Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Cell Rep ; 42(9): 113013, 2023 09 26.
Article En | MEDLINE | ID: mdl-37632752

2-Hydroxyglutarate (2HG) is a byproduct of the tricarboxylic acid (TCA) cycle and is readily detected in the tissues of healthy individuals. 2HG is found in two enantiomeric forms: S-2HG and R-2HG. Here, we investigate the differential roles of these two enantiomers in cluster of differentiation (CD)8+ T cell biology, where we find they have highly divergent effects on proliferation, differentiation, and T cell function. We show here an analysis of structural determinants that likely underlie these differential effects on specific α-ketoglutarate (αKG)-dependent enzymes. Treatment of CD8+ T cells with exogenous S-2HG, but not R-2HG, increased CD8+ T cell fitness in vivo and enhanced anti-tumor activity. These data show that S-2HG and R-2HG should be considered as two distinct and important actors in the regulation of T cell function.


Neoplasms , T-Lymphocytes, Cytotoxic , Humans , T-Lymphocytes, Cytotoxic/metabolism , CD8-Positive T-Lymphocytes/metabolism , Glutarates/metabolism , Neoplasms/metabolism , Isocitrate Dehydrogenase
2.
Cell Rep ; 42(5): 112419, 2023 05 30.
Article En | MEDLINE | ID: mdl-37074914

Potent T cell responses against infections and malignancies require a rapid yet tightly regulated production of toxic effector molecules. Their production level is defined by post-transcriptional events at 3' untranslated regions (3' UTRs). RNA binding proteins (RBPs) are key regulators in this process. With an RNA aptamer-based capture assay, we identify >130 RBPs interacting with IFNG, TNF, and IL2 3' UTRs in human T cells. RBP-RNA interactions show plasticity upon T cell activation. Furthermore, we uncover the intricate and time-dependent regulation of cytokine production by RBPs: whereas HuR supports early cytokine production, ZFP36L1, ATXN2L, and ZC3HAV1 dampen and shorten the production duration, each at different time points. Strikingly, even though ZFP36L1 deletion does not rescue the dysfunctional phenotype, tumor-infiltrating T cells produce more cytokines and cytotoxic molecules, resulting in superior anti-tumoral T cell responses. Our findings thus show that identifying RBP-RNA interactions reveals key modulators of T cell responses in health and disease.


Cytokines , T-Lymphocytes , Humans , T-Lymphocytes/metabolism , 3' Untranslated Regions , Cytokines/metabolism , RNA-Binding Proteins/metabolism , Butyrate Response Factor 1/genetics , Butyrate Response Factor 1/metabolism
3.
J Immunol ; 207(12): 2966-2975, 2021 12 15.
Article En | MEDLINE | ID: mdl-34782446

CD4+ T cells are key contributors in the induction of adaptive immune responses against pathogens. Even though CD4+ T cells are primarily classified as noncytotoxic helper T cells, it has become appreciated that a subset of CD4+ T cells is cytotoxic. However, tools to identify these cytotoxic CD4+ T cells are lacking. We recently showed that CD29 (integrin ß1, ITGB1) expression on human CD8+ T cells enriches for the most potent cytotoxic T cells. In this study, we questioned whether CD29 expression also associates with cytotoxic CD4+ T cells. We show that human peripheral blood-derived CD29hiCD4+ T cells display a cytotoxic gene expression profile, which closely resembles that of CD29hi cytotoxic CD8+ T cells. This CD29hi cytotoxic phenotype was observed ex vivo and was maintained in in vitro cultures. CD29 expression enriched for CD4+ T cells, which effectively produced the proinflammatory cytokines IFN-γ, IL-2, and TNF-α, and cytotoxic molecules. Lastly, CD29-expressing CD4+ T cells transduced with a MART1-specific TCR showed target cell killing in vitro. In conclusion, we demonstrate in this study that CD29 can be employed to enrich for cytotoxic human CD4+ T cells.


CD8-Positive T-Lymphocytes , Integrin beta1/metabolism , CD4-Positive T-Lymphocytes , Cytokines/metabolism , Humans , T-Lymphocytes, Cytotoxic , T-Lymphocytes, Helper-Inducer
4.
Proc Natl Acad Sci U S A ; 117(12): 6686-6696, 2020 03 24.
Article En | MEDLINE | ID: mdl-32161126

Cytotoxic CD8+ T cells can effectively kill target cells by producing cytokines, chemokines, and granzymes. Expression of these effector molecules is however highly divergent, and tools that identify and preselect CD8+ T cells with a cytotoxic expression profile are lacking. Human CD8+ T cells can be divided into IFN-γ- and IL-2-producing cells. Unbiased transcriptomics and proteomics analysis on cytokine-producing fixed CD8+ T cells revealed that IL-2+ cells produce helper cytokines, and that IFN-γ+ cells produce cytotoxic molecules. IFN-γ+ T cells expressed the surface marker CD29 already prior to stimulation. CD29 also marked T cells with cytotoxic gene expression from different tissues in single-cell RNA-sequencing data. Notably, CD29+ T cells maintained the cytotoxic phenotype during cell culture, suggesting a stable phenotype. Preselecting CD29-expressing MART1 TCR-engineered T cells potentiated the killing of target cells. We therefore propose that CD29 expression can help evaluate and select for potent therapeutic T cell products.


CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Integrin beta1/metabolism , Interferon-gamma/metabolism , Interleukin-2/metabolism , Melanoma/pathology , T-Lymphocytes, Cytotoxic/immunology , Gene Expression Profiling , Humans , Melanoma/immunology , Melanoma/metabolism , Prognosis , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Survival Rate
5.
Eur J Immunol ; 50(7): 949-958, 2020 07.
Article En | MEDLINE | ID: mdl-32112565

Long-lasting CD8+ T cell responses are critical in combatting infections and tumors. The pro-inflammatory cytokine IFN-γ is a key effector molecule herein. We recently showed that in murine T cells the production of IFN-γ is tightly regulated through adenylate uridylate-rich elements (AREs) that are located in the 3' untranslated region (UTR) of the Ifng mRNA molecule. Loss of AREs resulted in prolonged cytokine production in activated T cells and boosted anti-tumoral T cell responses. Here, we investigated whether these findings can be translated to primary human T cells. Utilizing CRISPR-Cas9 technology, we deleted the ARE region from the IFNG 3' UTR in peripheral blood-derived human T cells. Loss of AREs stabilized the IFNG mRNA in T cells and supported a higher proportion of IFN-γ protein-producing T cells. Importantly, combining MART-1 T cell receptor engineering with ARE-Del gene editing showed that this was also true for antigen-specific activation of T cells. MART-1-specific ARE-Del T cells showed higher percentages of IFN-γ producing T cells in response to MART-1 expressing tumor cells. Combined, our study reveals that ARE-mediated posttranscriptional regulation is conserved between murine and human T cells. Furthermore, generating antigen-specific ARE-Del T cells is feasible, a feature that could potentially be used for therapeutical purposes.


AU Rich Elements , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , CRISPR-Cas Systems , Cell Line, Tumor , Female , Humans , Interferon-gamma/genetics , MART-1 Antigen/genetics , MART-1 Antigen/immunology , Male , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
6.
Oncoimmunology ; 8(11): e1648170, 2019.
Article En | MEDLINE | ID: mdl-31646094

Non-small cell lung cancer (NSCLC) is the second most prevalent type of cancer. With the current treatment regimens, the mortality rate remains high. Therefore, better therapeutic approaches are necessary. NSCLCs generally possess many genetic mutations and are well infiltrated by T cells (TIL), making TIL therapy an attractive option. Here we show that T cells from treatment naive, stage I-IVa NSCLC tumors can effectively be isolated and expanded, with similar efficiency as from normal lung tissue. Importantly, 76% (13/17) of tested TIL products isolated from NSCLC lesions exhibited clear reactivity against primary tumor digests, with 0.5%-30% of T cells producing the inflammatory cytokine Interferon (IFN)-γ. Both CD4+ and CD8+ T cells displayed tumor reactivity. The cytokine production correlated well with CD137 and CD40L expression. Furthermore, almost half (7/17) of the TIL products contained polyfunctional T cells that produced Tumor Necrosis Factor (TNF)-α and/or IL-2 in addition to IFN-γ, a hallmark of effective immune responses. Tumor-reactivity in the TIL products correlated with high percentages of CD103+CD69+CD8+ T cell infiltrates in the tumor lesions, with PD-1hiCD4+ T cells, and with FoxP3+CD25+CD4+ regulatory T cell infiltrates, suggesting that the composition of T cell infiltrates may predict the level of tumor reactivity. In conclusion, the effective generation of tumor-reactive and polyfunctional TIL products implies that TIL therapy will be a successful treatment regimen for NSCLC patients.

