Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 30
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Nature ; 631(8022): 867-875, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38987588

RÉSUMÉ

Chronic hepatitis B virus (HBV) infection affects 300 million patients worldwide1,2, in whom virus-specific CD8 T cells by still ill-defined mechanisms lose their function and cannot eliminate HBV-infected hepatocytes3-7. Here we demonstrate that a liver immune rheostat renders virus-specific CD8 T cells refractory to activation and leads to their loss of effector functions. In preclinical models of persistent infection with hepatotropic viruses such as HBV, dysfunctional virus-specific CXCR6+ CD8 T cells accumulated in the liver and, as a characteristic hallmark, showed enhanced transcriptional activity of cAMP-responsive element modulator (CREM) distinct from T cell exhaustion. In patients with chronic hepatitis B, circulating and intrahepatic HBV-specific CXCR6+ CD8 T cells with enhanced CREM expression and transcriptional activity were detected at a frequency of 12-22% of HBV-specific CD8 T cells. Knocking out the inhibitory CREM/ICER isoform in T cells, however, failed to rescue T cell immunity. This indicates that CREM activity was a consequence, rather than the cause, of loss in T cell function, further supported by the observation of enhanced phosphorylation of protein kinase A (PKA) which is upstream of CREM. Indeed, we found that enhanced cAMP-PKA-signalling from increased T cell adenylyl cyclase activity augmented CREM activity and curbed T cell activation and effector function in persistent hepatic infection. Mechanistically, CD8 T cells recognizing their antigen on hepatocytes established close and extensive contact with liver sinusoidal endothelial cells, thereby enhancing adenylyl cyclase-cAMP-PKA signalling in T cells. In these hepatic CD8 T cells, which recognize their antigen on hepatocytes, phosphorylation of key signalling kinases of the T cell receptor signalling pathway was impaired, which rendered them refractory to activation. Thus, close contact with liver sinusoidal endothelial cells curbs the activation and effector function of HBV-specific CD8 T cells that target hepatocytes expressing viral antigens by means of the adenylyl cyclase-cAMP-PKA axis in an immune rheostat-like fashion.


Sujet(s)
Lymphocytes T CD8+ , Hépatite B chronique , Foie , Animaux , Humains , Mâle , Souris , Lymphocytes T CD8+/enzymologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lymphocytes T CD8+/anatomopathologie , Modulateur de l'élément de réponse à l'AMP cyclique/métabolisme , Cyclic AMP-Dependent Protein Kinases/métabolisme , Virus de l'hépatite B/immunologie , Hépatite B chronique/immunologie , Hépatite B chronique/virologie , Hépatocytes/immunologie , Hépatocytes/virologie , Foie/immunologie , Foie/virologie , Phosphorylation , Transduction du signal , Activation des lymphocytes
2.
Redox Biol ; 75: 103211, 2024 09.
Article de Anglais | MEDLINE | ID: mdl-38908072

RÉSUMÉ

Ferroptosis is a pervasive non-apoptotic form of cell death highly relevant in various degenerative diseases and malignancies. The hallmark of ferroptosis is uncontrolled and overwhelming peroxidation of polyunsaturated fatty acids contained in membrane phospholipids, which eventually leads to rupture of the plasma membrane. Ferroptosis is unique in that it is essentially a spontaneous, uncatalyzed chemical process based on perturbed iron and redox homeostasis contributing to the cell death process, but that it is nonetheless modulated by many metabolic nodes that impinge on the cells' susceptibility to ferroptosis. Among the various nodes affecting ferroptosis sensitivity, several have emerged as promising candidates for pharmacological intervention, rendering ferroptosis-related proteins attractive targets for the treatment of numerous currently incurable diseases. Herein, the current members of a Germany-wide research consortium focusing on ferroptosis research, as well as key external experts in ferroptosis who have made seminal contributions to this rapidly growing and exciting field of research, have gathered to provide a comprehensive, state-of-the-art review on ferroptosis. Specific topics include: basic mechanisms, in vivo relevance, specialized methodologies, chemical and pharmacological tools, and the potential contribution of ferroptosis to disease etiopathology and progression. We hope that this article will not only provide established scientists and newcomers to the field with an overview of the multiple facets of ferroptosis, but also encourage additional efforts to characterize further molecular pathways modulating ferroptosis, with the ultimate goal to develop novel pharmacotherapies to tackle the various diseases associated with - or caused by - ferroptosis.


