scholarly journals PReS-FINAL-2069: T cells secreting granulocyte-macrophage colony stimulating factor (GM-CSF) within the inflammed joint originate from an "EX-Th17" population

2013 ◽  
Vol 11 (S2) ◽  
Author(s):  
K Nistala ◽  
C Piper ◽  
A Pesenacker ◽  
D Bending ◽  
B Thirugnanabalan ◽  
...  
1990 ◽  
Vol 10 (3) ◽  
pp. 1281-1286 ◽  
Author(s):  
R Schreck ◽  
P A Baeuerle

The expression of the gene encoding the granulocyte-macrophage colony-stimulating factor (GM-CSF) is induced upon activation of T cells with phytohemagglutinin and active phorbolester and upon expression of tax1, a transactivating protein of the human T-cell leukemia virus type I. The same agents induce transcription from the interleukin-2 receptor alpha-chain and interleukin-2 genes, depending on promoter elements that bind the inducible transcription factor NF-kappa B (or an NF-kappa B-like factor). We therefore tested the possibility that the GM-CSF gene is also regulated by a cognate motif for the NF-kappa B transcription factor. A recent functional analysis by Miyatake et al. (S. Miyatake, M. Seiki, M. Yoshida, and K. Arai, Mol. Cell. Biol. 8:5581-5587, 1988) described a short promoter region in the GM-CSF gene that conferred strong inducibility by T-cell-activating signals and tax1, but no NF-kappa B-binding motifs were identified. Using electrophoretic mobility shift assays, we showed binding of purified human NF-kappa B and of the NF-kappa B activated in Jurkat T cells to an oligonucleotide comprising the GM-CSF promoter element responsible for mediating responsiveness to T-cell-activating signals and tax1. As shown by a methylation interference analysis and oligonucleotide competition experiments, purified NF-kappa B binds at positions -82 to -91 (GGGAACTACC) of the GM-CSF promoter sequence with an affinity similar to that with which it binds to the biologically functional kappa B motif in the beta interferon promoter (GGGAAATTCC). Two kappa B-like motifs at positions -98 to -108 of the GM-CSF promoter were also recognized but with much lower affinities. Our data provide strong evidence that the expression of the GM-CSF gene following T-cell activation is controlled by binding of the NF-kappa B transcription factor to a high-affinity binding site in the GM-CSF promoter.


Blood ◽  
1987 ◽  
Vol 69 (4) ◽  
pp. 1259-1261
Author(s):  
J Horiguchi ◽  
MK Warren ◽  
D Kufe

The macrophage-specific colony-stimulating factor (CSF-1, M-CSF) regulates the survival, growth and differentiation of monocytes. We have recently demonstrated that phorbol ester induces expression of CSF- 1 in human monocytes. These findings suggested that activated monocytes are capable of producing their own lineage-specific CSF. The present studies demonstrate that the granulocyte-macrophage colony-stimulating factor (GM-CSF) also induces CSF-1 transcripts in monocytes. Furthermore, we demonstrate that the detection of CSF-1 RNA in GM-CSF- treated monocytes is associated with synthesis of the CSF-1 gene product. The results thus suggest that GM-CSF may indirectly control specific monocyte functions through the regulation of CSF-1 production. These findings indicate another level of interaction between T cells and monocytes.


2003 ◽  
Vol 60 (5) ◽  
pp. 531-538 ◽  
Author(s):  
Miodrag Colic ◽  
Dusan Jandric ◽  
Zorica Stojic-Vukanic ◽  
Jelena Antic-Stankovic ◽  
Petar Popovic ◽  
...  

Several laboratories have developed culture systems that allow the generation of large numbers of human dendritic cells (DC) from monocytes using granulocyte-macrophage colony stimulating factor (GM-CSF), and interleukin-4 (IL-4). In this work we provided evidence that GM-CSF (100 ng/ml) in combination with a low concentration of IL-4 (5 ng/ml) was efficient in the generation of immature, non-adherent, monocyte-derived DC as the same concentration of GM-CSF, and ten times higher concentration of IL-4 (50 ng/ml). This conclusion was based on the similar phenotype profile of DC such as the expression of CD1a, CD80, CD86, and HLA-DR, down-regulation of CD14, and the absence of CD83, as well as on their similar allostimulatory activity for T cells. A higher number of cells remained adherent in cultures with lower concentrations of IL-4 than in cultures with higher concentrations of the cytokine. However, most of these adherent cells down-regulated CD14 and stimulated the proliferation of alloreactive T cells. In contrast adherent cells cultivated with GM-CSF alone were predominantly macrophages as judged by the expression of CD14 and the inefficiency to stimulate alloreactive T cells. DC generated in the presence of lower concentrations of IL-4 had higher proapoptotic potential for the Jurkat cell line than DC differentiated with higher concentrations of IL-4, suggesting their stronger cytotoxic, anti-tumor effect.


1998 ◽  
Vol 188 (1) ◽  
pp. 133-143 ◽  
Author(s):  
Chiara Zilocchi ◽  
Antonella Stoppacciaro ◽  
Claudia Chiodoni ◽  
Mariella Parenza ◽  
Nadia Terrazzini ◽  
...  

We analyzed the ability of interferon (IFN)-γ knockout mice (GKO) to reject a colon carcinoma transduced with interleukin (IL)-12 genes (C26/IL-12). Although the absence of IFN-γ impaired the early response and reduced the time to tumor onset in GKO mice, the overall tumor take rate was similar to that of BALB/c mice. In GKO mice, C26/IL-12 tumors had a reduced number of infiltrating leukocytes, especially CD8 and natural killer cells. Analysis of the tumor site, draining nodes, and spleens of GKO mice revealed reduced expression of IFN- inducible protein 10 and monokine induced by γ-IFN. Despite these defects, GKO mice that rejected C26/IL-12 tumor, and mice that were primed in vivo with irradiated C26/IL-12 cells, showed the same cytotoxic T lymphocyte activity but higher production of granulocyte/macrophage colony–stimulating factor (GM-CSF) as compared with control BALB/c mice. Treatment with monoclonal antibodies against GM-CSF abrogated tumor regression in GKO but not in BALB/c mice. CD4 T lymphocytes, which proved unnecessary or suppressive during rejection of C26/IL-12 cells in BALB/c mice, were required for tumor rejection in GKO mice. CD4 T cell depletion was coupled with a decline in GM-CSF expression by lymphocytes infiltrating the tumors or in the draining nodes, and with the reduction and disappearance of granulocytes and CD8 T cells, respectively, in tumor nodules. These results suggest that GM-CSF can substitute for IFN-γ in maintaining the CD8–polymorphonuclear leukocyte cross-talk that is a hallmark of tumor rejection.


Blood ◽  
1994 ◽  
Vol 83 (3) ◽  
pp. 713-723
Author(s):  
AM Stewart-Akers ◽  
JS Cairns ◽  
DJ Tweardy ◽  
SA McCarthy

The effects of granulocyte-macrophage colony-stimulating factor (GM- CSF) are not confined to cells of the myeloid lineage. GM-CSF has been shown to have effects on mature T cells and both mature and immature T- cell lines. We therefore examined the GM-CSF responsiveness of murine thymocytes to investigate whether GM-CSF also affected normal immature T lymphocytes. The studies presented here indicate that GM-CSF augments accessory cell (AC)-dependent T-cell receptor (TCR)-mediated proliferation of unseparated thymocyte populations. To identify the GM- CSF responsive cell type, thymic AC and T cells were examined for GM- CSF responsiveness. We found that GM-CSF augmentation of TCR-induced thymocyte proliferation appears to be mediated via augmentation of AC function, and not via direct effects on mature single-positive (SP) thymocytes. Enriched double-negative (DN) thymocytes were also tested for GM-CSF responsiveness. GM-CSF induced the proliferation of adult and fetal DN thymocytes in an AC-independent and TCR-independent single- cell assay. Thus, in contrast to the SP thymocytes, a DN thymocyte population was directly responsive to GM-CSF. GM-CSF therefore may play a direct role in the expansion of DN thymocytes and an indirect role in the expansion of SP thymocytes.


Blood ◽  
1987 ◽  
Vol 69 (4) ◽  
pp. 1259-1261 ◽  
Author(s):  
J Horiguchi ◽  
MK Warren ◽  
D Kufe

Abstract The macrophage-specific colony-stimulating factor (CSF-1, M-CSF) regulates the survival, growth and differentiation of monocytes. We have recently demonstrated that phorbol ester induces expression of CSF- 1 in human monocytes. These findings suggested that activated monocytes are capable of producing their own lineage-specific CSF. The present studies demonstrate that the granulocyte-macrophage colony-stimulating factor (GM-CSF) also induces CSF-1 transcripts in monocytes. Furthermore, we demonstrate that the detection of CSF-1 RNA in GM-CSF- treated monocytes is associated with synthesis of the CSF-1 gene product. The results thus suggest that GM-CSF may indirectly control specific monocyte functions through the regulation of CSF-1 production. These findings indicate another level of interaction between T cells and monocytes.


