monocytes and T cells, p=0.05) were present in high-cfDNA compared to low-cfDNA patients. While > 70% of the infiltrating T cells were exhausted in both groups, the pattern of exhaustion was significantly different in high- vs. low-cfDNA patients, with less CXCR5+CD69+ and more CXCR5-CD69- (p=0.008) progenitor exhausted T cells in cfDNA-high patients. CONCLUSIONS In this GBM pilot study, we demonstrated differences in the tumor immune infiltrate in patients with high vs. low baseline plasma cfDNA concentration. Preclinical studies will be needed to determine if this explains the association between high plasma cfDNA and poor outcomes previously observed in patients. Our results may have implications for the use of cfDNA concentration as a predictive biomarker for immunotherapy, as tumors with more intermediate progenitor (CXCR5-CD69-) exhausted T cells may respond better to PD-1 checkpoint blockade.


2012 ◽  
Vol 188 (12) ◽  
pp. 6389-6398 ◽  
Author(s):  
Paul Spear ◽  
Amorette Barber ◽  
Agnieszka Rynda-Apple ◽  
Charles L. Sentman

2010 ◽  
Vol 30 (20) ◽  
pp. 4922-4939 ◽  
Author(s):  
Mark A. Zarnegar ◽  
Jing Chen ◽  
Ellen V. Rothenberg

ABSTRACT The transcription factor PU.1 is critical for multiple hematopoietic lineages, but different leukocyte types require strictly distinct patterns of PU.1 regulation. PU.1 is required early for T-cell lineage development but then must be repressed by a stage-specific mechanism correlated with commitment. Other lineages require steady, low expression or upregulation. Until now, only the promoter plus a distal upstream regulatory element (URE) could be invoked to explain nearly all Sfpi1 (PU.1) activation and repression, including bifunctional effects of Runx1. However, the URE is dispensable for most Sfpi1 downregulation in early T cells, and we show that it retains enhancer activity in immature T-lineage cells even where endogenous Sfpi1 is repressed. We now present evidence for another complex of conserved noncoding elements that mediate discrete, cell-type-specific regulatory features of Sfpi1, including a myeloid cell-specific activating element and a separate, pro-T-cell-specific silencer element. These elements yield opposite, cell-type-specific responses to Runx1. T-cell-specific repression requires Runx1 acting through multiple nonconsensus sites in the silencer core. These newly characterized sites recruit Runx1 binding in early T cells in vivo and define a functionally specific scaffold for dose-dependent, Runx-mediated repression.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 62-62
Author(s):  
Clint Piper ◽  
Vivan Zhou ◽  
Brian T. Edelson ◽  
Reshma Taneja ◽  
William R. Drobyski

Abstract Damage to the gastrointestinal tract is the major cause of morbidity during acute graft-versus-host disease (GVHD). While T cells are the proximate drivers of GVHD, disease induction and amplification rely on crosstalk between the innate and adaptive arms of the immune system. The cellular and cytokine networks which mediate this interplay, however, are not well understood. We previously identified a colitogenic CD4+ IL-23R+ CD11c+ T cell population that possesses an innate-like gene expression signature, indicating that these cells appear to be positioned at the interface of the innate and adaptive arms of the immune system. Notably, we also observed that these cells had increased expression of bhlhe40 which is a transcription factor that has been shown to regulate the production of GM-CSF. Given the well characterized ability of GM-CSF to activate myeloid cell populations in autoimmunity, we sought to define the role of this transcription factor and cytokine as a potential bridge between innate and adaptive immunity in GVHD. Using a well-defined murine GVHD model [C57BL/6 (H-2b)→Balb/c (H-2d)], we observed that mice transplanted with Rag-1-/- bone marrow (BM) and CD4+ bhlhe40-/- T cells were completely protected from GVHD, whereas animals transplanted with Rag-1-/- BM and wild type (WT) CD4+ T cells uniformly developed lethal disease. Further analysis revealed that CD4+ bhlhe40-/- T cells produced less GM-CSF and more IL-10 than their WT counterparts, and had preferentially less pathological damage in the colon. To examine the specific role of GM-CSF, we employed the same GVHD model along with a corresponding syngeneic control (B6→B6.PL). We observed robust GM-CSF production in allogeneic, but not syngeneic, recipients in all GVHD target tissues, but most prominently in the colon. This was largely attributable to donor-derived CD4+ T cells, as there was little GM-CSF produced by CD8+ T cells. Notably, whereas the vast majority (~80%) of these cells in the lung and liver also produced IFN-γ, ~50% of GM-CSF-expressing CD4+ T cells in the colon only produced GM-CSF, suggesting that these cells might represent a separate CD4+ T cell lineage. In that regard, antibody blockade of IL-6, IL-23 and IL-27 had no effect on the frequency of CD4+ GM-CSF+ T cells, indicating that the development of these cells was not regulated by cytokines affecting TH1 and TH17 differentiation. To define the functional significance of donor T cell-derived GM-CSF, recipients were transplanted with BM or BM plus splenocytes from WT or GM-CSF-/- animals. Recipients of GM-CSF-/- grafts had significantly increased survival when compared to WT controls. Furthermore, histological analysis demonstrated a significant reduction in pathology in the colon of animals that received GM-CSF-/- grafts, as well as a decrease in infiltrating TH1 cells, whereas there was no difference in pathological damage in the lung or liver. A similar outcome was observed in complementary experiments in which recipient animals that were treated with an anti-GM-CSF antibody had significantly increased survival compared to mice treated with an isotype control antibody. To confirm a role for GM-CSF signaling in CD4+ T cells, Balb/c recipients were transplanted with Rag-1-/- BM alone or together with purified CD4+ T cells from WT or GM-CSF-/- mice. Mice that received CD4+ GM-CSF-/- T cells had a significant increase in survival compared to those that received WT CD4+ T cells, confirming a proinflammatory role for GM-CSF production by donor CD4+ T cells. Given that GM-CSF acts on a diverse subset of innate immune cells, we then examined which myeloid cell subsets were responsive to GM-CSF two weeks post-transplantation when donor APCs have repopulated the APC compartment. Using established markers for macrophages, neutrophils, and dendritic cells, we observed no difference in the number of donor macrophages or neutrophils between groups. However, there was a significant reduction in dendritic cells (DCs) in the colon of mice receiving CD4+ GM-CSF-/- T cells, and donor-derived DCs were virtually absent from the mesenteric lymph nodes, indicating that GM-CSF facilitates the accumulation of DCs in the GI tract and associated lymphoid tissue during GVHD. Collectively, these studies demonstrate that a CD4+ T cell-intrinsic bhlhe40/GM-CSF axis potentiates gastrointestinal inflammation during GVHD by promoting inflammatory cytokine production and DC recruitment. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1993 ◽  
Vol 82 (11) ◽  
pp. 3401-3414 ◽  
Author(s):  
A Barcena ◽  
MO Muench ◽  
AH Galy ◽  
J Cupp ◽  
MG Roncarolo ◽  
...  