Sujet(s)
Ferroptose , Humains , Animaux , Fer/métabolisme , Tumeurs/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Peroxydation lipidique , Oxydoréduction , Prédisposition aux maladies
3.
Nature ; 629(8011): 426-434, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38658764

RÉSUMÉ

Expansion of antigen-experienced CD8+ T cells is critical for the success of tumour-infiltrating lymphocyte (TIL)-adoptive cell therapy (ACT) in patients with cancer1. Interleukin-2 (IL-2) acts as a key regulator of CD8+ cytotoxic T lymphocyte functions by promoting expansion and cytotoxic capability2,3. Therefore, it is essential to comprehend mechanistic barriers to IL-2 sensing in the tumour microenvironment to implement strategies to reinvigorate IL-2 responsiveness and T cell antitumour responses. Here we report that prostaglandin E2 (PGE2), a known negative regulator of immune response in the tumour microenvironment4,5, is present at high concentrations in tumour tissue from patients and leads to impaired IL-2 sensing in human CD8+ TILs via the PGE2 receptors EP2 and EP4. Mechanistically, PGE2 inhibits IL-2 sensing in TILs by downregulating the IL-2Rγc chain, resulting in defective assembly of IL-2Rß-IL2Rγc membrane dimers. This results in impaired IL-2-mTOR adaptation and PGC1α transcriptional repression, causing oxidative stress and ferroptotic cell death in tumour-reactive TILs. Inhibition of PGE2 signalling to EP2 and EP4 during TIL expansion for ACT resulted in increased IL-2 sensing, leading to enhanced proliferation of tumour-reactive TILs and enhanced tumour control once the cells were transferred in vivo. Our study reveals fundamental features that underlie impairment of human TILs mediated by PGE2 in the tumour microenvironment. These findings have therapeutic implications for cancer immunotherapy and cell therapy, and enable the development of targeted strategies to enhance IL-2 sensing and amplify the IL-2 response in TILs, thereby promoting the expansion of effector T cells with enhanced therapeutic potential.


Sujet(s)
Lymphocytes T CD8+ , Prolifération cellulaire , Dinoprostone , Interleukine-2 , Lymphocytes TIL , Mitochondries , Transduction du signal , Animaux , Humains , Souris , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Dinoprostone/métabolisme , Régulation négative , Ferroptose , Sous-unité gamma commune aux récepteurs des interleukines/biosynthèse , Sous-unité gamma commune aux récepteurs des interleukines/déficit , Sous-unité gamma commune aux récepteurs des interleukines/métabolisme , Interleukine-2/antagonistes et inhibiteurs , Interleukine-2/immunologie , Interleukine-2/métabolisme , Sous-unité bêta du récepteur à l'interleukine-2/métabolisme , Lymphocytes TIL/cytologie , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Mitochondries/métabolisme , Stress oxydatif , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Sous-type EP2 des récepteurs des prostaglandines E/antagonistes et inhibiteurs , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/antagonistes et inhibiteurs , Sérine-thréonine kinases TOR/métabolisme , Microenvironnement tumoral/immunologie
4.
Nature ; 629(8011): 417-425, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38658748

