scholarly journals Dasatinib, a small-molecule protein tyrosine kinase inhibitor, inhibits T-cell activation and proliferation

Blood ◽  
2008 ◽  
Vol 111 (3) ◽  
pp. 1366-1377 ◽  
Author(s):  
Andrew E. Schade ◽  
Gary L. Schieven ◽  
Robert Townsend ◽  
Anna M. Jankowska ◽  
Vojkan Susulic ◽  
...  

Abstract Dasatinib is an oral small molecule inhibitor of Abl and Src family tyrosine kinases (SFK), including p56Lck (Lck). Given the central importance of Lck in transmitting signals from the T-cell receptor (TCR) signaling complex and the potent ability of dasatinib to inhibit Lck activity, we hypothesized this agent could provide a novel route of immunomodulation via targeted inhibition of antigen-induced signaling. Herein, we show that dasatinib inhibits TCR-mediated signal transduction, cellular proliferation, cytokine production, and in vivo T-cell responses. However, dasatinib-mediated inhibition does not induce apoptosis because the effect is reversible or may be overcome by signals bypassing the TCR, such as phorbol ester. Signal transduction and proliferative responses via IL-2 remain essentially unperturbed, suggesting that dasatinib displays specificity for TCR signaling. In addition, dasatinib combined with cyclosporine A or rapamycin led to a much more potent inhibition of T-cell activation, suggesting that targeted inhibition of Lck could be a useful adjunct for enhanced immunomodulation. In combination with currently available immunomodulatory agents, SFK inhibition could potentially increase immunomodulatory efficacy while minimizing toxicity of individual agents.

1995 ◽  
Vol 310 (1) ◽  
pp. 243-248 ◽  
Author(s):  
T Dubois ◽  
J P Oudinet ◽  
F Russo-Marie ◽  
B Rothhut

In order to understand how signal transduction occurs during T cell activation, it is necessary to identify the key regulatory molecules whose function is influenced by phosphorylation. Annexins II (A-II) and V (A-V) belong to a large family of Ca(2+)-dependent phospholipid-binding proteins. Among many putative functions, annexins may be involved in signal transduction during cellular proliferation and differentiation. In the present study we show that A-II is phosphorylated in vivo in the Jurkat human T cell line. Indeed, A-II is phosphorylated after stimulation by phorbol myristate acetate and on serine residues after T cell antigen receptor (TcR) stimulation. In cytosol from Jurkat cells, A-II is phosphorylated only by Ca2+/phospholipid-stimulated kinases such as Ca(2+)-dependent protein kinases C (cPKCs). A-V inhibits the phosphorylation of A-II and other substrates of cPKCs and has no effect on kinases activated only by phospholipids. In conclusion, A-II is phosphorylated both in vitro and in vivo in Jurkat cells, and may play a role as a substrate during signal transduction in lymphocytes via the TcR through the PKC pathway. On the other hand, A-V could act as a potent modulator of cPKCs in Jurkat cells.


2004 ◽  
Vol 199 (3) ◽  
pp. 369-379 ◽  
Author(s):  
Magdalena M. Gorska ◽  
Susan J. Stafford ◽  
Osman Cen ◽  
Sanjiv Sur ◽  
Rafeul Alam

The first step in T cell receptor for antigen (TCR) signaling is the activation of the receptor-bound Src kinases, Lck and Fyn. The exact mechanism of this process is unknown. Here, we report that the novel Src homology (SH) 3/SH2 ligand–Uncoordinated 119 (Unc119) associates with CD3 and CD4, and activates Lck and Fyn. Unc119 overexpression increases Lck/Fyn activity in T cells. In Unc119-deficient T cells, Lck/Fyn activity is dramatically reduced with concomitant decrease in interleukin 2 production and cellular proliferation. Reconstitution of cells with Unc119 reverses the signaling and functional outcome. Thus, Unc119 is a receptor-associated activator of Src-type kinases. It provides a novel mechanism of signal generation in the TCR complex.


2020 ◽  
Vol 3 (1) ◽  
Author(s):  
Eliot Morrison ◽  
Tatjana Wegner ◽  
Andres Ernesto Zucchetti ◽  
Miguel Álvaro-Benito ◽  
Ashley Zheng ◽  
...  

