Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Front Oncol ; 12: 858379, 2022.
Article in English | MEDLINE | ID: mdl-35656514

ABSTRACT

The immune checkpoint programmed death-ligand 1 (PD-L1) is expressed on the cell surface of tumor cells and is key for maintaining an immunosuppressive microenvironment through its interaction with the programmed death 1 (PD-1). Clear cell renal cell carcinoma (ccRCC) is a highly immunogenic cancer characterized by an aberrant aerobic glycolytic metabolism and is known to overexpress PD-L1. Multiple immunotherapies have been approved for the treatment of ccRCC, including cytokines and immune checkpoint inhibitors. Recently the intrinsic role of PD-L1 and interferon gamma (IFNγ) signaling have been studied in several types of tumor cells, yet it remains unclear how they affect the metabolism and signaling pathways of ccRCC. Using metabolomics, metabolic assays and RNAseq, we showed that IFNγ enhanced aerobic glycolysis and tryptophan metabolism in ccRCC cells in vitro and induced the transcriptional expression of signaling pathways related to inflammation, cell proliferation and cellular energetics. These metabolic and transcriptional effects were partially reversed following transient PD-L1 silencing. Aerobic glycolysis, as well as signaling pathways related to inflammation, were not induced by IFNγ when PD-L1 was silenced, however, tryptophan metabolism and activation of Jak2 and STAT1 were maintained. Our data demonstrate that PD-L1 expression is required to mediate some of IFNγ's effect in ccRCC cells and highlight the importance of PD-L1 signaling in regulating the metabolism of ccRCC cells in response to inflammatory signals.

2.
Front Oncol ; 11: 626748, 2021.
Article in English | MEDLINE | ID: mdl-33718196

ABSTRACT

The use of immune checkpoint blockade, in particular PD-1 and PD-L1 inhibitors, is now commonplace in many clinical settings including the treatment of muscle-invasive bladder cancer (MIBC). Notwithstanding, little information exists regarding the expression of the alternative PD-1 ligand, PD-L2 in urothelial bladder cancer (UBC). We therefore set out to characterise the expression of PD-L2 in comparison to PD-L1. Firstly, we assessed PD-L2 expression by immunohistochemistry and found widespread expression of PD-L2 in UBC, albeit with reduced expression in MIBC. We further investigated these findings using RNA-seq data from a cohort of 575 patients demonstrating that PDCD1LG2 (PD-L2) is widely expressed in UBC and correlated with CD274 (PD-L1). However, in contrast to our immunohistochemistry findings, expression was significantly increased in advanced disease. We have also provided detailed evidence of constitutive PD-L2 expression in normal urothelium and propose a mechanism by which PD-L2 is cleaved from the cell surface in MIBC. These data provide a comprehensive assessment of PD-L2 in UBC, showing PD-L2 is abundant in UBC and, importantly, constitutively present in normal urothelium. These data have implications for future development of immune checkpoint blockade, and also the understanding of the function of the immune system in the normal urinary bladder.

3.
Front Immunol ; 11: 574491, 2020.
Article in English | MEDLINE | ID: mdl-33193363

ABSTRACT

Cutaneous Leishmaniasis (CL) affects up to one million people every year and treatments are costly and toxic. The regulation of the host immune response is complex and the knowledge of how CD4+ T cells are activated and maintained during Leishmania infection is still limited. Current therapies aim to target programmed cell death (PD)-1 and programmed cell death ligand (PD-L)-1 in order to boost T cell activity. However, the role of the PD-1/PD-L1 axis during Leishmania infection is still unclear. In this study, we found that patients with active and post-treatment CL displayed different subsets of CD4+PD-1+ T cells. Accordingly, L. major-infected mice upregulated PD-1 on activated CD4+ T effector cells and PD-L1 on resident macrophages and infiltrating monocytes at the site of infection. L. major-infected Pdl1-/- mice expressed lower levels of MHCII and higher levels of CD206 on macrophages and monocytes and, more importantly, the lack of PD-L1 contributed to a reduced frequency of CD4+Ly6Chi T effector cells and an increase of CD4+Foxp3+ regulatory T cells at the site of infection and in draining lymph nodes. Additionally, the lack of PD-L1 was associated with lower production of IL-27 by infiltrating monocytes and lower levels of the Th1 cytokines IFN-γ and TNF-α produced by CD4+ T effector cells. Pdl1-/- mice initially exhibited larger lesions despite having a similar parasite load. Our results describe for the first time how the interruption of the PD-1/PD-L1 axis influences the immune response against CL and suggests that this axis regulates the balance between CD4+Ly6Chi T effector cells and CD4+Foxp3+ regulatory T cells.