RÉSUMÉ

Cancer-specific TCF1+ stem-like CD8+ T cells can drive protective anticancer immunity through expansion and effector cell differentiation1-4; however, this response is dysfunctional in tumours. Current cancer immunotherapies2,5-9 can promote anticancer responses through TCF1+ stem-like CD8+ T cells in some but not all patients. This variation points towards currently ill-defined mechanisms that limit TCF1+CD8+ T cell-mediated anticancer immunity. Here we demonstrate that tumour-derived prostaglandin E2 (PGE2) restricts the proliferative expansion and effector differentiation of TCF1+CD8+ T cells within tumours, which promotes cancer immune escape. PGE2 does not affect the priming of TCF1+CD8+ T cells in draining lymph nodes. PGE2 acts through EP2 and EP4 (EP2/EP4) receptor signalling in CD8+ T cells to limit the intratumoural generation of early and late effector T cell populations that originate from TCF1+ tumour-infiltrating CD8+ T lymphocytes (TILs). Ablation of EP2/EP4 signalling in cancer-specific CD8+ T cells rescues their expansion and effector differentiation within tumours and leads to tumour elimination in multiple mouse cancer models. Mechanistically, suppression of the interleukin-2 (IL-2) signalling pathway underlies the PGE2-mediated inhibition of TCF1+ TIL responses. Altogether, we uncover a key mechanism that restricts the IL-2 responsiveness of TCF1+ TILs and prevents anticancer T cell responses that originate from these cells. This study identifies the PGE2-EP2/EP4 axis as a molecular target to restore IL-2 responsiveness in anticancer TILs to achieve cancer immune control.


Sujet(s)
Lymphocytes T CD8+ , Prolifération cellulaire , Dinoprostone , Lymphocytes TIL , Tumeurs , Cellules souches , Échappement de la tumeur à la surveillance immunitaire , Animaux , Femelle , Humains , Mâle , Souris , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Différenciation cellulaire , Lignée cellulaire tumorale , Dinoprostone/métabolisme , Modèles animaux de maladie humaine , Facteur nucléaire hépatocytaire HNF-1 alpha/métabolisme , Interleukine-2 , Noeuds lymphatiques/cytologie , Noeuds lymphatiques/immunologie , Lymphocytes TIL/cytologie , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Souris de lignée C57BL , Tumeurs/immunologie , Tumeurs/prévention et contrôle , Sous-type EP2 des récepteurs des prostaglandines E/déficit , Sous-type EP2 des récepteurs des prostaglandines E/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/déficit , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Transduction du signal , Cellules souches/cytologie , Cellules souches/immunologie , Cellules souches/métabolisme , Échappement de la tumeur à la surveillance immunitaire/immunologie
5.
Nature ; 627(8003): 407-415, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38383779

RÉSUMÉ

Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen1. The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP42. However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.


Sujet(s)
Aquaporine-4 , Autoanticorps , Autoantigènes , Lymphocytes B , Tolérance immunitaire , Neuromyélite optique , Animaux , Humains , Souris , , Aquaporine-4/déficit , Aquaporine-4/génétique , Aquaporine-4/immunologie , Aquaporine-4/métabolisme , Autoanticorps/immunologie , Autoantigènes/immunologie , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Antigènes CD40/immunologie , Centre germinatif/cytologie , Centre germinatif/immunologie , Neuromyélite optique/immunologie , Neuromyélite optique/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Lymphocytes T/immunologie , Thymus (glande)/cytologie , Thymus (glande)/immunologie , Cellules épithéliales thyroïdiennes/immunologie , Cellules épithéliales thyroïdiennes/métabolisme , Transcriptome
6.
Cancer Cell ; 41(8): 1498-1515.e10, 2023 08 14.
Article de Anglais | MEDLINE | ID: mdl-37451271

RÉSUMÉ

Type 1 conventional dendritic cells (cDC1) can support T cell responses within tumors but whether this determines protective versus ineffective anti-cancer immunity is poorly understood. Here, we use imaging-based deep learning to identify intratumoral cDC1-CD8+ T cell clustering as a unique feature of protective anti-cancer immunity. These clusters form selectively in stromal tumor regions and constitute niches in which cDC1 activate TCF1+ stem-like CD8+ T cells. We identify a distinct population of immunostimulatory CCR7neg cDC1 that produce CXCL9 to promote cluster formation and cross-present tumor antigens within these niches, which is required for intratumoral CD8+ T cell differentiation and expansion and promotes cancer immune control. Similarly, in human cancers, CCR7neg cDC1 interact with CD8+ T cells in clusters and are associated with patient survival. Our findings reveal an intratumoral phase of the anti-cancer T cell response orchestrated by tumor-residing cDC1 that determines protective versus ineffective immunity and could be exploited for cancer therapy.


