scholarly journals Mapping the extent of heterogeneity of human CCR5+ CD4+ T cells in peripheral blood and lymph nodes

AIDS ◽  
2020 ◽  
Vol 34 (6) ◽  
pp. 833-848
Author(s):  
John Zaunders ◽  
C. Mee Ling Munier ◽  
Helen M. McGuire ◽  
Hannah Law ◽  
Annett Howe ◽  
...  
Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 132-132
Author(s):  
Pengcheng Xu ◽  
Yajing Zhao ◽  
Ming Hou ◽  
Panpan Han

Abstract Immune thrombocytopenia (ITP) is an acquired autoimmune bleeding disorder, accounting for about 1/3 of clinical hemorrhagic diseases. Loss of peripheral immune tolerance through simultaneous decrease of CD4+CD25+Foxp3+ regulatory T cells (Tregs) as well as unrestricted proliferation and activation of peripheral CD4+ effector T cells underpin the pathophysiology of ITP. Atorvastatin (AT), an inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, could competitively combine with HMG-CoA reductase and inhibit the production of cholesterol, accompanying with the decrease of some intermediate metabolites, such as small GTPase. Recent studies have found that statins could regulate the homeostasis of effector T cells and Tregs in some autoimmune diseases and enhance bone marrow endothelial cell function in corticosteroid-resistant ITP. However, whether AT could target the Tregs/effector T cell-axis to restore the peripheral immune tolerance in ITP is unknown. To assess the effect of AT in ITP, CD4+ T cells were isolated magnetically from peripheral blood mononuclear cells of ITP patients and cultured with different doses of AT (0μM, 5μM, 10μM, 20μM) for 3 days. The activation of CD4+ T cells were analyzed by flow cytometry. It was shown that AT could significantly inhibit the expression of CD25 on CD4+ T cells, CD4+CD45RA+ naïve T cells and CD4+CD45RO+ memory T cells and impede the switching from CD45RA to RO dose-dependently. Moreover, AT was also effective in reducing the early activation of CD4+ T cells by decreasing the expression of CD69. The dampened activation of CD4+ T cells could be reversed after blocking AT by L-mevalonate (L-MA). These results suggested that AT can inhibit the activation of CD4+ T cells and naïve T cells in vitro. We further analyzed the influence of AT on the proliferation, apoptosis and cell cycle progression of CD4+ T cells. The isolated CD4+ T cells were labeled with CFSE and cultured with AT for 7 days. AT was observed to significantly inhibit the proliferation of CD4+ T cells in a dose-dependent manner and found to induce the apoptosis of CD4+ T cells with the cell arrest in G1 phase. In line with the previous studies about the promotion of Tregs after AT treatment, our in vitro study showed that the ratio of CD4+CD25+Foxp3+ Tregs among CD4+CD25+ T cells were elevated after AT treatment, suggesting that AT could increase the proportion of Treg in activated CD4+ T cells. Furthermore, as it was reported that AT could target some small GTPase to exert its regulation on T cells, we tested the regulation role of AT on the activation of Rho, Rac and Ras by western blot. It was shown that the expression of Ras and Rho of CD4+ T cells was decreased after AT administration in the culture system, and further influence on activation of small GTPase will be confirmed by pull-down assays. Finally, in the in vivo study, we established the murine passive ITP models by injecting anti-CD41 antibody and divided them randomly into AT group (AT 40mg/kg/d) and control group (same dose of PBS). The platelet count were detected every other day and the expression of CD25 and Foxp3 on CD4+T in thymus, lymph nodes, spleen and peripheral blood of mice were determined after 7 days. There was no difference on the expression of CD25+ on CD4+ T cells in peripheral blood, lymph nodes, thymus and spleen between the two groups. But increased number of Tregs in the lymph nodes, peripheral blood and spleen of the AT group and decreased number of Tregs in thymus were observed compared to the control group, suggesting that AT could induce the development of peripheral Tregs and facilitate the migration of Tregs from thymus to peripheral organs in the ITP murine models. Due to the short period of the mice model, we didn't observe a significant increase in the platelet number after AT treatment. Our current results showed that AT played an important role in regulating peripheral immune tolerance by inhibiting the activation, proliferation, expansion and survival of CD4+ T cells, whereas increasing the number of Tregs with dampened GTPase activation. The regulatory role of AT was recapitulated in the ITP murine models. This novel mechanism of AT underlies the potential therapeutic strategy for ITP. Figure. Figure. Disclosures No relevant conflicts of interest to declare.