1993 ◽  
Vol 13 (12) ◽  
pp. 7399-7407
Author(s):  
E S Masuda ◽  
H Tokumitsu ◽  
A Tsuboi ◽  
J Shlomai ◽  
P Hung ◽  
...  

Expression of the granulocyte-macrophage colony-stimulating factor (GM-CSF) gene in T cells is activated by the combination of phorbol ester (phorbol myristate acetate) and calcium ionophore (A23187), which mimic antigen stimulation through the T-cell receptor. We have previously shown that a fragment containing bp -95 to +27 of the mouse GM-CSF promoter can confer inducibility to reporter genes in the human Jurkat T-cell line. Here we use an in vitro transcription system to demonstrate that a cis-acting element (positions -54 to -40), referred to as CLE0, is a target for the induction signals. We observed induction with templates containing intact CLE0 but not with templates with deleted or mutated CLE0. We also observed that two distinct signals were required for the stimulation through CLE0, since only extracts from cells treated with both phorbol myristate acetate and A23187 supported optimal induction. Stimulation probably was mediated by CLE0-binding proteins because depletion of these proteins specifically reduced GM-CSF transcription. One of the binding factors possessed biochemical and immunological features identical to those of the transcription factor AP1. Another factor resembled the T-cell-specific factor NFAT. The characteristics of these two factors are consistent with their involvement in GM-CSF induction. The presence of CLE0-like elements in the promoters of interleukin-3 (IL-3), IL-4, IL-5, GM-CSF, and NFAT sites in the IL-2 promoter suggests that the factors we detected, or related factors that recognize these sites, may account for the coordinate induction of these genes during T-cell activation.


2020 ◽  
Vol 7 (4) ◽  
pp. e729 ◽  
Author(s):  
Farinaz Safavi ◽  
Rodolfo Thome ◽  
Zichen Li ◽  
Guang-Xian Zhang ◽  
Abdolmohamad Rostami

ObjectiveTo study the immunomodulatory effect of dimethyl fumarate (DF) on granulocyte macrophage colony-stimulating factor (GM-CSF) production in CD4+ T cells in experimental autoimmune encephalomyelitis (EAE) and human peripheral blood mononuclear cells (PBMCs).MethodsWe collected splenocytes and CD4+ T cells from C57BL/6 wild-type and interferon (IFN)-γ–deficient mice. For human PBMCs, venous blood was collected from healthy donors, and PBMCs were collected using the Percoll gradient method. Cells were cultured with anti-CD3/28 in the presence/absence of DF for 3 to 5 days. Cells were stained and analyzed by flow cytometry. Cytokines were measured by ELISA in cell supernatants. For in vivo experiments, EAE was induced by myelin oligodendrocyte glycoprotein35–55 and mice were treated with oral DF or vehicle daily.ResultsDF acts directly on CD4+ T cells and suppresses GM-CSF–producing Th1 not Th17 or single GM-CSF+ T cells in EAE. In addition, GM-CSF suppression depends on the IFN-γ pathway. We also show that DF specifically suppresses Th1 and GM-CSF–producing Th1 cells in PBMCs from healthy donors.ConclusionsWe suggest that DF exclusively suppresses GM-CSF–producing Th1 cells in both animal and human CD4+ T cells through an IFN-γ–dependent pathway. These findings indicate that DF has a better therapeutic effect on patients with Th1-dominant immunophenotype. However, future longitudinal study to validate this finding in MS is needed.


Author(s):  
Н.Д. Газатова ◽  
В.В. Малащенко ◽  
М.Е. Меняйло ◽  
О.Б. Мелащенко ◽  
Е.М. Морозова ◽  
...  

Актуальность. Исследовали прямые эффекты гранулоцит-макрофагального колониестимулирующего фактора (GM-CSF) человека на функциональную активность субпопуляций T-лимфоцитов. Методы. CD3+ Т-лимфоциты были выделены из крови здоровых доноров методом позитивной магнитной сепарации. T-клетки активировали частицами, конъюгированными с антителами (АТ) к молекулам CD3, СD28 и СD2 человека. Мембранную экспрессию CD3, CD4, СD45RA, СD197, CD25 и CD38 оценивали методом проточной цитофлюорометрии. Содержание интерферона- Результаты. Установлено, что GM-CSF в диапазоне концентраций 0,01-10,0 нг/мл не оказывал существенного влияния на содержание CD25+ клеток, среди активированных Т-лимфоцитов. Вместе с тем, GM-CSF в концентрации 0,1 и 1,0 нг/мл обладал способностью заметно увеличивать содержание CD38+ клеток среди наивных СD45RA+/СD197+ Т-клеток, а также среди СD45RA-/СD197+ Т-клеток центральной памяти, не оказывая при этом существенного влияния на экспрессию CD38, выявляемую среди эффекторных СD45RA-/СD197- и терминально дифференцированных СD45RA+/СD197- эффекторных Т-клеток. В относительно низкой концентрации (0,01 нг/мл) GM-CSF заметно снижал Т-клеточную продукцию INF-γ, тогда как в высокой концентрации (10,0 нг/мл) усиливал продукцию IL-2 и IL-4, снижая при этом выработку IL-10. Заключение. Полученные данные предполагают, что GM-CSF способен поддерживать активацию относительно низкодифференцированных Т-клеток, не оказывая при этом значимого влияния на активацию высоко дифференцированных Т-клеток. Background. We investigated direct effects of granulocyte-macrophage colony-stimulating factor (GM-CSF) on the functionality of T-lymphocyte subsets. Methods. CD3 + T cells were isolated from the blood of healthy donors by positive magnetic separation. The isolated T cells were activated with particles conjugated with antibodies (Abs) to human CD3, CD28, and CD2 molecules. The membrane expression of CD3, CD4, СD45RA, СD197, CD25, and CD38 was evaluated by flow cytofluorometry. Contents of interferon-γ (IFN- γ), interleukin-2 (IL-2), IL-4, and IL-10 in culture supernatants were determined by the enzyme immunoassay. Results. GM-CSF at 0.01-10.0 ng/ml had no significant effect on the content of CD25+ cells among activated T lymphocytes. At the same time, GM-CSF at 0.1-1.0 ng/ml was able to noticeably increase the content of CD38+ cells among both naive CD45RA+/CD197+ T cells and central memory CD45RA-/CD197+ T cells without affecting the СD38 expression on effector CD45RA- /CD197- and terminally differentiated CD45RA +/ CD197- effector T cells. At a relatively low concentration (0.01 ng/ml), GM-CSF significantly decreased T-cell production of INF-γ whereas at a high concentration (10.0 ng/ml), GM-CSF detectably enhanced the secretion of IL-2 and IL-4 while lowering IL-10 production. Conclusion. The findings suggest that GM-CSF is capable of supporting the activation of relatively low-differentiated T cells without significantly affecting the activation of highly differentiated T cells.


Blood ◽  
2000 ◽  
Vol 95 (10) ◽  
pp. 3011-3019
Author(s):  
Ivan Borrello ◽  
Eduardo M. Sotomayor ◽  
Frédérique-Marie Rattis ◽  
Sara K. Cooke ◽  
Lingping Gu ◽  
...  

For many cancers, autologous bone marrow transplantation (BMT) achieves a minimal residual disease state, yet relapse rates remain high. Using a syngeneic murine bone marrow transplant model, we demonstrate that vaccination with irradiated granulocyte-macrophage colony-stimulating factor (GM-CSF)–producing autologous tumor cells is effective in the post-BMT period and actually results in a greater tumor-free survival than vaccination in the nontransplant setting. Employing T cells specific for a model tumor-antigen, we find that transplantation of the tumor-bearing host results in a massive expansion and activation of tumor-specific T cells in the early posttransplant period, but this response rapidly declines in association with tumor progression. Immunization with irradiated GM-CSF tumor cells during the period of immune reconstitution results in the sustained amplification and activation of this response that closely correlates with freedom from relapse. These results demonstrate the feasibility of integrating GM-CSF vaccines in the postautologous BMT setting and suggest mechanisms that may contribute to the observed efficacy of immunization during the critical period of immune reconstitution.


Sign in / Sign up

Export Citation Format

Share Document