Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 13: 940577, 2022.
Article in English | MEDLINE | ID: mdl-36248873

ABSTRACT

Background: Aging is usually accompanied by functional declines of the immune system, especially in T-cell responses. However, little is known about ways to alleviate this. Methods: Here, 37 middle-aged healthy participants were recruited, among which 32 were intravenously administrated with expanded NK cells and 5 with normal saline. Then, we monitored changes of peripheral senescent and exhausted T cells within 4 weeks after infusion by flow cytometry, as well as serum levels of senescence-associated secretory phenotype (SASP)-related factors. In vitro co-culture assays were performed to study NK-mediated cytotoxic activity against senescent or exhausted T cells. Functional and phenotypic alteration of NK cells before and after expansion was finally characterized. Results: After NK cell infusion, senescent CD28-, CD57+, CD28-CD57+, and CD28-KLRG1+ CD4+ and CD8+ T-cell populations decreased significantly, so did PD-1+ and TIM-3+ T cells. These changes were continuously observed for 4 weeks. Nevertheless, no significant changes were observed in the normal saline group. Moreover, SASP-related factors including IL-6, IL-8, IL-1α, IL-17, MIP-1α, MIP-1ß, and MMP1 were significantly decreased after NK cell infusion. Further co-culture assays showed that expanded NK cells specifically and dramatically eliminated senescent CD4+ T cells other than CD28+CD4+ T cells. They also showed improved cytotoxic activity, with different expression patterns of activating and inhibitory receptors including NKG2C, NKG2A, KLRG1, LAG3, CD57, and TIM3. Conclusion: Our findings imply that T-cell senescence and exhaustion is a reversible process in healthy individuals, and autologous NK cell administration can be introduced to alleviate the aging. Clinical Trial Registration: ClinicalTrials.gov, ChiCTR-OOh-17011878.


Subject(s)
CD28 Antigens , Hepatitis A Virus Cellular Receptor 2 , CD28 Antigens/metabolism , Chemokine CCL3/metabolism , Chemokine CCL4/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Killer Cells, Natural , Matrix Metalloproteinase 1/metabolism , Programmed Cell Death 1 Receptor/metabolism , Randomized Controlled Trials as Topic , Saline Solution/metabolism
2.
Cell Oncol (Dordr) ; 45(6): 1297-1309, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36260222

ABSTRACT

PURPOSE: Our study intended to explore how low-dose anti-angiogenic drugs affected anti-tumor immunity of tumor-infiltrating exhausted CD8+T cells and achieved better clinical response when combined with immunotherapy. We set out to find potential targets or predictive biomarker on CD8+T cells for immunotherapy. METHODS: We tested different doses of anti-VEGFR2 antibody combined with anti-PD1 antibody to treat LUAD in vivo and analyzed tumor-infiltrating CD8+T cells by flow cytometry. CD8+T cells overexpressing LAYN were co-cultured with LA795 cell lines to identify the function of LAYN in CD8+T cells. We also analyzed clinical samples from advanced LUAD patients treated with anti-angiogenesis therapy combined with immunotherapy. RESULTS: Low-dose anti-VEGFR2 antibody combined with anti-PD1 antibody treatment delayed tumor growth and prolonged the survival time of tumor-bearing mice. The number of tumor-infiltrating CD8+T cells was reduced and the expression of LAYN was down-regulated in tumor-infiltrating CD8+T cells in the low-dose anti-VEGFR2 combination group. It was found that LAYN inhibited the killing function of CD8+T cells. In patients with advanced LUAD who received anti-angiogenesis therapy combined with immunotherapy, the LAYN+CD8+T cell subpopulation in good responders was significantly higher than that in poor responders. Furthermore, we demonstrated the expression of LAYN was regulated by upstream transcription factor NR4A1. CONCLUSION: Low-dose anti-VEGFR2 antibody combined with anti-PD1 antibody therapy promoted anti-tumor immunity and the downregulation of LAYN in tumor-infiltrating CD8+T cells played an important role in this process. These findings had implications for improving the efficacy of immune checkpoint blockade therapy and further optimized clinical treatment guidelines in advanced LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Mice , Animals , Lymphocytes, Tumor-Infiltrating , CD8-Positive T-Lymphocytes , Immunotherapy , Adenocarcinoma of Lung/metabolism , Lung Neoplasms/pathology , Tumor Microenvironment , Carrier Proteins/metabolism , Membrane Glycoproteins/metabolism
3.
J Immunother Cancer ; 10(7)2022 07.
Article in English | MEDLINE | ID: mdl-35817479

