scholarly journals Fyn tyrosine kinase, a downstream target of receptor tyrosine kinases, modulates antiglioma immune responses

2020 ◽  
Vol 22 (6) ◽  
pp. 806-818 ◽  
Author(s):  
Andrea Comba ◽  
Patrick J Dunn ◽  
Anna E Argento ◽  
Padma Kadiyala ◽  
Maria Ventosa ◽  
...  

Abstract Background High-grade gliomas are aggressive and immunosuppressive brain tumors. Molecular mechanisms that regulate the inhibitory immune tumor microenvironment (TME) and glioma progression remain poorly understood. Fyn tyrosine kinase is a downstream target of the oncogenic receptor tyrosine kinase pathway and is overexpressed in human gliomas. Fyn’s role in vivo in glioma growth remains unknown. We investigated whether Fyn regulates glioma initiation, growth and invasion. Methods We evaluated the role of Fyn using genetically engineered mouse glioma models (GEMMs). We also generated Fyn knockdown stem cells to induce gliomas in immune-competent and immune-deficient mice (nonobese diabetic severe combined immunodeficient gamma mice [NSG], CD8−/−, CD4−/−). We analyzed molecular mechanism by RNA sequencing and bioinformatics analysis. Flow cytometry was used to characterize immune cellular infiltrates in the Fyn knockdown glioma TME. Results We demonstrate that Fyn knockdown in diverse immune-competent GEMMs of glioma reduced tumor progression and significantly increased survival. Gene ontology (GO) analysis of differentially expressed genes in wild-type versus Fyn knockdown gliomas showed enrichment of GOs related to immune reactivity. However, in NSG and CD8−/− and CD4−/− immune-deficient mice, Fyn knockdown gliomas failed to show differences in survival. These data suggest that the expression of Fyn in glioma cells reduces antiglioma immune activation. Examination of glioma immune infiltrates by flow cytometry displayed reduction in the amount and activity of immune suppressive myeloid derived cells in the Fyn glioma TME. Conclusions Gliomas employ Fyn mediated mechanisms to enhance immune suppression and promote tumor progression. We propose that Fyn inhibition within glioma cells could improve the efficacy of antiglioma immunotherapies.

2019 ◽  
Author(s):  
Andrea Comba ◽  
Patrick J Dunn ◽  
Anna E Argento ◽  
Padma Kadiyala ◽  
Maria Ventosa ◽  
...  

ABSTRACTBackgroundHigh grade gliomas are aggressive and immunosuppressive brain tumors. Molecular mechanisms that regulate the inhibitory immune tumor microenvironment (TME) and glioma progression remain poorly understood. FYN tyrosine kinase is a downstream target of the oncogenic receptor tyrosine kinases pathway and is overexpressed in human gliomas. FYN’s rolein vivoin glioma growth remains unknown. We investigated whether FYN regulates glioma initiation, growth and invasion.MethodsWe evaluated the role of FYN using genetically engineered mouse glioma models (GEMM). We also generated FYN knockdown stem cells to induce gliomas in immune-competent and immune-deficient mice (NSG, CD8−/−, CD4−/−). We analyzed molecular mechanism by RNA-Seq and bioinformatics analysis. Flow cytometry was used to characterize immune cellular infiltrates in the FYN knockdown glioma TME.ResultsWe demonstrate that FYN knockdown in diverse immune-competent GEMMs of glioma reduced tumor progression and significantly increased survival. Gene ontologies (GOs) analysis of differentially expressed genes in wild type vs. FYN knockdown gliomas showed enrichment of GOs related to immune reactivity. However, in NSG, CD8−/− and CD4−/− immune-deficient mice, FYN knockdown gliomas failed to show differences in survival. These data suggest that the expression of FYN in glioma cells reduces anti-glioma immune activation. Examination of glioma immune infiltrates by flow-cytometry displayed reduction in the amount and activity of immune suppressive myeloid derived cells (MDSCs) in the FYN glioma TME.ConclusionsGliomas employ FYN mediated mechanisms to enhance immune-suppression and promote tumor progression. We propose that FYN inhibition within glioma cells could improve the efficacy of anti-glioma immunotherapies.Key pointsInhibition of FYN tyrosine kinase in genetically engineered mouse glioma models delays tumor initiation and progression. The oncogenic effects of FYN in vivo are mediated by downregulation of anti-glioma immunity.Importance of the StudyFYN is an effector of receptor tyrosine kinases (RTK) signaling in glioma. However, its rolein vivoremains unknown. Our study demonstrates that FYN tyrosine kinase is a novel regulator of the anti-glioma immune response. We show that FYN inactivation suppresses glioma growth, increases survival, and enhances anti-tumor immune reactivity. Our findings suggest that suppressing the expression of FYN in glioma cells could provide a novel therapeutic target.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi261-vi261
Author(s):  
Andrea Comba ◽  
Patrick J Dunn ◽  
Anna E Argento ◽  
Padma Kadiyala ◽  
Maria Ventosa ◽  
...  

Abstract There is currently much excitement for the use of immunotherapies in cancer. In spite of positive results using checkpoint inhibitors in melanoma and CAR T cells in leukemias, these strategies have not yet achieved robust clinical responses in human gliomas. A powerful inhibitory microenvironment is thought to be the culprit. Mechanisms that determine the inhibitory microenvironment remain poorly understood. Herein we demonstrate that FYN, a downstream target of receptor tyrosine kinases signaling, inhibits the anti-glioma immune response. We utilized genetically engineered mouse models (GEMM) of glioma based on the Sleeping Beauty Transposon method. We examined the activities of FYN in NP (N-ras + shp53), NPA (NP + shATRX), and NPD (NP + PDGF overexpression) tumors. We also generated FYN knockdown glioma stem cells in vitro to induce gliomas in immune-competent and immune-deficient mice of varying genetic backgrounds (NSG, CD8-/-). Flow cytometry was used to characterize immune cells within the glioma microenvironment. Our results show that FYN knockdown in NP, NPA, or NPD GEMM of glioma reduced tumor progression and increased survival by 25–77%. GSEA analysis of differential expressed genes of WT vs. FYN knockdown gliomas revealed enrichments of gene ontologies related to immune functions. In NSG and CD8-/- immune-deficient mice, FYN knockdown failed to inhibits tumor growth and increase animal survival. These results suggest that FYN enhances tumor progression through changes in anti-glioma immune activation. Examination of tumor immune infiltrates by flow cytometry indicate a 50–70% reduction in powerful immune inhibitory myeloid derived cells (MDSCs) with no changes in the total level of CD8+ and CD4+ cells. Our results show for the first time that FYN reduces anti-glioma immune responses, likely through a reduction in inhibitory MDSCs. The specific inhibition of FYN exclusively within glioma cells, could improve the efficacy of anti-glioma immunotherapies.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi260-vi260
Author(s):  
Andrea Comba ◽  
Patrick J Dunn ◽  
Anna E Argento ◽  
Padma Kadiyala ◽  
Sebastien Motsch ◽  
...  

Abstract Mesenchymal gliomas are the most aggressive tumors that carry the worst prognosis. The origins of mesenchymal cells within brain tumors, remains poorly understood. They could originate either from invading mesenchymal cells, from perivascular smooth muscle actin+ cells, or from a mesenchymal transformation of tumor cells. Identifying the origin and function of mesenchymal cells within gliomas is essential as these cells contribute to increased glioma aggressiveness and tumor progression. In this study we used human biopsies and implantable and genetically engineered mouse models (GEMM) of GBM to study tumor mesenchymal transformation. GBM implantable models were used to analyze the molecular landscape by laser microdissection followed by RNA-Seq and bioinformatics analysis. Time lapse confocal imagining was implemented to analyze GBM cells dynamics. Our results indicate the existence of a complex intratumoral and peritumoral dynamic organization of glioma cells (i.e., Oncostreams). Multicellular structures of elongated cells compatible with mesenchymal differentiation. These structures play important roles in intratumoral movements, peritumoral invasion of normal brain, and overall glioma progression. We also show that oncostreams are molecularly distinct and display increased expression of mesenchymal genes such as Col1a1. Knocking down of Col1a1 in a GEMM of aggressive gliomas reduced tumor progression and significantly increased animal survival. Histological examination confirmed absence of Col1a1, and absence of morphologically identifiable oncostreams. Our results show that tumor cells, especially within oncostreams, display a fibroblastic-like morphology and express proteins typical of mesenchymal cells. The knockout of Col1a1 from tumoral cells eliminated oncostreams from tumors and delayed tumor progression. These data suggest that tumor cells expressing mesenchymal genes regulate the organization of mesenchymal multicellular structures, and determine glioma progression. We propose that inhibiting mesenchymal transformation of glioma cells will assist in the treatment of glioblastoma.


2007 ◽  
Vol 27 (43) ◽  
pp. 11543-11551 ◽  
Author(s):  
D. Beacham ◽  
M. Ahn ◽  
W. A. Catterall ◽  
T. Scheuer

2014 ◽  
Vol 34 (11) ◽  
pp. 1826-1836 ◽  
Author(s):  
Jun Ma ◽  
Yilong Yao ◽  
Ping Wang ◽  
Yunhui Liu ◽  
Lini Zhao ◽  
...  

Blood-tumor barrier (BTB) constitutes an efficient organization of tight junctions that impairs the delivery of therapeutic drugs. However, the methods and molecular mechanisms underlying the BTB opening remain elusive. MicroRNAs (miRNAs) have recently emerged as key regulators of various biologic processes and therapeutic targets. In this study, we have identified microRNA-181a (miR-181a) as a critical miRNA in opening BTB. MicroRNA-181a expression was upregulated in glioma endothelial cells (GECs), which were obtained by coculturing endothelial cells (ECs) with glioma cells. Overexpression of miR-181a resulted in an impaired and permeability increased BTB, and meanwhile reduced the expression of zonula occluden (ZO)-1, occludin, and claudin-5. Kruppel-like factor 6 (KLF6), a transcription factor of the zinc-finger family, was downregulated in GECs. Mechanistic investigations defined it as a direct and functional downstream target of miR-181a, which was involved in the regulation of BTB permeability and the expression of ZO-1, occludin, and claudin-5. Furthermore, luciferase assays and chromatin immunoprecipitation assays showed that KLF6 upregulated the promoter activities and interacted with the promoters of ZO-1, occludin, and claudin-5 in GECs. Collectively, we showed the possibility that overexpression of miR-181a contributes to the increased permeability of BTB by targeting KLF6, thereby revealing potential therapeutic targets for the treatment of brain gliomas.


2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 942.2-942
Author(s):  
Y. Zhang ◽  
Y. Qin ◽  
Z. Chen

Background:Reduction and dysregulation of ILC2 was linked to delayed resolution of arthritis. The neuropeptide Neuromedin U (NMU) has been reported to rapidly activate ILC2 and initiate a Th2 type immune response through NMUR1 expressed on the surface of ILC2. However, one previous study reported that NMU promoted autoantibody-mediated arthritis.Objectives:The aim of this work was to investigate the effect of NMU on collagen-induced arthritis (CIA) mice and the potential mechanisms.Methods:CIA was induced in C57BL/6 WT and C57BL/6Nmudeficient mice on day 1. WT mice were treated i.p. daily by NMU-23 (20ug/mice) or by PBS for 10 days from day 1 to 5 and day 21 to 25. The clinical scores of CIA mice were assessed every two days from day 22 and determined on a scale of 0–4 for each paw. The proportion of ILC2 as well as Th1, Th2, Th17 and Treg in spleen, mesenteric lymph node (mLN) and joints of arthritic mice were analyzed by flow cytometry on day 42.Results:NMU-23 dramatically inhibited clinical onset and severity of arthritis in treated WT mice compared with control mice. Interestingly, NMU-deficient mice also developed significantly less severe arthritis compared with WT control (Fig 1). Flow cytometry analyses showed that the proportion of ILC2, which defined as Lin-CD45+CD127+KLRG1+ICOS+ST2+, was elevated in the joint but not in the spleen and mLN of arthritic mice treated with NMU-23. In contrast, the proportion of ILC2 was significantly lower in the spleen of NMU-deficient mice than WT control. The percentage of Th2 cells in the spleen and mLN tend to be higher in NMU-23 treated mice, but there is no statistical significance. Surprisingly, Th1 cells were increased in the mLN of NMU-23 treated and NMU-deficient mice compared with control whereas Th17 was comparable among groups. In addition, the proportion of Treg was decreased in the joint of NMU-23 treated and NMU-deficient mice compared with control mice.Conclusion:Our preliminary results show that repeated injection of NMU-23 during induction (early) and development (late) stage of CIA strongly suppressed clinical onset and severity of arthritis, which might be ascribed to activation of ILC2 in the joint. Further study is needed to explore other cellular and molecular mechanisms in the effect.References:[1] Cardoso V, Chesne J, Ribeiro H et al (2017) Neuronal regulation of type 2 innate lymphoid cells via neuromedin U. Nature 549 (7671):277-281.[2] Klose CSN, Mahlakoiv T, Moeller JB et al (2017) The neuropeptide neuromedin U stimulates innate lymphoid cells and type 2 inflammation. Nature 549 (7671):282-286.[3] Wallrapp A, Riesenfeld SJ, Burkett PR et al (2017) The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation. Nature 549 (7672):351-356.[4] Sindhuja M Rao, Jennifer L Auger, Philippe Gaillard et al (2012) The Neuropeptide Neuromedin U Promotes Autoantibody-Mediated Arthritis. Arthritis Res Ther, 14 (1), R29.Disclosure of Interests:None declared


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 573-573
Author(s):  
Satu Mustjoki ◽  
Marja Ekblom ◽  
T. Petteri Arstila ◽  
Ingunn Dybedal ◽  
PK Epling- Burnette ◽  
...  

