Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 9(6)2021 06.
Article in English | MEDLINE | ID: mdl-34103351

ABSTRACT

BACKGROUND: T follicular helper cells (Tfh) are essential to shape B cell response during germinal center formation. Tfh accumulation has been reported in various human cancers, with positive or negative prognostic roles. However, the mechanisms explaining the accumulation of Tfh and their role in cancer remain obscure. METHODS: In vitro differentiated and mouse cell sorted Tfh phenotype was evaluated by flow cytometry and quantitative PCR (qPCR). Antitumor effect of Tfh was evaluated by adoptive transfer in different tumor-bearing mice models. The involvement of immune cells, cytokines and chemokines was evaluated, using depleting antibodies. Chemokines and cytokines expression and production were evaluated by qPCR and ELISA. In human, the impact of immune cells and chemokines on survival was evaluated by analyzing transcriptomic data from public databases and from our own patient cohorts. RESULTS: In this study, we show that Tfh exert an antitumor immune effect in a CD8+-dependent manner. Tfh produce interleukin-21, which sustains proliferation, viability, cytokine production and cytotoxic functions of exhausted T cells. The presence of Tfh is required for efficacy of antiprogrammed cell death ligand-1 therapy. Tfh accumulate in the tumor bed and draining lymph nodes in different mouse cancer models. This recruitment is due to the capacity of transforming growth factor ß to drive Chemokine (C-X-C motif) Ligand 13 expression, a chemoattractant of Tfh, by intratumor CD8+ T cells. Accumulation of Tfh and exhausted CD8+ T cells predicts cancer outcome in various cancer types. In patients treated with anti-programmed cell death-1 mAb, accumulation of Tfh and CD8+ at the tumor site is associated with outcome. CONCLUSION: This study provides evidence that CD8+/Tfh crosstalk is important in shaping antitumor immune response generated by immunotherapy.


Subject(s)
Brain Neoplasms/therapy , Breast Neoplasms/therapy , Glioblastoma/therapy , Immune Checkpoint Inhibitors/administration & dosage , T Follicular Helper Cells/transplantation , Adoptive Transfer , Animals , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/pharmacology , Brain Neoplasms/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Interleukins/genetics , Interleukins/metabolism , Mice , T Follicular Helper Cells/immunology , Treatment Outcome , Xenograft Model Antitumor Assays
2.
Cell Death Dis ; 10(7): 485, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31217433

ABSTRACT

Limitation of 5-fluorouracil (5-FU) anticancer efficacy is due to IL-1ß secretion by myeloid-derived suppressor cells (MDSC), according to a previous pre-clinical report. Release of mature IL-1ß is a consequence of 5-FU-mediated NLRP3 activation and subsequent caspase-1 activity in MDSC. IL-1ß sustains tumor growth recovery in 5-FU-treated mice. Docosahexaenoic acid (DHA) belongs to omega-3 fatty acid family and harbors both anticancer and anti-inflammatory properties, which could improve 5-FU chemotherapy. Here, we demonstrate that DHA inhibits 5-FU-induced IL-1ß secretion and caspase-1 activity in a MDSC cell line (MSC-2). Accordingly, we showed that DHA-enriched diet reduces circulating IL-1ß concentration and tumor recurrence in 5-FU-treated tumor-bearing mice. Treatment with 5-FU led to JNK activation through ROS production in MDSC. JNK inhibitor SP600125 as well as DHA-mediated JNK inactivation decreased IL-1ß secretion. The repression of 5-FU-induced caspase-1 activity by DHA supplementation is partially due to ß-arrestin-2-dependent inhibition of NLRP3 inflammasome activity but was independent of JNK pathway. Interestingly, we showed that DHA, through ß-arrestin-2-mediated inhibition of JNK pathway, reduces V5-tagged mature IL-1ß release induced by 5-FU, in MDSC stably overexpressing a V5-tagged mature IL-1ß form. Finally, we found a negative correlation between DHA content in plasma and the induction of caspase-1 activity in HLA-DR- CD33+ CD15+ MDSC of patients treated with 5-FU-based chemotherapy, strongly suggesting that our data are clinical relevant. Together, these data provide new insights on the regulation of IL-1ß secretion by DHA and on its potential benefit in 5-FU-based chemotherapy.