ABSTRACT

BACKGROUND: Foxp3+ regulatory T (Treg) cells facilitate tumor immune evasion by forming a suppressive tumor microenvironment. Therefore, immune therapies promoting Treg fragility may greatly enhance immune checkpoint blockade (ICB) efficacy in cancers. METHODS: We have screened 2640 compounds and identified the gut microbial metabolite gallic acid, which promotes Foxp3 degradation and Treg instability by repressing Usp21 gene transcription. In vivo and in vitro experiments have been performed to explore the roles of Usp21 in Treg cells. Importantly, we treated tumor-bearing mice with gallic acid and anti-PD-1 antibody to explore the potential therapeutic value of gallic acid in clinical cancer immunotherapy. RESULTS: Mechanistically, gallic acid prevents STAT3 phosphorylation and the binding of phosphorylated STAT3 to Usp21 gene promoter. The deubiquitinated Usp21 and stabilized PD-L1 proteins boost the function of Treg cells. Combination of gallic acid and anti-PD-1 antibody, in colorectal cancer (CRC) treatment, not only significantly dampen Treg cell function by impairing PD-L1/PD-1 signaling and downregulating Foxp3 stability, but also promote CD8+ T cells' production of IFN-γ and limited tumor growth. CONCLUSION: Our findings have implications for improving the efficacy of ICB therapy in CRC by inducing T-helper-1-like Foxp3lo Treg cells.


Subject(s)
B7-H1 Antigen , Neoplasms , Animals , CD8-Positive T-Lymphocytes , Forkhead Transcription Factors/metabolism , Gallic Acid/pharmacology , Gallic Acid/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice , Tumor Microenvironment
4.
FEBS J ; 289(2): 319-335, 2022 01.
Article in English | MEDLINE | ID: mdl-33529458

ABSTRACT

Regulatory T (Treg) cells are critical for the maintenance of immune homeostasis. Dysregulation of Treg cells has been implicated in the pathogenesis of autoimmunity and chronic inflammation, while aging is characterized by an accumulation of inflammatory markers in the peripheral blood, a phenomenon known as 'inflammaging'. The relationship between Treg cells and age-related diseases remains to be further studied. Increasing evidence revealed that Treg cells' dysfunction occurs in aged patients, suggesting that immune therapies targeting Treg cells may be a promising approach to treat diseases such as cancers and autoimmune diseases. Furthermore, drugs targeting Treg cells show encouraging results and contribute to CD8+ T-cell-mediated cytotoxic killing of tumor and infected cells. In general, a better understanding of Treg cell function may help us to develop new immune therapies against aging. In this review, we discuss potential therapeutic strategies to modify immune responses of relevance for aging to prevent and treat age-related diseases, as well as the challenges posed by the translation of novel immune therapies into clinical practice.


Subject(s)
Aging/immunology , Forkhead Transcription Factors/genetics , Immunity/immunology , T-Lymphocytes, Regulatory/immunology , Aging/pathology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/immunology , Homeostasis/immunology , Humans , Immunity/genetics , Inflammation/immunology , Inflammation/therapy , Leukocytes, Mononuclear/immunology , Neoplasms/immunology , Neoplasms/therapy
5.
Nat Immunol ; 22(9): 1175-1185, 2021 09.
Article in English | MEDLINE | ID: mdl-34429546

ABSTRACT

Systematic characterizations of adipose regulatory T (Treg) cell subsets and their phenotypes remain uncommon. Using single-cell ATAC-sequencing and paired single-cell RNA and T cell receptor (TCR) sequencing to map mouse adipose Treg cells, we identified CD73hiST2lo and CD73loST2hi subsets with distinct clonal expansion patterns. Analysis of TCR-sharing data implied a state transition between CD73hiST2lo and CD73loST2hi subsets. Mechanistically, we revealed that insulin signaling occurs through a HIF-1α-Med23-PPAR-γ axis to drive the transition of CD73hiST2lo into a CD73loST2hi adipose Treg cell subset. Treg cells deficient in insulin receptor, HIF-1α or Med23 have decreased PPAR-γ expression that in turn promotes accumulation of CD73hiST2lo adipose Treg cells and physiological adenosine production to activate beige fat biogenesis. We therefore unveiled a developmental trajectory of adipose Treg cells and its dependence on insulin signaling. Our findings have implications for understanding the dynamics of adipose Treg cell subsets in aged and obese contexts.


Subject(s)
Adipose Tissue/immunology , Insulin Resistance/immunology , Insulin/metabolism , Receptor, Insulin/metabolism , T-Lymphocytes, Regulatory/immunology , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Adipose Tissue/cytology , Aging/immunology , Animals , Cells, Cultured , High-Throughput Nucleotide Sequencing , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Male , Mediator Complex/metabolism , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/immunology , PPAR gamma/metabolism , Receptors, Antigen, T-Cell/genetics , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/cytology
SELECTION OF CITATIONS
SEARCH DETAIL