Sujet(s)
Lymphocytes T CD8+ , Tumeurs , Humains , Récepteurs CCR7/métabolisme , Tumeurs/thérapie , Antigènes néoplasiques , Cellules dendritiques
7.
Immunity ; 56(6): 1341-1358.e11, 2023 06 13.
Article de Anglais | MEDLINE | ID: mdl-37315536

RÉSUMÉ

Type 1 conventional dendritic cells (cDC1s) are critical for anti-cancer immunity. Protective anti-cancer immunity is thought to require cDC1s to sustain T cell responses within tumors, but it is poorly understood how this function is regulated and whether its subversion contributes to immune evasion. Here, we show that tumor-derived prostaglandin E2 (PGE2) programmed a dysfunctional state in intratumoral cDC1s, disabling their ability to locally orchestrate anti-cancer CD8+ T cell responses. Mechanistically, cAMP signaling downstream of the PGE2-receptors EP2 and EP4 was responsible for the programming of cDC1 dysfunction, which depended on the loss of the transcription factor IRF8. Blockade of the PGE2-EP2/EP4-cDC1 axis prevented cDC1 dysfunction in tumors, locally reinvigorated anti-cancer CD8+ T cell responses, and achieved cancer immune control. In human cDC1s, PGE2-induced dysfunction is conserved and associated with poor cancer patient prognosis. Our findings reveal a cDC1-dependent intratumoral checkpoint for anti-cancer immunity that is targeted by PGE2 for immune evasion.


Sujet(s)
Dinoprostone , Tumeurs , Humains , Anticorps , Lymphocytes T CD8+ , Cellules dendritiques , Récepteur prostaglandine E
8.
Eur J Immunol ; 53(11): e2249923, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-36623939

RÉSUMÉ

This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various non-lymphoid tissues. Here, we provide detailed procedures for a variety of multiparameter fluorescence microscopy imaging methods to explore the spatial organization of DC in tissues and to dissect how DC migrate, communicate, and mediate their multiple functional roles in immunity in a variety of tissue settings. The protocols presented here entail approaches to study DC dynamics and T cell cross-talk by intravital microscopy, large-scale visualization, identification, and quantitative analysis of DC subsets and their functions by multiparameter fluorescence microscopy of fixed tissue sections, and an approach to study DC interactions with tissue cells in a 3D cell culture model. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.


Sujet(s)
Cellules dendritiques , Lymphocytes T , Humains , Microscopie de fluorescence/méthodes
9.
Sci Transl Med ; 14(653): eabm9043, 2022 07 13.
Article de Anglais | MEDLINE | ID: mdl-35857639

RÉSUMÉ

T cell-directed cancer immunotherapy often fails to generate lasting tumor control. Harnessing additional effectors of the immune response against tumors may strengthen the clinical benefit of immunotherapies. Here, we demonstrate that therapeutic targeting of the interferon-γ (IFN-γ)-interleukin-12 (IL-12) pathway relies on the ability of a population of natural killer (NK) cells with tissue-resident traits to orchestrate an antitumor microenvironment. In particular, we used an engineered adenoviral platform as a tool for intratumoral IL-12 immunotherapy (AdV5-IL-12) to generate adaptive antitumor immunity. Mechanistically, we demonstrate that AdV5-IL-12 is capable of inducing the expression of CC-chemokine ligand 5 (CCL5) in CD49a+ NK cells both in tumor mouse models and tumor specimens from patients with cancer. AdV5-IL-12 imposed CCL5-induced type I conventional dendritic cell (cDC1) infiltration and thus increased DC-CD8 T cell interactions. A similar observation was made for other IFN-γ-inducing therapies such as Programmed cell death 1 (PD-1) blockade. Conversely, failure to respond to IL-12 and PD-1 blockade in tumor models with low CD49a+ CXCR6+ NK cell infiltration could be overcome by intratumoral delivery of CCL5. Thus, therapeutic efficacy depends on the abundance of NK cells with tissue-resident traits and, specifically, their capacity to produce the DC chemoattractant CCL5. Our findings reveal a barrier for T cell-focused therapies and offer mechanistic insights into how T cell-NK cell-DC cross-talk can be enhanced to promote antitumor immunity and overcome resistance.


Sujet(s)
Intégrine alpha1 , Tumeurs , Animaux , Cellules dendritiques , Immunothérapie , Intégrine alpha1/métabolisme , Interleukine-12/métabolisme , Cellules tueuses naturelles , Souris , Tumeurs/anatomopathologie , Récepteur-1 de mort cellulaire programmée/métabolisme , Microenvironnement tumoral
10.
Eur J Immunol ; 52(11): 1750-1758, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35106759

RÉSUMÉ

DCs play a pivotal role in orchestrating innate and adaptive antitumor immunity. Activated DCs can produce large amounts of various proinflammatory cytokines, initiate T-cell responses, and exhibit direct cytotoxicity against tumor cells. They also efficiently enhance the antitumoral properties of NK cells and T lymphocytes. Based on these capabilities, immunogenic DCs promote tumor elimination and are associated with improved survival of patients. Furthermore, they can essentially contribute to the clinical efficacy of immunotherapeutic strategies for cancer patients. However, depending on their intrinsic properties and the tumor microenvironment, DCs can be rendered dysfunctional and mediate tolerance by producing immunosuppressive cytokines and activating Treg cells. Such tolerogenic DCs can foster tumor progression and are linked to poor prognosis of patients. Here, we focus on recent studies exploring the phenotype, functional orientation, and clinical relevance of tumor-infiltrating conventional DC1, conventional DC2, plasmacytoid DCs, and monocyte-derived DCs in translational and clinical settings. In addition, recent findings demonstrating the influence of DCs on the efficacy of immunotherapeutic strategies are summarized.


Sujet(s)
Cellules dendritiques , Tumeurs , Humains , Tumeurs/thérapie , Cellules tueuses naturelles , Cytokines , Phénotype , Microenvironnement tumoral
11.
Sci Immunol ; 6(65): eabf7235, 2021 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-34739338

RÉSUMÉ

Deficiency in X-linked inhibitor of apoptosis protein (XIAP) is the cause for X-linked lymphoproliferative syndrome 2 (XLP2). About one-third of these patients suffer from severe and therapy-refractory inflammatory bowel disease (IBD), but the exact cause of this pathogenesis remains undefined. Here, we used XIAP-deficient mice to characterize the mechanisms underlying intestinal inflammation. In Xiap−/− mice, we observed spontaneous terminal ileitis and microbial dysbiosis characterized by a reduction of Clostridia species. We showed that in inflamed mice, both TNF receptor 1 and 2 (TNFR1/2) cooperated in promoting ileitis by targeting TLR5-expressing Paneth cells (PCs) or dendritic cells (DCs). Using intestinal organoids and in vivo modeling, we demonstrated that TLR5 signaling triggered TNF production, which induced PC dysfunction mediated by TNFR1. TNFR2 acted upon lamina propria immune cells. scRNA-seq identified a DC population expressing TLR5, in which Tnfr2 expression was also elevated. Thus, the combined activity of TLR5 and TNFR2 signaling may be responsible for DC loss in lamina propria of Xiap−/− mice. Consequently, both Tnfr1−/−Xiap−/− and Tnfr2−/−Xiap−/− mice were rescued from dysbiosis and intestinal inflammation. Furthermore, RNA-seq of ileal crypts revealed that in inflamed Xiap−/− mice, TLR5 signaling was abrogated, linking aberrant TNF responses with the development of a dysbiosis. Evidence for TNFR2 signaling driving intestinal inflammation was detected in XLP2 patient samples. Together, these data point toward a key role of XIAP in mediating resilience of TLR5-expressing PCs and intestinal DCs, allowing them to maintain tissue integrity and microbiota homeostasis.