2014 ◽  
Vol 52 (08) ◽  
Author(s):  
M Rau ◽  
AK Schilling ◽  
J Meertens ◽  
I Hering ◽  
T Kudlich ◽  
...  

Author(s):  
Kuan Lai ◽  
Wenjing Zhang ◽  
Songshan Li ◽  
Zhiwen Zhang ◽  
Shuangde Xie ◽  
...  

Abstract Pemphigus vulgaris (PV) is a chronic and potentially life-threatening autoimmune blistering disease. Aberrant mTOR pathway activity is involved in many autoimmune diseases. This study investigated the correlation of mTOR pathway (PI3K/AKT/mTOR/p70S6K) activity with the loss of balance in T helper 2/regulatory T (Th2/Treg) cells in the peripheral blood of PV patients. CD4+ T cells were isolated from 15 PV patients and 15 healthy controls (HCs), the ratios of Th2/CD4+ T cells and Treg/CD4+ T cells, the activity of the mTOR pathway (PI3K/AKT/mTOR/p70S6K), the transcription factors and cytokines of Th2 and Treg cells were detected. Primary CD4+ T cells from PV patients were cultured under Th2- or Treg-polarizing conditions with or without rapamycin in vitro. We found that PV patients showed significantly elevated serum IL-4 when compared with HCs, and serum IL-4 level was positively correlated with the titer of anti-Dsg1/3 antibody and disease severity, while the serum TGF-β level was negatively correlated with the titer of anti-Dsg3 antibody and disease severity. Meanwhile, PV patients showed increased Th2/CD4+ T cell ratio; decreased Treg/CD4+ T cell ratio; elevated mRNA of PI3K, AKT, mTOR and protein of PI3K (P85), AKT, p-AKT (Ser473), mTOR, p-mTOR (Ser2448), p-p70S6K (Thr389), GATA3; reduced protein of forkhead box protein 3. Rapamycin inhibited Th2 cell differentiation and promoted Treg cell differentiation in vitro. These data suggest a close association between mTOR pathway activation and the loss of balance in Th2/Treg cells in peripheral blood of PV patients. Inhibiting mTORC1 can help restore the Th2/Treg balance.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii89-ii89
Author(s):  
Subhajit Ghosh ◽  
Ran Yan ◽  
Sukrutha Thotala ◽  
Arijita Jash ◽  
Anita Mahadevan ◽  
...  

Abstract BACKGROUND Patients with glioblastoma (GBM) are treated with radiation (RT) and temozolomide (TMZ). These treatments can cause prolonged severe lymphopenia, which is associated with shorter survival. NT-I7 (efineptakin alfa) is a long-acting recombinant human IL-7 that supports the proliferation and survival CD4+ and CD8+ cells in both human and mice. We tested whether NT-I7 would protect T cells from treatment-induced lymphopenia and improve survival. METHODS C57BL/6 mice bearing intracranial tumors (GL261 or CT2A) were treated with RT (1.8 Gy/day x 5 days), TMZ (33 mg/kg/day x 5 days) and/or NT-17 (10 mg/kg on the final day of RT completion). We followed for survival and profiled CD3, CD8, CD4, FOXP3 in peripheral blood over time. In parallel, we assessed cervical lymph nodes, bone marrow, thymus, spleen, and the tumor 6 days after NT-I7 treatment. RESULTS Median survival in mice treated with NT-I7 combined with RT was significantly better than RT alone (GL261: 40d vs 34d, p< 0.0021; CT2A: 90d vs 40d, p< 0.0499) or NT-I7 alone (GL261: 40d vs 24d, p< 0.008; CT2A: 90d vs 32d, p< 0.0154). NT-17 with RT was just as effective as NT-I7 combined with RT and TMZ in both GL261 (40d vs 47d) and CT2A (90d vs 90d). NT-I7 treatment significantly increased the amount of CD8+ cells in the peripheral blood and tumor. NT- I7 rescued CD8+ T cells from RT induced lymphopenia in peripheral blood, spleen, and lymph nodes. NT-I7 alone or NT-I7 in combination with RT increased the CD8+ T cells in peripheral blood and tumor while reducing the FOXP3+ T-reg cells in the tumor microenvironment. CONCLUSIONS NT-I7 protects T-cells from RT induced lymphopenia, improves cytotoxic CD8+ T lymphocytes systemically and in the tumor, and improves survival. Presently, a phase I/II trial to evaluate NT-I7 in patients with high-grade gliomas is ongoing (NCT03687957).