7.
Oncoimmunology ; 8(2): e1532762, 2019.
Article En | MEDLINE | ID: mdl-30713785

Protective T cell responses against tumors require the production of Interferon gamma (IFN-γ). However, tumor-infiltrating T cells (TILs) gradually lose their capacity to produce IFN-γ and therefore fail to clear malignant cells. Dissecting the underlying mechanisms that block cytokine production is thus key for improving T cell products. Here we show that although TILs express substantial levels of Ifng mRNA, post-transcriptional mechanisms impede the production of IFN-γ protein due to loss of mRNA stability. CD28 triggering, but not PD1 blocking antibodies, effectively restores the stability of Ifng mRNA. Intriguingly, TILs devoid of AU-rich elements within the 3'untranslated region maintain stabilized Ifng mRNA and produce more IFN-γ protein than wild-type TILs. This sustained IFN-γ production translates into effective suppression of tumor outgrowth, which is almost exclusively mediated by direct effects on the tumor cells. We therefore conclude that post-transcriptional mechanisms could be modulated to potentiate effective T cell therapies in cancer.

8.
J Immunol ; 202(3): 714-723, 2019 02 01.
Article En | MEDLINE | ID: mdl-30578304

Optimal T cell activation requires Ag recognition through the TCR, engagement of costimulatory molecules, and cytokines. T cells can also directly recognize danger signals through the expression of TLRs. Whether TLR ligands have the capacity to provide costimulatory signals and enhance Ag-driven T cell activation is not well understood. In this study, we show that TLR2 and TLR7 ligands potently lower the Ag threshold for cytokine production in T cells. To investigate how TLR triggering supports cytokine production, we adapted the protocol for flow cytometry-based fluorescence in situ hybridization to mouse T cells. The simultaneous detection of cytokine mRNA and protein with single-cell resolution revealed that TLR triggering primarily drives de novo mRNA transcription. Ifng mRNA stabilization only occurs when the TCR is engaged. TLR2-, but not TLR7-mediated costimulation, can enhance mRNA stability at low Ag levels. Importantly, TLR2 costimulation increases the percentage of polyfunctional T cells, a hallmark of potent T cell responses. In conclusion, TLR-mediated costimulation effectively potentiates T cell effector function to suboptimal Ag levels.


CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Toll-Like Receptor 2/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Flow Cytometry , Humans , In Situ Hybridization, Fluorescence , Interferon-gamma/metabolism , Ligands , Melanoma, Experimental , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Specific Pathogen-Free Organisms , Toll-Like Receptor 2/genetics , Toll-Like Receptor 7/immunology
9.
Nat Immunol ; 19(8): 828-837, 2018 08.
Article En | MEDLINE | ID: mdl-29988089

Memory T cells are critical for the immune response to recurring infections. Their instantaneous reactivity to pathogens is empowered by the persistent expression of cytokine-encoding mRNAs. How the translation of proteins from pre-formed cytokine-encoding mRNAs is prevented in the absence of infection has remained unclear. Here we found that protein production in memory T cells was blocked via a 3' untranslated region (3' UTR)-mediated process. Germline deletion of AU-rich elements (AREs) in the Ifng-3' UTR led to chronic cytokine production in memory T cells. This aberrant protein production did not result from increased expression and/or half-life of the mRNA. Instead, AREs blocked the recruitment of cytokine-encoding mRNA to ribosomes; this block depended on the ARE-binding protein ZFP36L2. Thus, AREs mediate repression of translation in mouse and human memory T cells by preventing undesirable protein production from pre-formed cytokine-encoding mRNAs in the absence of infection.