AbstractPalmitoylation is the reversible addition of palmitate to cysteine via a thioester linkage. The reversible nature of this modification makes it a prime candidate as a mechanism for regulating signal transduction in T-cell receptor signaling. Following stimulation of the T-cell receptor we find a number of proteins are newly palmitoylated, including those involved in vesicle-mediated transport and Ras signal transduction. Among these stimulation-dependent palmitoylation targets are the v-SNARE VAMP7, important for docking of vesicular LAT during TCR signaling, and the largely undescribed palmitoyl acyltransferase DHHC18 that is expressed in two isoforms in T cells. Using our newly developed On-Plate Palmitoylation Assay (OPPA), we show DHHC18 is capable of palmitoylating VAMP7 at Cys183. Cellular imaging shows that the palmitoylation-deficient protein fails to be retained at the Golgi and to localize to the immune synapse upon T cell activation.


2020 ◽  
Author(s):  
Hridesh Banerjee ◽  
Hector Nieves-Rosado ◽  
Aditi Kulkarni ◽  
Benjamin Murter ◽  
Uma R. Chandran ◽  
...  

AbstractRegulatory T cells (Treg) are critical mediators of self-tolerance but can also limit effective anti-tumor immunity. We and others previously reported that 40-60% percent of Treg-infiltrating head and neck cancer (HNC) and other tumors highly express Tim-3, compared with about 5% in lymphoid organs. Tumor-infiltrating Tim-3+ Treg also have enhanced suppressive function and display a more effector-like phenotype. Using a novel mouse model with cell type-specific Tim-3 expression, we show here that expression of Tim-3 by Treg is sufficient to drive Treg to a more effector-like phenotype, resulting in enhanced suppressive activity and increased tumor growth. These findings may help to reconcile previous reports that some Tim-3 antibodies enhance T cell responses in vivo, while expression of Tim-3 has a cell-intrinsic ability to enhance TCR signaling and T cell activation. Thus, we propose that Tim-3 regulates anti-tumor immunity at least in part through enhancement of Treg function. To our knowledge, this is the first example in which expression of a single co-stimulatory molecule is sufficient to drive differentiation of Treg in this manner.


2019 ◽  
Vol 2 (1) ◽  
pp. e201800282 ◽  
Author(s):  
Takayuki Imanishi ◽  
Midori Unno ◽  
Wakana Kobayashi ◽  
Natsumi Yoneda ◽  
Satoshi Matsuda ◽  
...  

Stimulator of interferon genes (STING) plays a key role in detecting cytosolic DNA and induces type I interferon (IFN-I) responses for host defense against pathogens. Although T cells highly express STING, its physiological role remains unknown. Here, we show that costimulation of T cells with the STING ligand cGAMP and TCR leads to IFN-I production and strongly inhibits T-cell growth. TCR-mediated mTORC1 activation and sustained activation of IRF3 are required for cGAMP-induced IFN-I production, and the mTORC1 activity is partially counteracted by cGAMP, thereby blocking proliferation. This mTORC1 inhibition in response to costimulation depends on IRF3 and IRF7. Effector T cells produce much higher IFN-I levels than innate cells in response to cGAMP. Finally, we demonstrated that STING stimulation in T cells is effective in inducing antitumor responses in vivo. Our studies demonstrate that the outputs of STING and TCR signaling pathways are mutually regulated through mTORC1 to modulate T-cell functions.


2003 ◽  
Vol 197 (8) ◽  
pp. 955-966 ◽  
Author(s):  
Fred Lühder ◽  
Yun Huang ◽  
Kevin M. Dennehy ◽  
Christine Guntermann ◽  
Ingrid Müller ◽  
...  

Full activation of naive T cells requires both engagement of the T cell antigen receptor (TCR; signal 1) and costimulatory signaling by CD28 (signal 2). We previously identified two types of rat CD28-specific monoclonal antibodies (mAbs): “conventional,” TCR signaling–dependent costimulatory mAbs and “superagonistic” mAbs capable of inducing the full activation of primary resting T cells in the absence of TCR ligation both in vitro and in vivo. Using chimeric rat/mouse CD28 molecules, we show that the superagonists bind exclusively to the laterally exposed C′′D loop of the immunoglobulin-like domain of CD28 whereas conventional, costimulatory mAbs recognize an epitope close to the binding site for the natural CD80/CD86 ligands. Unexpectedly, the C′′D loop reactivity of a panel of new antibodies raised against human CD28 could be predicted solely on the basis of their superagonistic properties. Moreover, mouse CD28 molecules engineered to express the rat or human C′′D loop sequences activated T cell hybridomas without TCR ligation when cross-linked by superagonistic mAbs. Finally, biochemical analysis revealed that superagonistic CD28 signaling activates the nuclear factor κB pathway without inducing phosphorylation of either TCRζ or ZAP70. Our findings indicate that the topologically constrained interactions of anti-CD28 superagonists bypass the requirement for signal 1 in T cell activation. Antibodies with this property may prove useful for the development of T cell stimulatory drugs.


Blood ◽  
2009 ◽  
Vol 113 (23) ◽  
pp. 5793-5800 ◽  
Author(s):  
Manoj Saini ◽  
Claire Pearson ◽  
Benedict Seddon

Abstract Interleukin-7 (IL-7) plays a central role in the homeostasis of the T-cell compartment by regulating T-cell survival and proliferation. Whether IL-7 can influence T-cell receptor (TCR) signaling in T cells remains controversial. Here, using IL-7–deficient hosts and TCR-transgenic T cells that conditionally express IL-7R, we examined antigen-specific T-cell responses in vitro and in vivo to viral infection and lymphopenia to determine whether IL-7 signaling influences TCR-triggered cell division events. In vitro, we could find no evidence that IL-7 signaling could costimulate T-cell activation over a broad range of conditions, suggesting that IL-7 does not directly tune TCR signaling. In vivo, however, we found an acute requirement for IL-7 signaling for efficiently triggering T-cell responses to influenza A virus challenge. Furthermore, we found that IL-7 was required for the enhanced homeostatic TCR signaling that drives lymphopenia-induced proliferation by a mechanism involving efficient contacts of T cells with dendritic cells. Consistent with this, saturating antigen-presenting capacity in vivo overcame the triggering defect in response to cognate peptide. Thus, we demonstrate a novel role for IL-7 in regulating T cell–dendritic cell interactions that is essential for both T-cell homeostasis and activation in vivo.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1349-1349
Author(s):  
Andrew E. Schade ◽  
Anna M. Jankowska ◽  
Hadrian Szpurka ◽  
Jaroslaw P. Maciejewski

Abstract It is well-recognized that the T cell receptor (TCR) signaling pathway, catalyzed by the Src family kinase (SFK) Lck, is the essential first step in T cell immune responses. The long-held goal of specifically targeting SFK activity for immunomodulatory therapy is becoming more of a reality as SFK inhibitors are entering into clinical use. In particular, dasatinib is an oral small molecule inhibitor of Abl and SFK, including Lck. Given the central importance of Lck in transmitting signals from the TCR signaling complex, and the potent ability of dasatinib to inhibit Lck activity, we hypothesized this agent could provide a novel route of immunomodulation via targeted inhibition of antigen-induced signaling, particularly in combination with currently available immunomodulatory agents. Using intracellular phospho-flow cytometry and western blotting, we show that low nanomolar concentrations of dasatinib potently inhibit TCR induced global protein tyrosine phosphoryation, including the key signaling elements CD3, ZAP-70, LAT, and PLC. In addition, the second messenger pathway Ras/Raf/MEK/ERK is potently blocked, whereas the PI3K-AKT pathway is minimally affected. Furthermore, induction of many pro-inflammatory cytokines, including TNF-α, IFN-γ, IL-2, GM-CSF, IL-17, IP-10, RANTES, and IL-6 (measured by cytokine antibody array) was significantly inhibited when the TCR was triggered in the presence of dasatinib. T cell proliferation, assessed by flow cytometric CFSE dilution analysis, was inhibited in a dose-dependent manner with dasatinib, showing complete inhibtion at 10 nM. Cell cycle analysis based on DNA content by PI staining revealed that dasatinib-treated T cells failed to enter the cell cycle. Considering that the PI3K-AKT pathway was only minimally affected by dasatinib, we examined the ability to achieve enhanced inhibition by using sub-optimal doses of dasatinib and rapamycin that individually produce mild-moderate inhibition of proliferation (20% and 40% inhibition, respectively). When dasatinib and rapamycin are combined at these lower doses, there was a consistently enhanced inhibitory effect on T cell proliferation (80–95% inhibition). Concurrent CD28 stimulation failed to overcome the inhibitory effects of SFK inhibition when the inhibitor was present throughout the stimulation. Interestingly, when SFK inhibition was delayed for at least 24 hours after the initiation of T cell activation with concurrent CD28 stimulation, we observed a divergent effect with regard to cytokine production and proliferation. Pro-inflammatory cytokine production was significantly decreased even if SFK activity was inhibited 48 hours after stimulating T cells, suggesting that ongoing TCR-dependent SFK activity is essential for cytokine production. However, late inhibition of SFK activity with dasatinib increased proliferation, with greater numbers of T cells achieving more rounds of cell division. Thus, while necessary for cytokine production, once the cells have entered the cell cycle, SFK activity regulates negative feedback in CD28-mediated proliferation. In conclusion, targeted inhibition of SFK activity is a promising approach for novel immunomodulatory therapy, particularly in combination with other signaling pathway inhibitors, but timing of SFK inhibition in relation to T cell activation could have an important impact on the immunomodulatory effect.


1999 ◽  
Vol 189 (1) ◽  
pp. 111-121 ◽  
Author(s):  
Jean-François Cloutier ◽  
André Veillette

Antigen receptor–triggered T-cell activation is mediated by the sequential action of the Src and Syk/Zap-70 families of protein tyrosine kinases (PTKs). Previously, we reported that another PTK termed p50csk was a potent negative regulator of T-cell receptor (TCR) signaling because of its ability to inactivate Src-related kinases. This inhibitory effect required the catalytic activity of Csk, as well as its Src homology (SH)3 and SH2 domains. Subsequent studies uncovered that, via its SH3 domain, p50csk was associated with PEP, a proline-enriched protein tyrosine phosphatase (PTP) of unknown function expressed in hemopoietic cells. Herein, we have attempted to identify the role of the Csk-PEP complex in T lymphocytes. The results of our experiments showed that, like Csk, PEP was a strong repressor of TCR signaling. This property was dependent on the phosphatase activity of PEP, as well as on the sequence mediating its binding to p50csk. Through reconstitution experiments in Cos-1 cells, evidence was obtained that Csk and PEP act synergistically to inhibit protein tyrosine phosphorylation by Src-related kinases, and that this effect requires their association. Finally, experiments with a substrate-trapping mutant of PEP suggested that PEP functions by dephosphorylating and inactivating the PTKs responsible for T-cell activation. In addition to giving novel insights into the mechanisms involved in the negative regulation of T-cell activation, these findings indicate that the association of an inhibitory PTK with a PTP constitutes a more efficient means of inhibiting signal transduction by Src family kinases in vivo.


Cells ◽  
2021 ◽  
Vol 11 (1) ◽  
pp. 19
Author(s):  
Jusong Kim ◽  
Haeyeon Jang ◽  
Gyu Jin Lee ◽  
Yelim Hur ◽  
Juhee Keum ◽  
...  

In this study, we describe a novel kinase inhibitor AX-0085 which can suppress the induction of PD-L1 expression by Interferon-γ (IFN-γ) in lung adenocarcinoma (LUAD) cells. AX-0085 effectively blocks JAK2/STAT1 signaling initiated by IFN-γ treatment and prevents nuclear localization of STAT1. Importantly, we demonstrate that AX-0085 reverses the IFN-γ-mediated repression of T cell activation in vitro and enhances the anti-tumor activity of anti-PD-1 antibody in vivo when used in combination. Finally, transcriptomic analyses indicated that AX-0085 is highly specific in targeting the IFN-γ-pathway, thereby raising the possibility of applying this reagent in combination therapy with checkpoint inhibitor antibodies. It may be particularly relevant in cases in which PD-L1-mediated T cell exhaustion leads to immunoevasive phenotypes.


Sign in / Sign up

Export Citation Format

Share Document