Non-Redundant Activity of GSK-3α and GSK-3β in T Cell-Mediated Tumour Rejection

2021 ◽  
Author(s):  
Lynette Steele ◽  
Aarren Mannion ◽  
Gary Shaw ◽  
Ken Maclennan ◽  
Graham P. Cook ◽  
...  
Keyword(s):  
T Cell ◽  
iScience ◽  
2021 ◽  
pp. 102555
Author(s):  
Lynette Steele ◽  
Aarren J. Mannion ◽  
Gary Shaw ◽  
Ken Maclennan ◽  
Graham P. Cook ◽  
...  
Keyword(s):  
T Cell ◽  

2020 ◽  
Author(s):  
Yanlian Xiong ◽  
Yueming Wang ◽  
Jiashen Zhang ◽  
Nannan Zhao ◽  
Hengchao Zhang ◽  
...  

Abstract Background: Mesenchymal stem cells (MSCs) was considered as regenerative therapeutic approach in both acute and chronic diseases. However, whether MSCs regulate the antioxidant metabolism of CD4+ T cells and weaken immunosenescence remains unclear. Here, we reported the protective effects of hPMSCs in aging-related CD4+ T cell senescence and identified the underlying mechanisms using a D-gal induced mouse aging model.Methods: In vivo study, 40 male C57BL/6 mice (8 weeks) were randomly divided into four groups: control group, D-gal group, hPMSC group and PBS group. In in vitro experiment, human naive CD4+ T (CD4CD45RA) cells were prepared using a naive CD4+ T cell isolation kit II and pretreated with the Akt inhibitor LY294002 and Nrf2 inhibitor ML385. Then, isolated naive CD4+ T cell were cocultured with hPMSCs for 72 h in the absence or presence of anti-CD3/CD28 Dynabeads and IL-2 as a mitogenic stimulus. Intracellular ROS changes were detected by flow cytometry. The activities of the antioxidant enzymes superoxide dismutase, glutathione peroxidase and catalase were measured by colorimetric analysis. The senescent T cells were detected SA-β-gal stain. The expression of aging related proteins were detected by Western blotting, RT-PCR and confocal microscopy.Results: We found that hPMSC treatment markedly decreased the ROS level, SA-β-gal positive cells number, senescence-associated secretory phenotype (IL-6 and OPN) expression and aging-related protein (P16 and P21) expression in senescent CD4+ T cells. Furthermore, hPMSC treatment effectively upregulated Nrf2 nuclear translocation and the expression of downstream target genes (HO-1, CAT, GCLC and NQO1) in senescent CD4+ T cells. Moreover, in vitro studies revealed that hPMSCs attenuated CD4+ T cell senescence by upregulating the Akt/GSK-3β/Fyn pathway to activate Nrf2 functions. Conversely, the antioxidant effects of hPMSCs were blocked by the Akt inhibitor LY294002 and Nrf2 inhibitor ML385 in senescent CD4+ T cells.Conclusions: Our results indicate that hPMSCs attenuate D-gal induced CD4+ T cell senescence by activating Nrf2-mediated antioxidant defenses and that upregulation of Nrf2 by hPMSCs is regulated via the Akt/GSK-3β/Fyn pathway.


1991 ◽  
Vol 34 (2) ◽  
pp. 103-110 ◽  
Author(s):  
P. A. Steerenberg ◽  
E. Geerse ◽  
W. H. De Jong ◽  
R. Burger ◽  
R. J. Scheper ◽  
...  

1985 ◽  
Vol 6 (2) ◽  
pp. 55-58 ◽  
Author(s):  
R.A. Robins ◽  
R.W. Baldwin

2013 ◽  
Vol 41 (12) ◽  
pp. 1016-1027.e1 ◽  
Author(s):  
Sylvie Shen ◽  
Guy Klamer ◽  
Ning Xu ◽  
Tracey A. O’Brien ◽  
Alla Dolnikov

2020 ◽  
Author(s):  
Yanlian Xiong ◽  
Yueming Wang ◽  
Jiashen Zhang ◽  
Nannan Zhao ◽  
Aiping Zhang ◽  
...  