3' Untranslated Regions/genetics , AU Rich Elements/genetics , Interferon-gamma/genetics , RNA, Messenger/genetics , T-Lymphocytes/immunology , Animals , Cells, Cultured , Epigenetic Repression , Immunologic Memory , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Chain Elongation, Translational , Ribosomes/metabolism , Tristetraprolin/genetics , Tristetraprolin/metabolism
10.
J Immunol ; 198(2): 962-970, 2017 01 15.
Article En | MEDLINE | ID: mdl-27927969

Effective T cell responses entail the coproduction of IFN-γ, TNF-α, and IL-2. Cytokine production is determined by transcriptional and posttranscriptional events. However, increased transcript levels do not always translate into protein production, and therefore simultaneous transcripts and protein measurement are essential for the appropriate analysis of T cell responses. In this study, we optimized flow cytometry-based fluorescence in situ hybridization (Flow-FISH) for IFN-γ to multicolor flow cytometry that allows for single-cell measurement of mRNA and protein levels. This high-throughput analysis detected Ag-specific human T cells of low frequency. We also employed Flow-FISH for single-tube analysis of IFN-γ transcript and protein profile to simultaneously study the responsiveness of different T cell subsets, that is, naive, effector, and memory T cells. Importantly, the simultaneous transcript and protein analysis of IFN-γ and of TNF-α and IL-2 revealed that T cell responses consist of two types: one subtype loses mRNA expression during activation, whereas the other maintains high transcript levels throughout stimulation. High cytokine transcript levels correlated with increased protein production. Intriguingly, this mRNAhi-expressing T cell population also produced higher levels of other cytokines, indicating that Flow-FISH helps identify the best cytokine producers during T cell activation. We conclude that Flow-FISH is a rapid, sensitive, and cost-effective method to determine the quality of T cell responses induced by, for instance, T cell vaccines.


Cytokines/analysis , Cytokines/biosynthesis , Flow Cytometry/methods , In Situ Hybridization, Fluorescence/methods , T-Lymphocytes/immunology , High-Throughput Screening Assays/methods , Humans , Polymerase Chain Reaction , RNA, Messenger/analysis , T-Lymphocytes/metabolism
11.
J Immunol ; 196(9): 3695-705, 2016 05 01.
Article En | MEDLINE | ID: mdl-27016606

CD8(+) T cells can respond to unrelated infections in an Ag-independent manner. This rapid innate-like immune response allows Ag-experienced T cells to alert other immune cell types to pathogenic intruders. In this study, we show that murine CD8(+) T cells can sense TLR2 and TLR7 ligands, resulting in rapid production of IFN-γ but not of TNF-α and IL-2. Importantly, Ag-experienced T cells activated by TLR ligands produce sufficient IFN-γ to augment the activation of macrophages. In contrast to Ag-specific reactivation, TLR-dependent production of IFN-γ by CD8(+) T cells relies exclusively on newly synthesized transcripts without inducing mRNA stability. Furthermore, transcription of IFN-γ upon TLR triggering depends on the activation of PI3K and serine-threonine kinase Akt, and protein synthesis relies on the activation of the mechanistic target of rapamycin. We next investigated which energy source drives the TLR-induced production of IFN-γ. Although Ag-specific cytokine production requires a glycolytic switch for optimal cytokine release, glucose availability does not alter the rate of IFN-γ production upon TLR-mediated activation. Rather, mitochondrial respiration provides sufficient energy for TLR-induced IFN-γ production. To our knowledge, this is the first report describing that TLR-mediated bystander activation elicits a helper phenotype of CD8(+) T cells. It induces a short boost of IFN-γ production that leads to a significant but limited activation of Ag-experienced CD8(+) T cells. This activation suffices to prime macrophages but keeps T cell responses limited to unrelated infections.