Abstract Tyrosine kinase inhibitors (TKIs) targeting the BCR-ABL fusion protein are an effective therapy for Philadelphia chromosome positive (Ph+) leukemia. Dasatinib, a 2nd generation pan-TKI, also inhibits wild-type kinases, which may result in unexpected clinical responses. Recent data suggest that dasatinib has a potent immunosuppressive effect on T- and NK-cells in vitro. In contrast, we have noticed a marked expansion of lymphocytes in blood in a subset of dasatinib patients, but its clinical significance and molecular mechanisms are unclear. In this multicenter study, 19 Ph+ leukemia patients with marked lymphoproliferation in blood while on dasatinib (9 CML CP, 2 CML AP, 2 CML BC, 6 Ph+ALL) were identified. Clonality, immunophenotype, and intracellular signaling was analyzed and related clinical information was collected. In addition, prevalence and prognostic significance of the phenomenon was retrospectively assessed in a Phase II clinical study on 46 Ph+ ALL patients. An abrupt lymphocytosis (peak count range 4–20×109/L) with large granular lymphocyte (LGL) morphology was observed after a median of 3 months from the start of therapy (range 1–8 months). In most patients, LGL lymphocytosis was long-lasting and continued throughout dasatinib therapy, albeit with marked fluctuations in absolute lymphocyte count (Fig. 1a). In all evaluable patients (n=4), lymphocyte counts normalized after drug discontinuation. All patients were previously exposed to imatinib without similar changes in lymphocyte count or morphology. By immunophenotyping, 14 patients had a cytotoxic T-cell and 5 patients a NK-cell phenotype. After initiation of dasatinib therapy, the CD4/CD8 ratio shifted, expression of activation antigens HLA-DR and CD57 increased, expression of homing antigen CD62L decreased and relative numbers of regulatory T-cells were significantly decreased (p<0.001). All patients with T-LGL phenotype had mono/oligoclonal rearrangements in TCR g/d genes. Quantitative allele-specific oligonucleotide PCR analysis from samples before and during dasatinib therapy indicated that LGL lymphocytosis results from expansion of a pre-existing terminal memory cell clone (CD8+CD62Llow). By using single-cell profiling of nodal phosphoproteins (ERK1/2, STAT1, STAT3, STAT5, STAT6) by multi-color FACS, we showed that during dasatinib therapy LGL patients were more responsive to cytokine or growth factor stimuli (IL-2, IL-4, IL-6, IFNs, GM-CSF), when compared to dasatinib patients without lymphocytosis or to healthy controls. This indicates alternative signaling pathway usage not inhibited by dasatinib and/or cytokine hypersensitivity. Dasatinib-related severe adverse effects were common in patients with LGL lymphocytosis. Pleural effusions and/or pulmonary infiltrates were seen in 13/19 patients, modest CMV reactivation in 9/19 patients, colitis in 11/19 patients and long lasting mild fever in 14/19 patients. Accumulation of pheno- and genotypically identical cytotoxic T-cells was also detected in pleural effusion and colitis biopsy samples. Adverse effects were temporally related to dasatinib therapy and emerged after the appearance of LGL lymphocytosis. All patients with relapsed poor prognosis leukemia (Ph+ ALL n=5, blastic phase CML n=2) achieved durable complete molecular responses (CMR) during dasatinib therapy, CMR still ongoing in 3 patients (follow-up 18+ months). In the confirmatory Phase II dasatinib study in Ph+ ALL (START-L), patients with lymphocytosis had superior survival compared to patients without lymphocytosis (Fig. 1b). In conclusion, we propose that by inhibiting immunoregulatory kinases, dasatinib may induce a reversible state of aberrant immune reactivity in a distinct subset of patients and result in anti-leukemia and anti-host responses driven by cytotoxic T/NK LGL cells. Further studies pinpointing the mechanisms, such as the target antigens on the malignant cells and activation pathways are ongoing. Figure Figure


2004 ◽  
Vol 48 (3) ◽  
pp. 259-267 ◽  
Author(s):  
Kayoko Hamaguchi-Hamada ◽  
Chiaki Sanbo ◽  
Shun Hamada ◽  
Takeshi Yagi

Sign in / Sign up

Export Citation Format

Share Document