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A599-A599
Author(s):  
Subhajit Ghosh ◽  
Ran Yan ◽  
Sukrutha Thotala ◽  
Arijita Jash ◽  
Anita Mahadevan ◽  
...  

BackgroundRadiation (RT) and temozolomide (TMZ), which are standard of care for patients with glioblastoma (GBM), can cause prolonged severe lymphopenia. Lymphopenia, in turn, is an independent risk factor for shorter survival. Interleukin-7 (IL-7) is a cytokine that is required for T cell homeostasis and proliferation. IL-7 levels are inappropriately low in GBM patients with lymphopenia. NT-I7 (efineptakin alfa) is a long-acting recombinant human IL-7 that supports the proliferation and survival CD4+ and CD8+ cells in both human and mice. We tested whether NT-I7 rescues treatment-induced lymphopenia and improves survival.MethodsImmunocompetent C57BL/6 mice bearing two intracranial glioma models (GL261 and CT2A) were treated with RT (1.8 Gy/day x 5 days), TMZ (33 mg/kg/day x 5 days) and/or NT-I7 (10 mg/kg on the final day of RT completion). We profiled the CD3, CD8, CD4, FOXP3 cells in peripheral blood over time. We also immunoprofiled cervical lymph nodes, bone marrow, thymus, spleen, and the tumor 6 days after NT-I7 treatment. Survival was monitored daily.ResultsMedian survival in mice treated with NT-I7 combined with RT was significantly longer than RT alone (GL261: 40d vs 34d, p<0.0021; CT2A: 90d vs 40d, p<0.0499) or NT-I7 alone (GL261: 40d vs 24d, p<0.008; CT2A: 90d vs 32d, p<0.0154). NT-I7 with RT was just as effective as NT-I7 combined with RT and TMZ in both GL261(40d vs 47d) and CT2A (90d vs 90d). Cytotoxic CD8+ T cells were increased in both peripheral blood (0.66 x 105 to 3.34 x 105; P≤0.0001) and tumor (0.53 x 103 to 1.83 x 103; P≤0.0001) in mice treated with NT-I7 when compared to control. Similarly, NT-I7 in combination with RT increased the CD8+ T cells in peripheral blood (0.658 x 105 to 1.839 x 105 P≤0.0001) when compared to RT alone. There were decreases in tumor infiltrating FOXP3+ T-reg cells in mice treated with NT-I7 (1.9 x 104 to 0.75 x 104 P≤0.0001) and NT-I7 + RT (1.9 x 104 to 0.59 x 104 P≤0.0001) when compared to the control group without NT-I7. In addition, NT- I7 treatment increased CD8+ T cells in thymus, spleen, and lymph nodes.ConclusionsNT-I7 enhances cytotoxic CD8+ T lymphocytes systemically and in the tumor microenvironment, and improves survival. A phase I/II trial to evaluate NT-I7 in patients with high-grade gliomas is ongoing (NCT03687957).


Sign in / Sign up

Export Citation Format

Share Document