Subject(s)
Docosahexaenoic Acids/pharmacology , Fluorouracil/pharmacology , Inflammasomes/drug effects , Inflammasomes/metabolism , Interleukin-1beta/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Caspase 1/metabolism , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Docosahexaenoic Acids/therapeutic use , Female , Fluorouracil/therapeutic use , Humans , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , beta-Arrestin 2/metabolism
3.
Nat Commun ; 8(1): 2085, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29233972

ABSTRACT

Interferon regulatory factors (IRF) have critical functions in lymphoid development and in immune response regulation. Although many studies have described the function of IRF4 in CD4+ T cells, few have focused on the IRF4 homologue, IRF8. Here, we show that IRF8 is required for Th9 differentiation in vitro and in vivo. IRF8 functions through a transcription factor complex consisting of IRF8, IRF4, PU.1 and BATF, which binds to DNA and boosts Il9 transcription. By contrast, IRF8 deficiency promotes the expression of other genes such as Il4, as IRF8 dimerises with the transcriptional repressor ETV6 and inhibits Il4 expression. In vivo, IRF8 is essential for the anti-tumour effects of Th9 cells in mouse melanoma models. Our results show that IRF8 complexes boost the Th9 program and repress Il4 expression to modulate Th9 cell differentiation, thereby implicating IRF8 as a potential therapeutic target to affect Th9 responses in cancer therapy.


Subject(s)
Cell Differentiation/genetics , Interferon Regulatory Factors/metabolism , Melanoma/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Transcription, Genetic/immunology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/immunology , Female , Gene Knockdown Techniques , Humans , Interferon Regulatory Factors/genetics , Interleukin-9/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Trans-Activators/metabolism , Xenograft Model Antitumor Assays
4.
Cell Rep ; 19(4): 746-759, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28445726

ABSTRACT

Sirtuin-1 deacetylates proteins and has emerged as a critical regulator of different cellular processes, particularly inflammation. Basal SIRT1 activity was previously found to limit Th9 and enhance Th17 differentiation in mice, but the effect of pharmacological SIRT1 activation on T cell differentiation and antitumor responses remains unclear. Here, we find that SIRT1 pharmacological agonists selectively impede mouse and human Th17 cell differentiation. SIRT1 activation induces STAT3 deacetylation, thus reducing its ability to translocate into the nucleus, bind to Rorc promoter, and induce its transcription. SIRT1 agonists reduce tumor growth in mice by blocking Th17 cell differentiation. In cancer patients, the SIRT1 agonist metformin reduced the frequency of Th17 cells and STAT3 acetylation levels. Altogether, these data underscore that SIRT1 activation impedes Th17 cell differentiation and thereby limits tumor growth and suggest that SIRT1 activators may directly target IL-17A functions.


Subject(s)
STAT3 Transcription Factor/metabolism , Sirtuin 1/metabolism , Acetylation/drug effects , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Carbazoles/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Metformin/pharmacology , Metformin/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , STAT3 Transcription Factor/agonists , Sirtuin 1/chemistry , Th17 Cells/cytology , Th17 Cells/drug effects , Th17 Cells/metabolism , Transplantation, Heterologous
5.
Am J Physiol Endocrinol Metab ; 313(1): E26-E36, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28325733

ABSTRACT

Evidence has accumulated that obesity-related metabolic dysregulation is associated with overactivation of the endocannabinoid system (ECS), which involves cannabinoid receptor 1 (CB1R), in peripheral tissues, including adipose tissue (AT). The functional consequences of CB1R activation on AT metabolism remain unclear. Since excess fat mobilization is considered an important primary event contributing to the onset of insulin resistance, we combined in vivo and in vitro experiments to investigate whether activation of ECS could alter the lipolytic rate. For this purpose, the appearance of plasma glycerol was measured in wild-type and CB1R-/- mice after acute anandamide administration or inhibition of endocannabinoid degradation by JZL195. Additional experiments were conducted on rat AT explants to evaluate the direct consequences of ECS activation on glycerol release and signaling pathways. Treatments stimulated glycerol release in mice fasted for 6 h and injected with glucose but not in 24-h fasted mice or in CB1R-/-, suggesting that the effect was dependent on plasma insulin levels and mediated by CB1R. We concomitantly observed that Akt cascade activity was decreased, indicating an alteration of the antilipolytic action of insulin. Similar results were obtained with tissue explants exposed to anandamide, thus identifying CB1R of AT as a major target. This study indicates the existence of a functional interaction between CB1R and lipolysis regulation in AT. Further investigation is needed to test if the elevation of ECS tone encountered in obesity is associated with excess fat mobilization contributing to ectopic fat deposition and related metabolic disorders.