CD8-Positive T-Lymphocytes/immunology , Glycolysis , Interferon-gamma/biosynthesis , Lymphocyte Activation/immunology , Membrane Glycoproteins/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 7/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Class I Phosphatidylinositol 3-Kinases , Cytokines/biosynthesis , Cytokines/immunology , Immunity, Innate , Interferon-gamma/immunology , Interleukin-2/biosynthesis , Interleukin-2/immunology , Macrophage Activation , Mice , Mitochondria/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Respiration
12.
Cancer Immunol Immunother ; 64(10): 1241-50, 2015 Oct.
Article En | MEDLINE | ID: mdl-26105626

Targeted therapy with sunitinib, pazopanib or everolimus has improved treatment outcome for patients with metastatic renal cell carcinoma patients (RCC). However, despite considerable efforts in sequential or combined modalities, durable remissions are rare. Immunotherapy like cytokine therapy with interleukin-2, T cell checkpoint blockade or adoptive T cell therapies can achieve long-term benefit and even cure. This raises the question of whether combining targeted therapy with immunotherapy could also be an effective treatment option for RCC patients. Sunitinib, one of the most frequently administered therapeutics in RCC patients has been implicated in impairing T cell activation and proliferation in vitro. In this work, we addressed whether this notion holds true for expansion of tumor-infiltrating lymphocytes (TILs) in sunitinib-treated patients. We compared resected primary RCC tumor material of patients pretreated with sunitinib with resection specimen from sunitinib-naïve patients. We found improved TIL expansion from sunitinib-pretreated tumor digests. These TIL products contained more PD-1 expressing TIL, while the regulatory T cell infiltration was not altered. The improved TIL expansion was associated with reduced intratumoral myeloid-derived suppressor cell (MDSC) content. Depletion of MDSCs from sunitinib-naïve RCC tissue-digest improved TIL expansion, proving the functional relevance of the MDSC alteration by sunitinib. Our in vivo results do not support previous in vitro observations of sunitinib inhibiting T cell function, but do provide a possible rationale for the combination of sunitinib with immunotherapy.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/therapy , Immunotherapy , Kidney Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Molecular Targeted Therapy , Myeloid Cells/drug effects , T-Lymphocytes/immunology , Adult , Aged , Carcinoma, Renal Cell/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy , Everolimus , Female , Humans , Indazoles , Indoles/administration & dosage , Indoles/adverse effects , Interleukin-2/administration & dosage , Kidney Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Myeloid Cells/immunology , Neoplasm Metastasis , Programmed Cell Death 1 Receptor/metabolism , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrroles/administration & dosage , Pyrroles/adverse effects , Sirolimus/administration & dosage , Sirolimus/adverse effects , Sirolimus/analogs & derivatives , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Sunitinib , T-Lymphocytes/transplantation , Treatment Outcome
13.
Cancer Immunol Immunother ; 62(3): 503-15, 2013 Mar.
Article En | MEDLINE | ID: mdl-23001162

CD8(+) T cells undergoing homeostatic proliferation (HP) in a lymphopenic environment acquire a central memory-like phenotype (CD44(+) CD62L(+) Ly6c(+)). Such cells are readily functional in vitro, with a strong capacity to secrete IFNγ and IL-2 and to lyse target cells upon antigen recognition. In vivo, these memory-like T cells display potent anti-tumor reactivity. When addressing whether these remarkable properties were "acquired" or dependent on sustained HP, we observed, for the first time, that memory-like T cells retained full anti-tumor functions even when removed from their lymphopenic environment and retransferred into non-lymphopenic P14/Rag2(-/-) recipients (where HP is prevented). Moreover, memory-like T cells were superior to in vitro expanded effector T cells. We next sought to determine the conditions required to reproduce such a potent phenotype in vitro, in order to obtain optimal cells for adoptive cell transfer therapy. Assessing ex vivo lymph node cultures, dendritic cells, fibroblastic reticular cells, and HP-associated cytokines, we found that stimulation of naïve T cells with anti-CD3/CD28 beads and IL-15 (IL-7 was dispensable) led to the generation of memory-like T cell with a similar phenotype. Both in vitro and in vivo memory-like T cells retained the capacity to efficiently control tumor growth in non-lymphopenic hosts upon adoptive cell transfer. A similar phenotype could be imparted to human peripheral blood leukocytes with comparable culture conditions. Our data reinforce the idea that in vitro-generated memory-like T cells could benefit adoptive cell transfer therapies.


CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Immunotherapy, Adoptive/methods , Neoplasms/immunology , Neoplasms/therapy , Animals , CD28 Antigens/immunology , CD3 Complex/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Homeostasis/immunology , Humans , Lymphocyte Activation/immunology , Lymphopenia/immunology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , T-Lymphocyte Subsets/immunology
...