Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Nature ; 621(7980): 849-856, 2023 Sep.
Article En | MEDLINE | ID: mdl-37730993

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.


CD8-Positive T-Lymphocytes , Lymphocyte Activation , Cell Differentiation/genetics , Citric Acid Cycle , Oxidative Phosphorylation , Immunologic Memory/genetics
2.
Sci Immunol ; 8(87): eadf7579, 2023 09 29.
Article En | MEDLINE | ID: mdl-37738363

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.


COVID-19 , Parkinson Disease , Humans , CD8-Positive T-Lymphocytes , Memory T Cells , Mitophagy , Cell Death
3.
Cell Metab ; 35(1): 118-133.e7, 2023 01 03.
Article En | MEDLINE | ID: mdl-36599297

Immunoediting sculpts immunogenicity and thwarts host anti-tumor responses in tumor cells during tumorigenesis; however, it remains unknown whether metabolic programming of tumor cells can be guided by immunosurveillance. Here, we report that T cell-mediated immunosurveillance in early-stage tumorigenesis instructs c-Myc upregulation and metabolic reprogramming in tumor cells. This previously unexplored tumor-immune interaction is controlled by non-canonical interferon gamma (IFNγ)-STAT3 signaling and supports tumor immune evasion. Our findings uncover that immunoediting instructs deregulated bioenergetic programs in tumor cells to empower them to disarm the T cell-mediated immunosurveillance by imposing metabolic tug-of-war between tumor and infiltrating T cells and forming the suppressive tumor microenvironment.


Immune Evasion , Neoplasms , Humans , Neoplasms/pathology , Interferon-gamma/metabolism , T-Lymphocytes/metabolism , Carcinogenesis , Cell Transformation, Neoplastic , Tumor Microenvironment
4.
Nat Cell Biol ; 24(11): 1574-1583, 2022 11.
Article En | MEDLINE | ID: mdl-36229606

The metabolically hostile tumour microenvironment imposes barriers to tumour-infiltrating immune cells and impedes durable clinical remission following immunotherapy. Metabolic communication between cancer cells and their neighbouring immune cells could determine the amplitude and type of immune responses, highlighting a potential involvement of metabolic crosstalk in immune surveillance and escape. In this Review, we explore tumour-immune metabolic crosstalk and discuss potential nutrient-limiting strategies that favour anti-tumour immune responses.


Neoplasms , Tumor Microenvironment , Humans , Immunity , Immunotherapy , Neoplasms/metabolism
5.
Cancer Discov ; 12(7): 1615-1616, 2022 07 06.
Article En | MEDLINE | ID: mdl-35791695

SUMMARY: In this issue, Hu and colleagues unveil that IFNα administration combined with anti-PD-1 therapy can potentiate murine and human CD8+ T-cell antitumor response in hepatocellular carcinoma, highlighting a novel therapeutic strategy for hepatocellular carcinoma. See related article by Hu et al., p. 1718 (6) .


Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/drug therapy , Glucose , Humans , Immune Checkpoint Inhibitors , Immunotherapy , Liver Neoplasms/drug therapy , Mice , Tumor Microenvironment
6.
FEBS J ; 289(3): 634-646, 2022 02.
Article En | MEDLINE | ID: mdl-33570771

Viral infections are very common, and in most cases, the virus is well controlled and eliminated by the immune system. Nevertheless, in some cases, damage of the host tissue inflicted by the virus itself or by the elicited immune response may result in severe disease courses. Thus, regulatory mechanisms are necessary to control virus-induced and immune pathology. This ensures immune responses are elicited in a potent but controlled manner. In this review, we will outline how immune regulation may contribute to this process. We focus on regulatory T cells and co-inhibitory receptors and outline how these two regulatory immune components allow for and may even promote potent but not pathologic immune responses. By enabling a balanced immune response, regulatory mechanisms can thus contribute to pathogen control as well as tissue and host protection.


Host-Pathogen Interactions/genetics , Immune System/immunology , Immunity, Innate/genetics , Virus Diseases/genetics , Antiviral Agents/therapeutic use , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/immunology , T-Lymphocytes, Regulatory/immunology , Virus Diseases/drug therapy , Virus Diseases/immunology , Virus Diseases/virology
7.
Cell Metab ; 33(6): 1071-1072, 2021 06 01.
Article En | MEDLINE | ID: mdl-34077714

Tumor cells utilize glucose to engage in aerobic glycolysis, fulfilling their metabolic demands for extensive proliferation. A recent study in Nature discovers that tumor-infiltrating myeloid cells exhibit a superior glucose uptake capacity over tumor cells, which present enhanced glutamine metabolism, suggesting that nutrient partitioning in the TME might be more complex than previously thought.


Glycolysis , Neoplasms , Cell Proliferation , Citric Acid Cycle , Glucose , Glutamine/metabolism , Humans
8.
Nat Commun ; 11(1): 1522, 2020 03 23.
Article En | MEDLINE | ID: mdl-32251280

Foxp3+ regulatory T (Treg) cells are essential for maintaining peripheral tolerance and preventing autoimmunity. While genetic factors may predispose for autoimmunity, additional environmental triggers, such as viral infections, are usually required to initiate the onset of disease. Here, we show that viral infection with LCMV results in type I IFN-dependent Treg cell loss that is rapidly compensated by the conversion and expansion of Vß5+ conventional T cells into iTreg cells. Using Vß5-deficient mice, we show that these Vß5+ iTreg cells are dispensable for limiting anti-viral immunity. Rather, the delayed replenishment of Treg cells in Vß5-deficient mice compromises suppression of microbiota-dependent activation of CD8+ T cells, resulting in colitis. Importantly, recovery from clinical symptoms in IBD patients is marked by expansion of the corresponding Vß2+ Treg population in humans. Collectively, we provide a link between a viral trigger and an impaired Treg cell compartment resulting in the initiation of immune pathology.


Arenaviridae Infections/immunology , Autoimmunity , CD8-Positive T-Lymphocytes/immunology , Colitis/immunology , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Arenaviridae Infections/complications , Cell Line , Colitis/microbiology , Colon/immunology , Colon/microbiology , Forkhead Transcription Factors/metabolism , Gastrointestinal Microbiome/immunology , Healthy Volunteers , Host Microbial Interactions/immunology , Humans , Interferon Type I/immunology , Interferon Type I/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Lymphocyte Activation/genetics , Male , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes, Regulatory/metabolism
9.
Immun Ageing ; 15: 27, 2018.
Article En | MEDLINE | ID: mdl-30455721

BACKGROUND: Patients with end-stage renal disease (ESRD) exhibit a premature aging phenotype of the immune system. Nevertheless, the etiology and impact of these changes in ESRD patients remain unknown. RESULTS: Compared to healthy individuals, ESRD patients exhibit accelerated immunosenescence in both T cell and monocyte compartments, characterized by a dramatic reduction in naïve CD4+ and CD8+ T cell numbers but increase in CD8+ TEMRA cell and proinflammatory monocyte numbers. Notably, within ESRD patients, aging-related immune changes positively correlated not only with increasing age but also with longer dialysis vintage. In multivariable-adjusted logistic regression models, the combination of high terminally differentiated CD8+ T cell level and high intermediate monocyte level, as a composite predictive immunophenotype, was independently associated with prevalent coronary artery disease as well as cardiovascular disease, after adjustment for age, sex, systemic inflammation and presence of diabetes. Levels of terminally differentiated CD8+ T cells also positively correlated with the level of uremic toxin p-cresyl sulfate. CONCLUSIONS: Aging-associated adaptive and innate immune changes are aggravated in ESRD and are associated with cardiovascular diseases. For the first time, our study demonstrates the potential link between immunosenescence in ESRD and duration of exposure to the uremic milieu.

10.
J Immunol ; 199(2): 467-476, 2017 07 15.
Article En | MEDLINE | ID: mdl-28600287

Human oral squamous cell carcinoma (OSCC) constitutes an inflammatory microenvironment enriched with chemokines such as CCL20, which promote cancer cell invasion and tumor progression. We found that in OSCC there is a correlation between the expression of CCL20 and FOXP3 mRNA. Therefore, we hypothesized that OSCC may favor the recruitment and retention of regulatory T (Treg) cells that express the CCL20 receptor, CCR6. Interestingly, most (∼60%) peripheral blood Treg cells express CCR6, and CCR6+ Treg cells exhibit an activated effector/memory phenotype. In contrast, a significant portion (>30%) of CCR6- Treg cells were found to be CD45RA+ naive Treg cells. Compared to CCR6- naive or memory Treg cells, CCR6+ Treg cells exhibit stronger suppressive activity and display higher FOXP3 expression along with lower methylation at the Treg-specific demethylated region of the FOXP3 gene. This predominance of CCR6+ Treg cells was also found in the draining lymph nodes and tumor-infiltrating lymphocytes of OSCC patients with early or late clinical staging. Moreover, CCR6+ Treg cells isolated from tumor-infiltrating lymphocytes or draining lymph nodes maintained similar phenotypic and suppressive characteristics ex vivo as did their counterparts isolated from peripheral blood. These results suggest that CCR6 marks activated effector or memory Treg phenotypes with superior suppressive activity in humans.


Carcinoma, Squamous Cell/immunology , Mouth Neoplasms/immunology , Receptors, CCR6/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Carcinoma, Squamous Cell/pathology , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Humans , Immunologic Memory , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Methylation , Middle Aged , Receptors, CCR6/deficiency , Receptors, CCR6/genetics , T-Lymphocytes, Regulatory/physiology
...