Subject(s)
Adipose Tissue/physiology , Endocannabinoids/metabolism , Insulin Resistance/physiology , Insulin/blood , Lipolysis/physiology , Receptor, Cannabinoid, CB1/metabolism , Animals , Fatty Acids, Nonesterified/biosynthesis , Hydrolysis , Male , Mice , Mice, Inbred C57BL , Up-Regulation/physiology
6.
Oncoimmunology ; 5(1): e1055444, 2016.
Article in English | MEDLINE | ID: mdl-26942062

ABSTRACT

Th17 cells contribute to the development of some autoimmune and allergic diseases by driving tissue inflammation. However, the function of Th17 cells during cancer progression remains controversial. Here, we show that human memory CD25high Th17 cells suppress T cell immunity in breast cancer. Ectonucleotidase-expressing Th17 cells accumulated in breast cancer tumors and suppressed CD4+ and CD8+ T cell activation. These cells expressed both Rorγt and Foxp3 genes and secreted Th17 related cytokines. We further found that CD39 ectonucleotisase expression on tumor-infiltrating Th17 cells was driven by TGF-ßand IL-6. Finally, immunohistochemical analysis of localized breast cancer revealed that high-tumor infiltration by IL-17+ cells was associated with a poor clinical outcome and impeded the favorable effect of high CD8+ infiltration. Altogether, these findings suggest that intratumoral Th17 cells compromise anticancer immune responses in breast cancer patients.

8.
Nat Immunol ; 16(8): 859-70, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26098997

ABSTRACT

The receptor NLRP3 is involved in the formation of the NLRP3 inflammasome that activates caspase-1 and mediates the release of interleukin 1ß (IL-1ß) and IL-18. Whether NLRP3 can shape immunological function independently of inflammasomes is unclear. We found that NLRP3 expression in CD4(+) T cells specifically supported a T helper type 2 (TH2) transcriptional program in a cell-intrinsic manner. NLRP3, but not the inflammasome adaptor ASC or caspase-1, positively regulated a TH2 program. In TH2 cells, NLRP3 bound the Il4 promoter and transactivated it in conjunction with the transcription factor IRF4. Nlrp3-deficient TH2 cells supported melanoma tumor growth in an IL-4-dependent manner and also promoted asthma-like symptoms. Our results demonstrate the ability of NLRP3 to act as a key transcription factor in TH2 differentiation.


Subject(s)
Carrier Proteins/immunology , Cell Differentiation/immunology , Th2 Cells/immunology , Trans-Activators/immunology , Animals , Asthma/genetics , Asthma/immunology , Asthma/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , NLR Family, Pyrin Domain-Containing 3 Protein , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Protein Binding/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , Th2 Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
9.
Nat Immunol ; 15(8): 758-66, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24973819

ABSTRACT

The TH9 subset of helper T cells was initially shown to contribute to the induction of autoimmune and allergic diseases, but subsequent evidence has suggested that these cells also exert antitumor activities. However, the molecular events that account for their effector properties are elusive. Here we found that the transcription factor IRF1 enhanced the effector function of TH9 cells and dictated their anticancer properties. Under TH9-skewing conditions, interleukin 1ß (IL-1ß) induced phosphorylation of the transcription factor STAT1 and subsequent expression of IRF1, which bound to the promoters of Il9 and Il21 and enhanced secretion of the cytokines IL-9 and IL-21 from TH9 cells. Furthermore, IL-1ß-induced TH9 cells exerted potent anticancer functions in an IRF1- and IL-21-dependent manner. Our findings thus identify IRF1 as a target for controlling the function of TH9 cells.


Subject(s)
Interferon Regulatory Factor-1/immunology , Interleukins/immunology , Melanoma, Experimental/immunology , T-Lymphocytes, Helper-Inducer/immunology , 3T3 Cells , Animals , Base Sequence , Cell Line , Female , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Interferon Regulatory Factor-1/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/antagonists & inhibitors , Interleukin-10/immunology , Interleukin-9/genetics , Interleukin-9/immunology , Interleukin-9/metabolism , Interleukins/genetics , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Phosphorylation/immunology , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/genetics , RNA Interference , RNA, Small Interfering , STAT1 Transcription Factor/immunology , Sequence Analysis, RNA , T-Lymphocytes, Helper-Inducer/metabolism
10.
JAKSTAT ; 2(1): e23010, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-24058791

ABSTRACT

Cancer growth is controlled by cancer cells (cell intrinsic phenomenon), but also by the immune cells in the tumor microenvironment (cell extrinsic phenomenon). Thus cancer progression is mediated by the activation of transcription programs responsible for cancer cell proliferation, but also induced proliferation/activation of immunosuppressive cells such as Th17, Treg or myeloid derived suppressor cells (MDSCs). One of the key transcription factors involved in these pathways is the signal transducer and activator of transcription 3 (STAT3). In this review we will focus on STAT3 activation in immune cells, and how it impacts on tumor progression.

12.
PLoS One ; 8(6): e65181, 2013.
Article in English | MEDLINE | ID: mdl-23762310

ABSTRACT

Bleomycin (BLM) is an anticancer drug currently used for the treatment of testis cancer and Hodgkin lymphoma. This drug triggers cancer cell death via its capacity to generate radical oxygen species (ROS). However, the putative contribution of anticancer immune responses to the efficacy of BLM has not been evaluated. We make here the observation that BLM induces immunogenic cell death. In particular, BLM is able to induce ROS-mediated reticulum stress and autophagy, which result in the surface exposure of chaperones, including calreticulin and ERp57, and liberation of HMBG1 and ATP. BLM induces anti-tumor immunity which relies on calreticulin, CD8(+) T cells and interferon-γ. We also find that, in addition to its capacity to trigger immunogenic cell death, BLM induces expansion of Foxp3+ regulatory T (Treg) cells via its capacity to induce transforming growth factor beta (TGFß) secretion by tumor cells. Accordingly, Treg cells or TGFß depletion dramatically potentiates the antitumor effect of BLM. We conclude that BLM induces both anti-tumor CD8(+) T cell response and a counteracting Treg proliferation. In the future, TGFß or Treg inhibition during BLM treatment could greatly enhance BLM anti-tumor efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , Bleomycin/pharmacology , Immunity/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Immunity, Innate/drug effects , Mice , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/metabolism
13.
Cancer Res ; 73(12): 3578-90, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23619236

ABSTRACT

Activation of the transcription factor PPARγ by the n-3 fatty acid docosahexaenoic acid (DHA) is implicated in controlling proinflammatory cytokine secretion, but the intracellular signaling pathways engaged by PPARγ are incompletely characterized. Here, we identify the adapter-encoding gene SOCS3 as a critical transcriptional target of PPARγ. SOCS3 promoter binding and gene transactivation by PPARγ was associated with a repression in differentiation of proinflammatory T-helper (TH)17 cells. Accordingly, TH17 cells induced in vitro displayed increased SOCS3 expression and diminished capacity to produce interleukin (IL)-17 following activation of PPARγ by DHA. Furthermore, naïve CD4 T cells derived from mice fed a DHA-enriched diet displayed less capability to differentiate into TH17 cells. In two different mouse models of cancer, DHA prevented tumor outgrowth and angiogenesis in an IL-17-dependent manner. Altogether, our results uncover a novel molecular pathway by which PPARγ-induced SOCS3 expression prevents IL-17-mediated cancer growth.


Subject(s)
Mammary Neoplasms, Experimental/genetics , PPAR gamma/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Transcriptional Activation , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Diet , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/pharmacology , Female , Interleukin-17/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , PPAR gamma/agonists , PPAR gamma/metabolism , Promoter Regions, Genetic/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Th17 Cells/drug effects , Th17 Cells/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...