Abstract Background: Mesenchymal stem cells (MSCs) was considered as regenerative therapeutic approach in both acute and chronic diseases. However, whether MSCs regulate the antioxidant metabolism of CD4+ T cells and weaken immunosenescence remains unclear. Here, we reported the protective effects of hPMSCs in aging-related CD4+ T cell senescence and identified the underlying mechanisms using a D-gal induced mouse aging model.Methods: In vivo study, 40 male C57BL/6 mice (8 weeks) were randomly divided into four groups: control group, D-gal group, hPMSC group and PBS group. In in vitro experiment, human naive CD4+ T (CD4CD45RA) cells were prepared using a naive CD4+ T cell isolation kit II and pretreated with the Akt inhibitor LY294002 and Nrf2 inhibitor ML385. Then, isolated naive CD4+ T cell were cocultured with hPMSCs for 72 h in the absence or presence of anti-CD3/CD28 Dynabeads and IL-2 as a mitogenic stimulus. Intracellular ROS changes were detected by flow cytometry. The activities of the antioxidant enzymes superoxide dismutase, glutathione peroxidase and catalase were measured by colorimetric analysis. The senescent T cells were detected SA-β-gal stain. The expression of aging related proteins were detected by Western blotting, RT-PCR and confocal microscopy.Results: We found that hPMSC treatment markedly decreased the ROS level, SA-β-gal positive cells number, senescence-associated secretory phenotype (IL-6 and OPN) expression and aging-related protein (P16 and P21) expression in senescent CD4+ T cells. Furthermore, hPMSC treatment effectively upregulated Nrf2 nuclear translocation and the expression of downstream target genes (HO-1, CAT, GCLC and NQO1) in senescent CD4+ T cells. Moreover, in vitro studies revealed that hPMSCs attenuated CD4+ T cell senescence by upregulating the Akt/GSK-3β/Fyn pathway to activate Nrf2 functions. Conversely, the antioxidant effects of hPMSCs were blocked by the Akt inhibitor LY294002 and Nrf2 inhibitor ML385 in senescent CD4+ T cells.Conclusions: Our results indicate that hPMSCs attenuate D-gal induced CD4+ T cell senescence by activating Nrf2-mediated antioxidant defenses and that upregulation of Nrf2 by hPMSCs is regulated via the Akt/GSK-3β/Fyn pathway.


Open Biology ◽  
2020 ◽  
Vol 10 (2) ◽  
pp. 190235 ◽  
Author(s):  
Stephanie J. Crowley ◽  
Patrick T. Bruck ◽  
Md Aladdin Bhuiyan ◽  
Amelia Mitchell-Gears ◽  
Michael J. Walsh ◽  
...  

Cancer-specific mutations can lead to peptides of unique sequence presented on MHC class I to CD8 T cells. These neoantigens can be potent tumour-rejection antigens, appear to be the driving force behind responsiveness to anti-CTLA-4 and anti-PD1/L1-based therapies and have been used to develop personalized vaccines. The platform for delivering neoantigen-based vaccines has varied, and further optimization of both platform and adjuvant will be necessary to achieve scalable vaccine products that are therapeutically effective at a reasonable cost. Here, we developed a platform for testing potential CD8 T cell tumour vaccine candidates. We used a high-affinity alpaca-derived VHH against MHC class II to deliver peptides to professional antigen-presenting cells. We show in vitro and in vivo that peptides derived from the model antigen ovalbumin are better able to activate naive ovalbumin-specific CD8 T cells when conjugated to an MHC class II-specific VHH when compared with an irrelevant control VHH. We then used the VHH-peptide platform to evaluate a panel of candidate neoantigens in vivo in a mouse model of pancreatic cancer. None of the candidate neoantigens tested led to protection from tumour challenge; however, we were able to show vaccine-induced CD8 T cell responses to a melanoma self-antigen that was augmented by combination therapy with the synthetic cytokine mimetic Neo2/15.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1921-1921 ◽  
Author(s):  
Francesca Chiarini ◽  
Federica Fala ◽  
Francesca Ricci ◽  
PierLuigi Tazzari ◽  
Annalisa Astolfi ◽  
...  

Abstract Constitutively activated phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian Target of Rapamycin (mTOR) signaling is a common feature of T-cell acute lymphoblastic leukaemia (T-ALL). Recently, it was demonstrated that activated Notch-1 leads to constitutive activation of the PI3K/Akt/mTOR pathway by HES1-mediated transcriptional suppression of the PTEN gene. In addition, PTEN is mutated in about 20% of T-ALL patients, and virtually all T-ALL cell lines that are resistant to Notch-1 inhibition with γ-secretase inhibitors, contain mutations leading to either no or low PTEN expression. These findings lend compelling weight for the application of PI3K/Akt/mTOR inhibitors in T-ALL. However, our knowledge of PI3K/Akt/mTOR signalling in T-ALL is still limited and it is not clear whether it could be an effective target for innovative therapeutic strategies. Here, we have characterized PI3K/Akt/mTOR signalling in T-ALL cell lines (Jurkat, MOLT-4, CEM) lacking PTEN expression, including one (CEM-R) which over expresses high levels of the membrane transporter, 170-kDa P-glycoprotein (P-gp), one of the main determinants of multidrug-resistance. While MOLT-4 cells display wild-type p53, both Jurkat and CEM have a non-functional p53 pathway. Moreover, we have analyzed the therapeutic potential of the dual PI3K/mTOR inhibitor, PI-103, a small synthetic molecule of the pyridofuropyrimidine class, on both T-ALL cell lines and patient samples. T-ALL cell lines expressed p110α, p110β, p110γ, and p110α PI3K. Moreover, they expressed Akt1 and Akt2, both of which were found to be constitutively phosphorylated on Ser 473 and Ser 474, respectively, by immunoprecipitation experiments. Treatment of T-ALL cell lines with selective pharmacological inhibitors of p110 PI3K isoforms, demonstrated that only a p110α PI3K inhibitor (PIK75) was cytotoxic, resulting in a 40–50% reduction of cell growth when used at 2 μM. Consistently, only PIK75 induced Akt1 and Akt2 dephosphorylation on Ser 473 or Ser 474, respectively, hinting that p110α is the most important isoform for the activation of downstream signalling events. PI-103 was cytotoxic to all T-ALL cell lines including P-gp overexpressing cells, as it reduced cell growth by approximately 70% when employed at 2 μM for 24 h. PI-103 IC50 ranged from 0.5 to 1.0 μM at 24 h. PI-103 treatment resulted in apoptotic cell death (about 30% at 6 h of exposure, when employed at 0.75 μM), as demonstrated by Annexin V/propidium iodide staining and cytofluorimetric analysis. PI-103 caused both Akt1 and Akt2 dephosphorylation, accompanied by dephosphorylation of the Akt downstream target, glycogen synthase kinase (GSK) -3β. Also mTOR downstream targets were dephosphorylated in response to PI-103, including p70S6 kinase, ribosomal S6 protein, and 4E-BP1. Moreover, PI-103 resulted in lower levels of c-Myc expression. PI-103 activated caspase-3, -8, and -9. In contrast, an mTOR inhibitor (rapamycin) was less cytotoxic than PI-103 (25–30% reduction of cell growth at 100 nM after 24 h), blocked cells in the G1 phase of the cell cycle, and was much less effective in inducing apoptosis (about 5% at 6 h of treatment). Remarkably, rapamycin was almost completely ineffective against CEM-R cells. A combination consisting of PIK75 and rapamycin was less cytotoxic to T-ALL cell lines than PI-103 alone. Furthermore, rapamycin treatment, at variance with PI-103, resulted in an overactivation of the Akt/ GSK-3β axis, as documented by increased phosphorylation levels of both Akt and GSK- 3β. PI-103 was also cytotoxic to primary lymphoblasts from patients with T-ALL (IC50: 0.15 nM at 96 h), displaying constitutive phosphorylation of Akt and 4E-BP1, as well as low/absent PTEN expression. These data indicate that multi-targeted therapy towards PI3K and mTOR, may serve as an efficient treatment towards T-ALL cells (including those over expressing P-gp and independently from p53 state) which require upregulation of PI3K/Akt/mTOR signaling for their survival and growth.


2016 ◽  
Vol 291 (40) ◽  
pp. 21085-21095 ◽  
Author(s):  
Sihua Wang ◽  
Yuan Zhang ◽  
Yan Wang ◽  
Ping Ye ◽  
Jun Li ◽  
...  

Immunology ◽  
2008 ◽  
Vol 123 (3) ◽  
pp. 367-377 ◽  
Author(s):  
Dru S. Dace ◽  
Peter W. Chen ◽  
Jerry Y. Niederkorn
Keyword(s):  
T Cell ◽  

Sign in / Sign up

Export Citation Format

Share Document