Subject(s)
B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , Leishmaniasis, Cutaneous/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocyte Subsets/immunology , Adolescent , Adult , Animals , B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Female , Humans , Inflammation , Leishmania , Leishmaniasis, Cutaneous/blood , Leishmaniasis, Cutaneous/pathology , Lymph Nodes/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Phenotype , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Young Adult
4.
Cancers (Basel) ; 12(4)2020 Apr 10.
Article in English | MEDLINE | ID: mdl-32290052

ABSTRACT

PD-L1 expressed on tumor cells contributes to disease progression with evasion from tumor immunity. Plasma cells from multiple myeloma (MM) patients expressed higher levels of PD-L1 compared with healthy volunteers and monoclonal gammopathy of undetermined significance (MGUS) patients, and its expression is significantly upregulated in relapsed/refractory patients. Furthermore, high PD-L1 expression is induced by the myeloma microenvironment and PD-L1+ patients with MGUS and asymptomatic MM tend to show disease progression. PD-L1 expression on myeloma cells was associated with more proliferative potential and resistance to antimyeloma agents because of activation of the Akt pathway through PD-1-bound PD-L1 in MM cells. Those data suggest that PD-L1 plays a crucial role in the disease progression of MM.

5.
Front Immunol ; 9: 2129, 2018.
Article in English | MEDLINE | ID: mdl-30356906

ABSTRACT

Interferon-α (IFNα) has one of the longest histories of use amongst cytokines in clinical oncology and has been applied for the treatment of many types of cancers. Due to its immune-activating properties, IFNα is also an attractive candidate for combinatory anti-cancer therapies. Despite its extensive use in animal tumor models as well as in several clinical trials, the different mechanisms underlying patient responses and affecting desirable clinical benefits are still under investigation. Here we show that in addition to its immune-activating properties, IFNα induces the expression of a key negative regulator, immunosuppressive PD-L1 molecule, in the majority of the specific immune cell populations, particularly in the dendritic cells (DC). DC can modulate immune responses by a variety of mechanisms, including expression of T-cell regulatory molecules and cytokines. Our results showed that treatment of DC with IFNα-2b led to pronounced up-regulation of surface expression of PD-L1 molecules, increased IL-6 and decreased IL-12 production. Moreover, we present evidence that IFNα-treated DC exhibited a reduced capacity to stimulate interferon-γ production in T cells compared to control DC. This T-cell response after treatment of DC with IFNα was recovered by a pre-treatment with an anti-PD-L1 blocking antibody. Further analyses revealed that IFNα regulated PD-L1 expression through the STAT3 and p38 signaling pathways, since blocking of STAT3 and p38 activation with specific inhibitors prevented PD-L1 up-regulation. Our findings underline the important roles of p38 and STAT3 in the regulation of PD-L1 expression and prove that IFNα induces STAT3/p38-mediated expression of PD-L1 and thereby a reduced stimulatory ability of DC. The augmentation of PD-L1 expression in immune cells through IFNα treatment should be considered by use of IFNα in an anti-cancer therapy.