It has been proposed that the CD7 molecule is the first antigen expressed on the membrane of cells committed to the T-cell lineage during human fetal T-cell ontogeny. To further identify the pre-T cell subpopulation that migrates to the thymus early in ontogeny, we analyzed the phenotypic and functional characteristics of the fetal liver populations separated on the basis of CD7 expression. Three populations expressing different levels of CD7 were observed: CD7bright, CD7dull, and CD7-. A CD7bright population depleted of mature T, B, and myeloid cells (lineage negative, lin-) and mostly composed of CD56+ CD34- natural killer cells did not mature into T cells in a fetal thymic organ culture (FTOC) assay and was devoid of myeloid progenitors in a clonal colony-forming cell assay. In contrast, the CD7-/dull CD34+ lin- populations were capable of differentiating into phenotypically mature T cells after injection into FTOC and contained early myeloid progenitors. Here we phenotypically compared the fetal liver CD7 populations with the most immature fetal thymic subset that differentiated in the FTOC assay, namely the triple negative (TN, CD3- CD4-CD8-) thymocytes. Fetal TN lin- expressed high levels of CD34 marker and were further subdivided by their expression of CD1 antigen, because CD1- TN thymocytes express higher levels of CD34 antigen compared with CD1+ TN cells. CD1- lin -TN thymocytes are characterized by expressing high levels of CD2, CD7, and CD34 markers and dull levels of CD5, CD10, and CD28 molecules. We could not find fetal liver pre-T cells with a phenotype equivalent to that of TN thymocytes. Our data show that CD7 does not necessarily identify T-cell precursors during fetal T-cell development and strongly support the hypothesis that the acquisition of early T-cell markers as CD2, CD28, and CD5 molecules on the cell surface of T-cell progenitors takes place intrathymically.


2021 ◽  
Vol 9 (10) ◽  
pp. e002758
Author(s):  
Wan-Lun Yan ◽  
Chiao-Chieh Wu ◽  
Kuan-Yin Shen ◽  
Shih-Jen Liu

BackgroundThe major challenge of antitumor immunotherapy is dealing with the immunosuppressive tumor microenvironment, which involves immature myeloid cell accumulation that results in T cell dysfunction. Myeloid cell activation is induced by Toll-like receptor agonists. Additionally, granulocyte/macrophage colony stimulating factor (GM-CSF) promotes myelopoiesis and recruits myeloid cells. Here, we combined the Toll-like receptor 2 (TLR2) agonist lipoprotein and GM-CSF to assess whether this bifunctional immunotherapy has synergistic effects on myeloid cells and could be further developed as a therapeutic intervention that enhances the antitumor response.MethodsWe investigated the synergistic effects of biadjuvanted tumor antigen on antigen-presenting cell (APC) activation in bone marrow-derived dendritic cells. Furthermore, therapeutic efficacy was monitored in different tumor models treated via intratumoral or subcutaneous administration routes. The immune effects of the bifunctional fusion protein on myeloid cells in the tumor mass and draining lymph nodes were analyzed by flow cytometry. The induction of cytotoxic T lymphocytes was evaluated via intracellular cytokine levels, perforin/granzyme B staining and an in vivo killing assay.ResultsThe TLR2 agonist lipoprotein combined with GM-CSF synergistically induced DC maturation, which subsequently enhanced antitumor immunity. In addition, rlipoE7m-MoGM modulated tumor-infiltrating myeloid cell populations. Vaccination with rlipoE7m-MoGM therapy increased the number of CCR7+CD103+ cDC1s, whereas the number of suppressive tumor-associated macrophages was reduced in the tumor lesions. Consistent with this observation, proliferating antigen-specific CD8+ T cells are highly infiltrated within the tumor, and the expression of IFN-r and perforin was most pronounced within antigen-specific CD8+ T cells in mice administered rlipoE7m-MoGM therapy. This finding corresponded with observation that the combination of a TLR2 agonist and GM-CSF provides increased antitumor activity by inhibiting established tumor outgrowth and protecting against metastatic cancer compared with a TLR2 agonist alone. Importantly, tumor growth inhibition was not due to the direct effects of the TLR2 agonist or GM-CSF but was instead due to the induction of antigen-specific immunity.ConclusionsThe combination of a TLR2 agonist and GM-CSF has synergistic effects that inhibit tumor growth and modulate tumor-infiltrating APCs. This therapeutic approach could be applied to other tumor antigens to treat different cancers.


Sign in / Sign up

Export Citation Format

Share Document