Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Nat Immunol ; 22(6): 746-756, 2021 06.
Article in English | MEDLINE | ID: mdl-34031618

ABSTRACT

T cell exhaustion presents one of the major hurdles to cancer immunotherapy. Among exhausted CD8+ tumor-infiltrating lymphocytes, the terminally exhausted subset contributes directly to tumor cell killing owing to its cytotoxic effector function. However, this subset does not respond to immune checkpoint blockades and is difficult to be reinvigorated with restored proliferative capacity. Here, we show that a half-life-extended interleukin-10-Fc fusion protein directly and potently enhanced expansion and effector function of terminally exhausted CD8+ tumor-infiltrating lymphocytes by promoting oxidative phosphorylation, a process that was independent of the progenitor exhausted T cells. Interleukin-10-Fc was a safe and highly efficient metabolic intervention that synergized with adoptive T cell transfer immunotherapy, leading to eradication of established solid tumors and durable cures in the majority of treated mice. These findings show that metabolic reprogramming by upregulating mitochondrial pyruvate carrier-dependent oxidative phosphorylation can revitalize terminally exhausted T cells and enhance the response to cancer immunotherapy.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-10/pharmacology , Neoplasms/therapy , Oxidative Phosphorylation/drug effects , T-Lymphocytes, Cytotoxic/drug effects , Animals , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Cell Line, Tumor , Combined Modality Therapy/methods , Disease Models, Animal , Drug Synergism , Female , HEK293 Cells , Half-Life , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin Fc Fragments/therapeutic use , Interleukin-10/therapeutic use , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Neoplasms/immunology , Neoplasms/pathology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Interleukin-10/metabolism , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
2.
Nature ; 621(7980): 849-856, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37730993

ABSTRACT

Protective immunity against pathogens or cancer is mediated by the activation and clonal expansion of antigen-specific naive T cells into effector T cells. To sustain their rapid proliferation and effector functions, naive T cells switch their quiescent metabolism to an anabolic metabolism through increased levels of aerobic glycolysis, but also through mitochondrial metabolism and oxidative phosphorylation, generating energy and signalling molecules1-3. However, how that metabolic rewiring drives and defines the differentiation of T cells remains unclear. Here we show that proliferating effector CD8+ T cells reductively carboxylate glutamine through the mitochondrial enzyme isocitrate dehydrogenase 2 (IDH2). Notably, deletion of the gene encoding IDH2 does not impair the proliferation of T cells nor their effector function, but promotes the differentiation of memory CD8+ T cells. Accordingly, inhibiting IDH2 during ex vivo manufacturing of chimeric antigen receptor (CAR) T cells induces features of memory T cells and enhances antitumour activity in melanoma, leukaemia and multiple myeloma. Mechanistically, inhibition of IDH2 activates compensating metabolic pathways that cause a disequilibrium in metabolites regulating histone-modifying enzymes, and this maintains chromatin accessibility at genes that are required for the differentiation of memory T cells. These findings show that reductive carboxylation in CD8+ T cells is dispensable for their effector response and proliferation, but that it mainly produces a pattern of metabolites that epigenetically locks CD8+ T cells into a terminal effector differentiation program. Blocking this metabolic route allows the increased formation of memory T cells, which could be exploited to optimize the therapeutic efficacy of CAR T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocyte Activation , Cell Differentiation/genetics , Citric Acid Cycle , Oxidative Phosphorylation , Immunologic Memory/genetics
3.
Eur J Immunol ; 52(4): 541-549, 2022 04.
Article in English | MEDLINE | ID: mdl-35253907

ABSTRACT

Cytotoxic CD8+ T cells are a key element of the adaptative immune system to protect the organism against infections and malignant cells. During their activation and response, T cells undergo different metabolic pathways to support their energetic needs according to their localization and function. However, it has also been recently appreciated that this metabolic reprogramming also directly supports T-cell lineage differentiation. Accordingly, metabolic deficiencies and prolonged stress exposure can impact T-cell differentiation and skew them into an exhausted state. Here, we review how metabolism defines CD8+ T-cell differentiation and function. Moreover, we cover the principal metabolic dysregulation that promotes the exhausted phenotype under tumor or chronic virus conditions. Finally, we summarize recent strategies to reprogram impaired metabolic pathways to promote CD8+ T-cell effector function and survival.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocyte Activation , Cell Differentiation , Lymphocyte Count , T-Lymphocytes, Cytotoxic
4.
Sci Immunol ; 9(98): eadn2717, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39178275

ABSTRACT

The formation of memory T cells is a fundamental feature of adaptative immunity, allowing the establishment of long-term protection against pathogens. Although emerging evidence suggests that metabolic reprogramming is crucial for memory T cell differentiation and survival, the underlying mechanisms that drive metabolic rewiring in memory T cells remain unclear. Here, we found that up-regulation of the nuclear receptor peroxisome proliferator-activated receptor ß/δ (PPARß/δ) instructs the metabolic reprogramming that occurs during the establishment of central memory CD8+ T cells. PPARß/δ-regulated changes included suppression of aerobic glycolysis and enhancement of oxidative metabolism and fatty acid oxidation. Mechanistically, exposure to interleukin-15 and expression of T cell factor 1 facilitated activation of the PPARß/δ pathway, counteracting apoptosis induced by antigen clearance and metabolic stress. Together, our findings indicate that PPARß/δ is a master metabolic regulator orchestrating a metabolic switch that may be favorable for T cell longevity.


Subject(s)
CD8-Positive T-Lymphocytes , Mice, Inbred C57BL , PPAR delta , PPAR-beta , Animals , PPAR-beta/metabolism , PPAR-beta/immunology , CD8-Positive T-Lymphocytes/immunology , PPAR delta/immunology , PPAR delta/metabolism , Mice , Immunologic Memory/immunology , Memory T Cells/immunology , Mice, Knockout , Interleukin-15/immunology , Interleukin-15/metabolism , Mice, Transgenic , Metabolic Reprogramming , Receptors, Cytoplasmic and Nuclear
5.
Sci Immunol ; 8(87): eadf7579, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37738363

ABSTRACT

Mitophagy, a central process guarding mitochondrial quality, is commonly impaired in human diseases such as Parkinson's disease, but its impact in adaptive immunity remains unclear. The differentiation and survival of memory CD8+ T cells rely on oxidative metabolism, a process that requires robust mitochondrial quality control. Here, we found that Parkinson's disease patients have a reduced frequency of CD8+ memory T cells compared with healthy donors and failed to form memory T cells upon vaccination against COVID-19, highlighting the importance of mitochondrial quality control for memory CD8+ T cell formation. We further uncovered that regulators of mitophagy, including Parkin and NIX, were up-regulated in response to interleukin-15 (IL-15) for supporting memory T cell formation. Mechanistically, Parkin suppressed VDAC1-dependent apoptosis in memory T cells. In contrast, NIX expression in T cells counteracted ferroptosis by preventing metabolic dysfunction resulting from impaired mitophagy. Together, our results indicate that the mitophagy machinery orchestrates survival and metabolic dynamics required for memory T cell formation, as well as highlight a deficit in T cell-mediated antiviral responses in Parkinson's disease patients.


Subject(s)
COVID-19 , Parkinson Disease , Humans , CD8-Positive T-Lymphocytes , Memory T Cells , Mitophagy , Cell Death
6.
Sci Immunol ; 7(73): eabq6783, 2022 07.
Article in English | MEDLINE | ID: mdl-35776801

ABSTRACT

Androgen signaling compromises CD8+ T cell effector functions and contributes to sex-biased outcomes in many forms of cancer (see the related Research Article by Kwon et al.).


Subject(s)
Androgens , Neoplasms , CD8-Positive T-Lymphocytes , Humans , Signal Transduction
7.
Sci Rep ; 9(1): 18429, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804543

ABSTRACT

Hydrophobic zeolites are nanoporous materials that are attracting an increasing interest, especially for catalysis, desalination, energy storage and biomedical applications. Nevertheless, a more profound understanding and control of water infiltration in their nanopores is still desirable to rationally design zeolite-based materials with tailored properties. In this work, both atomistic simulations and previous experimental data are employed to investigate water infiltration in hydrophobic MFI zeolites with different concentration of hydrophilic defects. Results show that limited concentrations of defects (e.g. 1%) induce a change in the shape of infiltration isotherms (from type-V to type-I), which denotes a sharp passage from typical hydrophobic to hydrophilic behavior. A correlation parametrized on both energy and geometric characteristics of the zeolite (infiltration model) is then adopted to interpolate the infiltration isotherms data by means of a limited number of physically-meaningful parameters. Finally, the infiltration model is combined with the water-zeolite interaction energy computed by simulations to correlate the water intrusion mechanism with the atomistic details of the zeolite crystal, such as defects concentration, distribution and hydrophilicity. The suggested methodology may allow a faster (more than one order of magnitude) and more systematic preliminary computational screening of innovative zeolite-based materials for energy storage, desalination and biomedical purposes.

8.
Nat Commun ; 7: 12762, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27694935

ABSTRACT

A comprehensive understanding of molecular transport within nanoporous materials remains elusive in a broad variety of engineering and biomedical applications. Here, experiments and atomistic simulations are synergically used to elucidate the non-trivial interplay between nanopore hydrophilicity and surface barriers on the overall water transport through zeolite crystals. At these nanometre-length scales, these results highlight the dominating effect of surface imperfections with reduced permeability on the overall water transport. A simple diffusion resistance model is shown to be sufficient to capture the effects of both intracrystalline and surface diffusion resistances, thus properly linking simulation to experimental evidence. This work suggests that future experimental work should focus on eliminating/overcoming these surface imperfections, which promise an order of magnitude improvement in permeability.

SELECTION OF CITATIONS
SEARCH DETAIL