Subject(s)
B7-H1 Antigen/metabolism , Dendritic Cells/immunology , Interferon-alpha/metabolism , MAP Kinase Signaling System/immunology , Animals , B7-H1 Antigen/immunology , Blood Buffy Coat/cytology , Cells, Cultured , Coculture Techniques , Dendritic Cells/metabolism , Healthy Volunteers , Humans , Interferon-alpha/immunology , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Primary Cell Culture , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Up-Regulation/immunology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Transpl Immunol ; 51: 21-29, 2018 12.
Article in English | MEDLINE | ID: mdl-30092337

ABSTRACT

Studies have reported that bone marrow mesenchymal stem cells (BMSCs) play an important role in immune regulation after organ transplantation. Some of the BMSC-mediated regulatory mechanisms are related to PD-L1 expression. However, the regulatory mechanism of PD-L1 expression is not fully understood. In this experiment, we confirmed the regulatory role of BMSCs in the rejection of liver transplantation and explored the possible mechanism by which PD-L1 expression is regulated in BMSCs. An orthotopic liver transplantation model in rats was established based on the "double-cuff technique". Third-generation BMSCs were injected into the rejection model rats via portal vein. At the same time, sera were obtained from rats in the allograft, isograft and sham-operated groups. The sera from these groups were separately added to BMSCs complete medium to partially mimic the in vivo environment in which BMSCs are exposed. After predicting and analysing microRNAs that regulate PD-L1 expression, we examined the microRNA and PD-L1 expression in BMSCs of the three groups cultured in the conditioned media. Moreover, a luciferase reporter assay was used to confirm the interaction between PD-L1 and microRNA. The study found that the liver function and liver graft histopathology of the BMSC-treated group were better than those of the allograft group. The Kaplan-Meier survival curve analysis showed that the median survival time (MST) of the BMSC-treated group was longer than that of the allograft group. Bioinformatics prediction and analysis showed that miR-17-5p is highly likely to regulate PD-L1. Compared with the other two groups of cells, BMSCs cultured with serum from allograft models showed higher PD-L1 expression, but lower miR-17-5p expression. Pearson correlation analysis showed that miR-17-5p and PD-L1 expression was negatively correlated, and further luciferase reporter assays confirmed that miR-17-5p interacted directly with the 3´-untranslated region (UTR) of PD-L1 mRNA. The results of this study demonstrate that BMSCs can attenuate liver allograft rejection and provide us with the idea that BMSCs may further upregulate PD-L1 expression by downregulating miR-17-5p expression, thereby attenuating liver allograft rejection.


Subject(s)
B7-H1 Antigen/metabolism , Graft Rejection/immunology , Liver Transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , MicroRNAs/genetics , Allografts/immunology , Animals , B7-H1 Antigen/genetics , Cells, Cultured , Down-Regulation , Graft Rejection/prevention & control , Humans , Male , Portal Vein/surgery , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Transplantation, Homologous , Up-Regulation
7.
Oral Oncol ; 51(3): 221-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25500094

ABSTRACT

Recent clinical results for PD-1 blockade therapy have demonstrated durable tumor control with minimal immune-related adverse effects. PD-L1 is induced in non-lymphoid tissue cells and tumor cells, in addition to tissue-recruiting immune cells, under inflammatory conditions triggered by several cytokines, especially IFN-γ, and exogenous stimuli delivered by pathogen-associated molecular patterns. Receptor-mediated signaling molecules that affect the cell cycle, proliferation, apoptosis, and survival (including NF-κB, MAPK, PI3K, mTOR, and JAK/STAT) are involved in PD-L1 induction. PD-L1 expression in tumor cells is also triggered by the signals described above, but in some instances, intrinsic cell alteration associated with carcinogenesis contributes to PD-L1 induction. The tumor suppressor genes PTEN and Lkb1 and epithelial-mesenchymal transition-related molecules are also involved in the regulation of PD-L1 expression. Notably, squamous cell carcinoma of the head and neck (SCCHN) often exhibits both host immunosuppression and cytogenetic alternations of tumor cells. Precise understanding of how PD-L1 expression is controlled will allow the development of effective approaches to PD-1 blockade therapy for patients with SCCHN.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Squamous Cell/metabolism , Epithelial Cells/metabolism , Head and Neck Neoplasms/metabolism , Animals , Epithelial-Mesenchymal Transition/physiology , Humans , Interferon-gamma/metabolism , Janus Kinases/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL