regulatory t cells
Recently Published Documents


TOTAL DOCUMENTS

11393
(FIVE YEARS 1832)

H-INDEX

260
(FIVE YEARS 21)

2022 ◽  
Vol 104 ◽  
pp. 108469
Author(s):  
Bo-Jin Chen ◽  
Jing-Wen Zhao ◽  
Da-Hong Zhang ◽  
Ai-Hong Zheng ◽  
Guo-Qing Wu

2022 ◽  
Vol 70 ◽  
pp. 101512
Author(s):  
Zahra Beyzaei ◽  
Alireza Shojazadeh ◽  
Bita Geramizadeh

2022 ◽  
Author(s):  
Sho Sato ◽  
Hirokazu Matsushita ◽  
Daisuke Shintani ◽  
Nao Fujieda ◽  
Akira Yabuno ◽  
...  

Abstract Background Regulatory T cells (Tregs) play an important role in the antitumor immune response in epithelial ovarian cancer (EOC). To understand the immune-inhibitory networks of EOC, we addressed the association between Tregs and immune checkpoint expression on T cells in the tumor microenvironment of EOC Methods A total of 41 patients with stage IIIC and IV EOC were included in the analysis. We harvested cells from malignant ascites and investigated them using multi-color flow cytometry. We categorized the Tregs into 3 groups: effector-type Tregs, naïve Tregs and non-Tregs, based on the expression patterns of CD45RA and Foxp3 in CD4+ T cells. Furthermore, the relationships between the expression of various immune checkpoint molecules, such as PD-1, on CD8+T cells and each of the Treg subtypes was also evaluated. Results The median frequency of naïve Tregs, effector-type Tregs and non-Tregs were 0.2% (0-0.8), 2.0% (0-11.4) and 1.5% (0.1-6.3) in CD4+ T cells of malignant ascites from EOC patients, respectively. A high frequency of effector-type Tregs was associated with high-grade serous carcinoma compared with the other histotypes. Patients with higher proportions of effector-type Tregs showed a trend towards increased progression-free survival. We also demonstrated a correlation between a higher proportion of effector-type Tregs and increased PD-1 expression on CD8+ T cells. In addition, C-C chemokine receptor 4 expression was also observed in effector-type Tregs. Conclusion These data suggest that multiple immune-inhibitory networks exist in malignant ascites from EOC patients, suggesting an approach towards combinational immunotherapies for advanced EOC patients.


Transfusion ◽  
2022 ◽  
Author(s):  
Tewodros Mamo ◽  
Keli L. Hippen ◽  
Margaret L. MacMillan ◽  
Claudio G. Brunstein ◽  
Jeffrey S. Miller ◽  
...  

2022 ◽  
Vol 23 (2) ◽  
pp. 732
Author(s):  
Katrin Peckert-Maier ◽  
Dmytro Royzman ◽  
Pia Langguth ◽  
Anita Marosan ◽  
Astrid Strack ◽  
...  

Chronic inflammatory diseases and transplant rejection represent major challenges for modern health care. Thus, identification of immune checkpoints that contribute to resolution of inflammation is key to developing novel therapeutic agents for those conditions. In recent years, the CD83 (cluster of differentiation 83) protein has emerged as an interesting potential candidate for such a “pro-resolution” therapy. This molecule occurs in a membrane-bound and a soluble isoform (mCD83 and sCD83, respectively), both of which are involved in resolution of inflammation. Originally described as a maturation marker on dendritic cells (DCs), mCD83 is also expressed by activated B and T cells as well as regulatory T cells (Tregs) and controls turnover of MHC II molecules in the thymus, and thereby positive selection of CD4+ T cells. Additionally, it serves to confine overshooting (auto-)immune responses. Consequently, animals with a conditional deletion of CD83 in DCs or regulatory T cells suffer from impaired resolution of inflammation. Pro-resolving effects of sCD83 became evident in pre-clinical autoimmune and transplantation models, where application of sCD83 reduced disease symptoms and enhanced allograft survival, respectively. Here, we summarize recent advances regarding CD83-mediated resolution of inflammatory responses, its binding partners as well as induced signaling pathways, and emphasize its therapeutic potential for future clinical trials.


Author(s):  
Alba Angelina ◽  
Rodrigo Jiménez‐Saiz ◽  
Mario Pérez‐Diego ◽  
Angel Maldonado ◽  
Beate Rückert ◽  
...  

Author(s):  
Yuumi Okuzono ◽  
Yo Muraki ◽  
Shuji Sato

Abstract Tumor necrosis factor receptor 2 (TNFR2), a membrane-bound tumor necrosis factor receptor expressed by regulatory T cells (Tregs), participates in Treg proliferation. Although a specific TNFR2 pathway has been reported, the signaling mechanism has not been completely elucidated. This study sought to clarify TNFR2 signaling in human Tregs using amplicon sequencing and single-cell RNA-sequencing to assess Tregs treated with a TNFR2 agonist antibody. Pathway enrichment analysis based on differentially expressed genes highlighted tumor necrosis factor α signaling via nuclear factor-kappa B, interleukin-2 signal transducer and activator of transcription 5 signaling, interferon-γ response, and cell proliferation-related pathways in Tregs after TNFR2 activation. TNFR2-high Treg-focused analysis found that these pathways were fully activated in cancer Tregs, showing high TNFR2 expression. Collectively, these findings suggest that TNFR2 orchestrates multiple pathways in cancer Tregs, which could help cancer cells escape immune surveillance, making TNFR2 signaling a potential anticancer therapy target.


2022 ◽  
Author(s):  
Zuochen Du ◽  
Jinzhi Wang ◽  
Di Yang ◽  
Xiaoyu Sun ◽  
Lu Huang ◽  
...  

Abstract Cell metabolism is crucial for orchestrating the differentiation and function of regulatory T cells (Tregs). However, the underlying signaling mechanism that coordinates cell metabolism to regulate Treg activity is not completely understood. As a pivotal molecule in lipid metabolism, the role of SHIP-1 has been studied extensively in B cells and CD4 T cells, yet its regulatory role in Tregs remains unknown. In this study, we generated “SHIP-1 KO mice” that have SHIP-1 specifically deleted in regulatory T cells by crossing Foxp3YFP-cre mice with SHIP-1fl/fl mice. Surprisingly, SHIP-1 KO mice had severe autoimmunity with increased Tregs in the thymus and disrupted peripheral T cell homeostasis. Mechanistically, CD4Cre SHIP-1flox/flox mice were found to have increased Treg precursors and SHIP-1 KO Tregs had reduced migration and stability, which caused decreased Tregs in the spleen. Additionally, the suppressive function of Tregs from SHIP-1 KO mice was diminished, along with their promotion of anti-tumor immunity. Interestingly, the PI3K-mTORC1, but not mTORC2, signaling axis was enhanced in SHIP-1 KO Tregs. In vivo treatment of SHIP-1 KO mice with rapamycin rescued the abnormal Treg percentages and peripheral T cell homeostasis, as well as Treg suppressive function. Furthermore, the treatment of wild-type mice with SHIP-1 inhibitor enhanced anti-tumor activity. Our study has revealed a previously unrecognized underlying function of SHIP-1 in Tregs, which highlights the SHIP-1-PI3K-mTORC1 axis that regulates Treg differentiation and function.


Sign in / Sign up

Export Citation Format

Share Document