Increased Expression of Cytoskeleton Coordinator Protein MACF1 at the Immune Synapse during Jurkat T Cell Activation

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 28-29
Author(s):  
Deepak Salem ◽  
Arpita Upadhyaya

Introduction: Activation of the T cell Receptor (TCR) by antigen on the surface of an Antigen Presenting Cell (APC) leads to a dramatic change in T cell morphology and formation of a specialized signaling interface with the APC known as the Immune Synapse (IS). The IS allows for controlled cytokine secretion and is an important regulator of T cell activation. IS formation has been found to be dysfunctional in many immune disorders including T cell acute lymphoblastic leukemia (ALL).The actin and microtubule (MT) cytoskeletons have individually been implicated in regulating both IS formation and TCR signaling, but the crosstalk between actin and MTs in T cells has not been well explored. We hypothesized that MACF1 (Microtubule Actin Crosslinking Factor 1), a giant cytoskeleton crosslinker from the spectraplakin family, is present at the IS following Jurkat T cell activation and coordinates actin and MTs. To investigate this, we used TIRFM (Total Internal Reflection Fluorescence Microscopy) and confocal microscopy to image MACF1 organization at the IS. Methods: Jurkat E6-1 cells were cultured in RPMI 1640 supplemented with 10% FBS and 1% penicillin/streptomycin. Glass coverslips were coated with Poly-L-Lysine 0.01% and then with 10 µg/mL anti-CD3 for 2 hours at 37 °C to create an activating surface. Cells were dropped carefully and allowed to spread on these coverslips at 37 °C for the indicated time. In the inhibition experiments,5 µM SB415286 or 5 µM PP2 was added to cells at 8 minutes following the initiation of activation and allowed to incubate for an additional 8 minutes. Cells were fixed with 3.7% paraformaldehyde following activation and then permeabilized with 0.1% Triton X-100. Cells were stained for anti-MACF1, anti-beta tubulin clone KMX-1, respective secondary antibodies, actin-stain 488 phalloidin, and DAPI (4',6-diamidino-2-phenylindole). Fluorescence of labeled cells was imaged using TIRFM with a Nikon Ti microscope and Andor Zyla CMOS camera with a 100x magnification objective (1.49 NA). TIRFM images were analyzed for Corrected Total Cell Fluorescence, which is the total fluorescence of the cell corrected for the background. Mann-Whitney U tests were used for all statistical comparisons. Cells were imaged on Leica SP5X laser confocal microscope with 63x objective for localization of MACF1 with actin and tubulin. Confocal images were deconvolved using the deconvolution lab2 plugin in FIJI (FIJI Is Just ImageJ). Results: Corrected Total Cell Fluorescence (CTCF) of MACF1 was higher at 4, 8 and 16 minutes following activation compared to the PLL control (Figures 1A and 1B). There was no significant difference in CTCF across the different activation times. To test whether MACF1 is dis-inhibited in Jurkat T cells during activation, we used SB415286 to inhibit Glycogen Synthase Kinase 3-Beta (GSK3B), a protein that inhibits MACF1 function, on PLL-only coverslips. We found MACF1 CTCF to be higher following SB415286 treatment compared to the control, indicating that MACF1 levels increase with GSK3B inhibition (Figure 1C). Thus, SB415286 inhibition of GSK3B mimics T cell activation where Akt upregulation downstream of TCR signaling inhibits GSK3B. To further test whether CD3 signaling activates MACF1, we stimulated Jurkat cells on anti-CD3 coated glass and then removed the activation signal with the src-kinase inhibitor PP2, which inhibits lymphocyte-specific protein tyrosine kinase (Lck) - an important regulator of early T cell signaling. MACF1 CTCF was lower after PP2 inhibition compared with the control, suggesting that continued CD3 stimulation is necessary for MACF1 accumulation at the IS (Figure 1D). We then used confocal microscopy to image the spatial distribution of MACF1 and assess whether it localizes with actin and MTs at the IS. We found that MACF1 puncta are present at the F-actin rich lamellipodial region of the IS and appear to localize with MT filaments, suggesting that MACF1 coordinates the two cytoskeleton components at the IS (Figure 2). Conclusion: Using high resolution microscopy, we show that MACF1 is upregulated at the IS during Jurkat T cell activation downstream of CD3 signaling through a GSK3B dependent pathway. At the IS, we found the novel localization of MACF1 with actin and MTs. This upregulation and expression of MACF1 during activation warrants more studies of the role MACF1 plays in T cell activation, cytoskeletal dynamics and IS function. Disclosures No relevant conflicts of interest to declare.

2006 ◽  
Vol 26 (14) ◽  
pp. 5497-5508 ◽  
Author(s):  
Kazuhiro Ishiguro ◽  
Todd Green ◽  
Joseph Rapley ◽  
Heather Wachtel ◽  
Cosmas Giallourakis ◽  
...  

ABSTRACT CARMA1 is a central regulator of NF-κB activation in lymphocytes. CARMA1 and Bcl10 functionally interact and control NF-κB signaling downstream of the T-cell receptor (TCR). Computational analysis of expression neighborhoods of CARMA1-Bcl10MALT 1 for enrichment in kinases identified calmodulin-dependent protein kinase II (CaMKII) as an important component of this pathway. Here we report that Ca2+/CaMKII is redistributed to the immune synapse following T-cell activation and that CaMKII is critical for NF-κB activation induced by TCR stimulation. Furthermore, CaMKII enhances CARMA1-induced NF-κB activation. Moreover, we have shown that CaMKII phosphorylates CARMA1 on Ser109 and that the phosphorylation facilitates the interaction between CARMA1 and Bcl10. These results provide a novel function for CaMKII in TCR signaling and CARMA1-induced NF-κB activation.


Blood ◽  
2009 ◽  
Vol 114 (3) ◽  
pp. 580-588 ◽  
Author(s):  
Kathrin Gollmer ◽  
François Asperti-Boursin ◽  
Yoshihiko Tanaka ◽  
Klaus Okkenhaug ◽  
Bart Vanhaesebroeck ◽  
...  

Abstract CD4+ T cells use the chemokine receptor CCR7 to home to and migrate within lymphoid tissue, where T-cell activation takes place. Using primary T-cell receptor (TCR)–transgenic (tg) CD4+ T cells, we explored the effect of CCR7 ligands, in particular CCL21, on T-cell activation. We found that the presence of CCL21 during early time points strongly increased in vitro T-cell proliferation after TCR stimulation, correlating with increased expression of early activation markers. CCL21 costimulation resulted in increased Ras- and Rac-GTP formation and enhanced phosphorylation of Akt, MEK, and ERK but not p38 or JNK. Kinase-dead PI3KδD910A/D910A or PI3Kγ-deficient TCR-tg CD4+ T cells showed similar responsiveness to CCL21 costimulation as control CD4+ T cells. Conversely, deficiency in the Rac guanine exchange factor DOCK2 significantly impaired CCL21-mediated costimulation in TCR-tg CD4+ T cells, concomitant with impaired Rac- but not Ras-GTP formation. Using lymph node slices for live monitoring of T-cell behavior and activation, we found that G protein-coupled receptor signaling was required for early CD69 expression but not for Ca2+ signaling. Our data suggest that the presence of CCL21 during early TCR signaling lowers the activation threshold through Ras- and Rac-dependent pathways leading to increased ERK phosphorylation.


2021 ◽  
Vol 14 (687) ◽  
pp. eaba0717
Author(s):  
Shunsuke Kataoka ◽  
Priyanka Manandhar ◽  
Judong Lee ◽  
Creg J. Workman ◽  
Hridesh Banerjee ◽  
...  

Expression of the transmembrane protein Tim-3 is increased on dysregulated T cells undergoing chronic activation, including during chronic infection and in solid tumors. Thus, Tim-3 is generally thought of as an inhibitory protein. We and others previously reported that under some circumstances, Tim-3 exerts paradoxical costimulatory activity in T cells (and other cells), including enhancement of the phosphorylation of ribosomal S6 protein. Here, we examined the upstream signaling pathways that control Tim-3–mediated increases in phosphorylated S6 in T cells. We also defined the localization of Tim-3 relative to the T cell immune synapse and its effects on downstream signaling. Recruitment of Tim-3 to the immune synapse was mediated exclusively by the transmembrane domain, replacement of which impaired the ability of Tim-3 to costimulate T cell receptor (TCR)–dependent S6 phosphorylation. Furthermore, enforced localization of the Tim-3 cytoplasmic domain to the immune synapse in a chimeric antigen receptor still enabled T cell activation. Together, our findings are consistent with a model whereby Tim-3 enhances TCR-proximal signaling under acute conditions.


Blood ◽  
2006 ◽  
Vol 107 (3) ◽  
pp. 1010-1017 ◽  
Author(s):  
Peta J. O'Connell ◽  
Xiangbin Wang ◽  
Matilde Leon-Ponte ◽  
Corrie Griffiths ◽  
Sandeep C. Pingle ◽  
...  

AbstractAdaptive immunity is triggered at the immune synapse, where peptide-major histocompatibility complexes and costimulatory molecules expressed by dendritic cells (DCs) are physically presented to T cells. Here we describe transmission of the inflammatory monoamine serotonin (5-hydroxytryptamine [5-HT]) between these cells. DCs take up 5-HT from the microenvironment and from activated T cells (that synthesize 5-HT) and this uptake is inhibited by the antidepressant, fluoxetine. Expression of 5-HT transporters (SERTs) is regulated by DC maturation, exposure to microbial stimuli, and physical interactions with T cells. Significantly, 5-HT sequestered by DCs is stored within LAMP-1+ vesicles and subsequently released via Ca2+-dependent exocytosis, which was confirmed by amperometric recordings. In turn, extracellular 5-HT can reduce T-cell levels of cAMP, a modulator of T-cell activation. Thus, through the uptake of 5-HT at sites of inflammation, and from activated T cells, DCs may shuttle 5-HT to naive T cells and thereby modulate T-cell proliferation and differentiation. These data constitute the first direct measurement of triggered exocytosis by DCs and reveal a new and rapid type of signaling that may be optimized by the intimate synaptic environment between DCs and T cells. Moreover, these results highlight an important role for 5-HT signaling in immune function and the potential consequences of commonly used drugs that target 5-HT uptake and release.


2020 ◽  
Vol 21 (5) ◽  
pp. 1568 ◽  
Author(s):  
Ferenc Papp ◽  
Peter Hajdu ◽  
Gabor Tajti ◽  
Agnes Toth ◽  
Eva Nagy ◽  
...  

The immunological synapse (IS) is a specialized contact area formed between a T cell and an antigen presenting cell (APC). Besides molecules directly involved in antigen recognition such as the TCR/CD3 complex, ion channels important in the membrane potential and intracellular free Ca2+ concentration control of T cells are also recruited into the IS. These are the voltage-gated Kv1.3 and Ca2+-activated KCa3.1 K+ channels and the calcium release-activated Ca2+ channel (CRAC). However, the consequence of this recruitment on membrane potential and Ca2+ level control is not known. Here we demonstrate that the membrane potential (MP) of murine T cells conjugated with APCs in an IS shows characteristic oscillations. We found that depolarization of the membrane by current injection or by increased extracellular K+ concentration produced membrane potential oscillations (MPO) significantly more frequently in conjugated T cells than in lone T cells. Furthermore, oscillation of the free intracellular Ca2+ concentration could also be observed more frequently in cells forming an IS than in lone cells. We suggest that in the IS the special arrangement of channels and the constrained space between the interacting cells creates a favorable environment for these oscillations, which may enhance the signaling process leading to T cell activation.


2017 ◽  
Vol 114 (30) ◽  
pp. E6117-E6126 ◽  
Author(s):  
Thomas C. J. Tan ◽  
John Knight ◽  
Thomas Sbarrato ◽  
Kate Dudek ◽  
Anne E. Willis ◽  
...  

Global transcriptomic and proteomic analyses of T cells have been rich sources of unbiased data for understanding T-cell activation. Lack of full concordance of these datasets has illustrated that important facets of T-cell activation are controlled at the level of translation. We undertook translatome analysis of CD8 T-cell activation, combining polysome profiling and microarray analysis. We revealed that altering T-cell receptor stimulation influenced recruitment of mRNAs to heavy polysomes and translation of subsets of genes. A major pathway that was compromised, when TCR signaling was suboptimal, was linked to ribosome biogenesis, a rate-limiting factor in both cell growth and proliferation. Defective TCR signaling affected transcription and processing of ribosomal RNA precursors, as well as the translation of specific ribosomal proteins and translation factors. Mechanistically, IL-2 production was compromised in weakly stimulated T cells, affecting the abundance of Myc protein, a known regulator of ribosome biogenesis. Consequently, weakly activated T cells showed impaired production of ribosomes and a failure to maintain proliferative capacity after stimulation. We demonstrate that primary T cells respond to various environmental cues by regulating ribosome biogenesis and mRNA translation at multiple levels to sustain proliferation and differentiation.


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.


Blood ◽  
2010 ◽  
Vol 116 (18) ◽  
pp. 3475-3484 ◽  
Author(s):  
Tobias Woehrle ◽  
Linda Yip ◽  
Abdallah Elkhal ◽  
Yuka Sumi ◽  
Yu Chen ◽  
...  

Abstract Engagement of T cells with antigen-presenting cells requires T-cell receptor (TCR) stimulation at the immune synapse. We previously reported that TCR stimulation induces the release of cellular adenosine-5′-triphosphate (ATP) that regulates T-cell activation. Here we tested the roles of pannexin-1 hemichannels, which have been implicated in ATP release, and of various P2X receptors, which serve as ATP-gated Ca2+ channels, in events that control T-cell activation. TCR stimulation results in the translocation of P2X1 and P2X4 receptors and pannexin-1 hemichannels to the immune synapse, while P2X7 receptors remain uniformly distributed on the cell surface. Removal of extracellular ATP or inhibition, mutation, or silencing of P2X1 and P2X4 receptors inhibits Ca2+ entry, nuclear factors of activated T cells (NFAT) activation, and induction of interleukin-2 synthesis. Inhibition of pannexin-1 hemichannels suppresses TCR-induced ATP release, Ca2+ entry, and T-cell activation. We conclude that pannexin-1 hemichannels and P2X1 and P2X4 receptors facilitate ATP release and autocrine feedback mechanisms that control Ca2+ entry and T-cell activa-tion at the immune synapse.


2019 ◽  
Author(s):  
Shunsuke Kataoka ◽  
Priyanka Manandhar ◽  
Creg J. Workman ◽  
Hridesh Banerjee ◽  
Andrea L. Szymczak-Workman ◽  
...  

AbstractExpression of the transmembrane protein Tim-3 is increased on dysregulated T cells undergoing chronic T cell activation, including in chronic infection and solid tumors. We and others previously reported that Tim-3 exerts apparently paradoxical co-stimulatory activity in T cells (and other cells), including enhancement of ribosomal S6 protein phosphorylation (pS6). Here we examined the upstream signaling pathways that control Tim3-mediated increases in pS6 in T cells. We have also defined the localization of Tim-3 relative to the T cell immune synapse and impacts on downstream signaling. Recruitment of Tim-3 to the immune synapse was regulated exclusively by the transmembrane domain, replacement of which impaired Tim-3 co-stimulation of pS6. Strikingly, enforced localization of the Tim-3 cytoplasmic domain to the immune synapse in the context of a chimeric antigen receptor still allowed for robust T cell activation. Our findings are consistent with a model whereby Tim-3 enhances TCR-proximal signaling under acute conditions.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 8045-8045 ◽  
Author(s):  
Christian Leisner ◽  
Leonardo Borras ◽  
Stephanie Jungmichel ◽  
Philipp Richle ◽  
Fabian Scheifele ◽  
...  

8045 Background: The BCMA-targeting bispecific T-cell engager AMG420 emerged as the first bispecific that achieved responses similar to CAR-T therapies in patients with relapsed/refractory (RR) multiple myeloma (MM). Despite improved ORR, the median duration until relapse is currently limited to approximately 12 months. Persistent minimal residual disease drives relapse and is characterized by increased expression of PD-1/PD-L1. Efficacy with checkpoint inhibitors is compromised by 1) their activity not been targeted specifically to the immune synapse between T cells and cancer cells, and 2) dose-limiting broadly distributed immune-related adverse events, which has halted several clinical trials. This underscores the need for localized checkpoint inhibition within the cytolytic synapse. We developed a Local Activator and T cell Engager (LocATE) antibody that combines binding to CD3 and BCMA with selective blockade of PD-L1 at the immune synapse in just one scaffold. Selectivity is achieved via low afffinity for PD-L1 and high affinity for BCMA. Methods: Antibody mediated Cytotoxicity (LDH assay) and T cell activation (IL-2 release) was measured in vitro using MM cell lines together with isolated human CD3+ T cells. Human ex vivo T cell activity and redirection was evaluated on fresh bone marrow biopsies from MM patients with different disease stages by automated microscopy (pharmacoscopy) and image analysis. Results: The LocATE antibody showed a 5-fold increase in T cell activation and MM cell killing in vitro compared to a BCMAxCD3 BiTE. Furthermore, patient-derived MM cells showed up to a 19-fold increase in T cell activation as compared to a BCMAxCD3 BiTE or a combination of BiTE and PD-L1 inhibitor, while no activity was observed on healthy cells. Conclusions: These results suggest that T cell redirection with simultaneous checkpoint inhibition in the synapse is highly potent while minimizing off-tumor toxicity, therefore, has high therapeutic potential for patients with relapsed MM.


Sign in / Sign up

Export Citation Format

Share Document