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.
J Immunol ; 211(12): 1767-1782, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37947442

ABSTRACT

Understanding the mechanisms underlying the acquisition and maintenance of effector function during T cell differentiation is important to unraveling how these processes can be dysregulated in the context of disease and manipulated for therapeutic intervention. In this study, we report the identification of a previously unappreciated regulator of murine T cell differentiation through the evaluation of a previously unreported activity of the kinase inhibitor, BioE-1197. Specifically, we demonstrate that liver kinase B1 (LKB1)-mediated activation of salt-inducible kinases epigenetically regulates cytokine recall potential in effector CD8+ and Th1 cells. Evaluation of this phenotype revealed that salt-inducible kinase-mediated phosphorylation-dependent stabilization of histone deacetylase 7 (HDAC7) occurred during late-stage effector differentiation. HDAC7 stabilization increased nuclear HDAC7 levels, which correlated with total and cytokine loci-specific reductions in the activating transcription mark histone 3 lysine 27 acetylation (H3K27Ac). Accordingly, HDAC7 stabilization diminished transcriptional induction of cytokine genes upon restimulation. Inhibition of this pathway during differentiation produced effector T cells epigenetically poised for enhanced cytokine recall. This work identifies a previously unrecognized target for enhancing effector T cell functionality.


Subject(s)
Cytokines , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases , Animals , Mice , Cell Differentiation , Cytokines/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism
2.
Cell Rep ; 42(1): 111987, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36640309

ABSTRACT

T cell activation, proliferation, function, and differentiation are tightly linked to proper metabolic reprogramming and regulation. By using [U-13C]glucose tracing, we reveal a critical role for GOT1 in promoting CD8+ T cell effector differentiation and function. Mechanistically, GOT1 enhances proliferation by maintaining intracellular redox balance and serine-mediated purine nucleotide biosynthesis. Further, GOT1 promotes the glycolytic programming and cytotoxic function of cytotoxic T lymphocytes via posttranslational regulation of HIF protein, potentially by regulating the levels of α-ketoglutarate. Conversely, genetic deletion of GOT1 promotes the generation of memory CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Memory T Cells , CD8-Positive T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic , Cell Differentiation/genetics , Glucose/metabolism , Immunologic Memory/genetics
3.
Curr Opin Pharmacol ; 53: 77-83, 2020 08.
Article in English | MEDLINE | ID: mdl-32781414

ABSTRACT

Adenosine signaling through A2AR serves as a negative regulator of the immune system. Unique to this suppressive pathway is its ability to impact numerous stromal and immune cells. Additionally, tumors exhibit elevated concentrations of adenosine further advancing the pathway's potential as a powerful target for activating anti-tumor immunity. The promise of this therapeutic strategy has been repeatedly demonstrated in mice, but has so far only yielded limited success in the clinic. Nonetheless, it is notable that many of these observed clinical responses have been in individuals resistant to prior immunotherapy. These observations suggest this pathway is indeed involved in tumor immune evasion. Thus, identifying the disparities between the translational and clinical implementation of this therapy becomes necessary. To this end, this review will revisit how and where adenosine-A2AR signaling regulates the immune system and anti-tumor immunity so as to reveal opportunities for improving the translational success of this immunotherapy.


Subject(s)
Immunotherapy , Neoplasms/therapy , Receptor, Adenosine A2A/immunology , Adenosine/immunology , Animals , Homeostasis , Humans , Neoplasms/immunology
4.
Angiogenesis ; 17(3): 573-85, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24197832

ABSTRACT

Most deaths associated with breast cancer, the most common malignancy in women, are caused by metastasis. Tumor associated macrophages significantly contribute to breast cancer progression and development of metastasis through the promotion of angiogenesis which involves a central regulator of macrophage functions: nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Macrophages are activated by macrophage colony stimulating factor (MCSF) and chemokine (C-C motif) ligand 2 (CCL2) to secrete angiogenic factors including vascular endothelial growth factor (VEGF). The release of MCSF from tumor cells is mediated by ectodomain shedding through tumor necrosis factor alpha converting enzyme activation (TACE). Here we determined whether tumor cells TACE-shed MCSF promotes angiogenesis through activation of the NF-κB pathway in macrophages and the subsequent release of VEGF. These interactions were modeled in vitro using a panel of mammary cells mimicking the breast cancer progression from normal murine mammary gland cells to metastatic 4T1 cells along with J774 macrophages, all derived from BALB/c mice. TACE and MCSF expressions were higher in metastatic cells compared to epithelial cells (p < 0.05). Tumor conditioned medias activated the expression of VEGF by macrophages through stimulation of the NF-κB pathway and resulting macrophage secretions that promoted high levels of endothelial cell tubes. Furthermore, the combinations of CCL2, also highly expressed by tumor cells, and MCSF promoted pro-angiogenic macrophages. These results highlight the key role of tumor cell TACE-shed MCSF and secreted CCL2 in stimulating pro-angiogenic macrophages.


Subject(s)
ADAM Proteins/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , NF-kappa B/metabolism , Neovascularization, Pathologic/metabolism , ADAM17 Protein , Animals , Cell Line, Tumor , Chemokine CCL2/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Macrophages/metabolism , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Metastasis , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
5.
Immunol Res ; 58(1): 87-100, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24072428

ABSTRACT

The role of the tumor microenvironment especially of tumor-associated macrophages (TAMs) in the progression and metastatic spread of breast cancer is well established. TAMs have primarily a M2 (wound-healing) phenotype with minimal cytotoxic activities. The mechanisms by which tumor cells influence TAMs to display a pro-tumor phenotype are still debated although the key roles of immunomodulatory cytokines released by tumor cells, including colony-stimulating factor 1, tumor necrosis factor (TNF) and soluble TNF receptors 1/2, soluble vascular cell adhesion molecule 1, soluble interleukin 6 receptor and amphiregulin, have been demonstrated. Importantly, these factors are released through ectodomain shedding by the activities of the tumor necrosis factor-alpha-converting enzyme (TACE/ADAM17). The role of TACE activation leading to autocrine effects on tumor progression has been extensively studied. In contrast, limited information is available on the role of tumor cell TACE activities on TAMs in breast cancer. TACE inhibitors, currently in clinical trials, will certainly affect TAMs and subsequently treatment outcomes based on the substrates it releases. Furthermore, whether targeting a subset of the molecules shed by TACE, specifically those leading to TAMs with altered functions and phenotype, holds greater therapeutic promises than past clinical trials of TACE antagonists' remains to be determined. Here, the potential roles of TACE ectodomain shedding in the breast tumor microenvironment are reviewed with a focus on the release of tumor-derived immunomodulatory factors shed by TACE that directs TAM phenotypes and functions.


Subject(s)
ADAM Proteins/immunology , Breast Neoplasms/immunology , Enzyme Activation/immunology , Macrophages/immunology , ADAM Proteins/antagonists & inhibitors , ADAM17 Protein , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Enzyme Inhibitors/pharmacology , Female , Humans , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...