Sujet(s)
Inflammation/immunologie , Intestins/immunologie , Récepteur au facteur de nécrose tumorale de type II/immunologie , Récepteur au facteur de nécrose tumorale de type I/immunologie , Récepteur de type Toll-5/immunologie , Protéine inhibitrice de l'apoptose liée au chromosome X/immunologie , Animaux , Cellules dendritiques/immunologie , Dysbiose/immunologie , Humains , Immunité innée/immunologie , Souris , Souris knockout , Cellules de Paneth/immunologie , Récepteur au facteur de nécrose tumorale de type I/déficit , Récepteur au facteur de nécrose tumorale de type II/déficit , Protéine inhibitrice de l'apoptose liée au chromosome X/déficit
12.
Immunity ; 54(10): 2288-2304.e7, 2021 10 12.
Article de Anglais | MEDLINE | ID: mdl-34437840

RÉSUMÉ

Upon viral infection, natural killer (NK) cells expressing certain germline-encoded receptors are selected, expanded, and maintained in an adaptive-like manner. Currently, these are thought to differentiate along a common pathway. However, by fate mapping of single NK cells upon murine cytomegalovirus (MCMV) infection, we identified two distinct NK cell lineages that contributed to adaptive-like responses. One was equivalent to conventional NK (cNK) cells while the other was transcriptionally similar to type 1 innate lymphoid cells (ILC1s). ILC1-like NK cells showed splenic residency and strong cytokine production but also recognized and killed MCMV-infected cells, guided by activating receptor Ly49H. Moreover, they induced clustering of conventional type 1 dendritic cells and facilitated antigen-specific T cell priming early during MCMV infection, which depended on Ly49H and the NK cell-intrinsic expression of transcription factor Batf3. Thereby, ILC1-like NK cells bridge innate and adaptive viral recognition and unite critical features of cNK cells and ILC1s.


Sujet(s)
Immunité acquise/immunologie , Lignage cellulaire/immunologie , Infections à Herpesviridae/immunologie , Immunité innée/immunologie , Cellules tueuses naturelles/immunologie , Animaux , Femelle , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Muromegalovirus
14.
Nature ; 592(7854): 444-449, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33762736

RÉSUMÉ

Nonalcoholic steatohepatitis (NASH) is a manifestation of systemic metabolic disease related to obesity, and causes liver disease and cancer1,2. The accumulation of metabolites leads to cell stress and inflammation in the liver3, but mechanistic understandings of liver damage in NASH are incomplete. Here, using a preclinical mouse model that displays key features of human NASH (hereafter, NASH mice), we found an indispensable role for T cells in liver immunopathology. We detected the hepatic accumulation of CD8 T cells with phenotypes that combined tissue residency (CXCR6) with effector (granzyme) and exhaustion (PD1) characteristics. Liver CXCR6+ CD8 T cells were characterized by low activity of the FOXO1 transcription factor, and were abundant in NASH mice and in patients with NASH. Mechanistically, IL-15 induced FOXO1 downregulation and CXCR6 upregulation, which together rendered liver-resident CXCR6+ CD8 T cells susceptible to metabolic stimuli (including acetate and extracellular ATP) and collectively triggered auto-aggression. CXCR6+ CD8 T cells from the livers of NASH mice or of patients with NASH had similar transcriptional signatures, and showed auto-aggressive killing of cells in an MHC-class-I-independent fashion after signalling through P2X7 purinergic receptors. This killing by auto-aggressive CD8 T cells fundamentally differed from that by antigen-specific cells, which mechanistically distinguishes auto-aggressive and protective T cell immunity.


Sujet(s)
Lymphocytes T CD8+/immunologie , Foie/immunologie , Foie/anatomopathologie , Stéatose hépatique non alcoolique/immunologie , Stéatose hépatique non alcoolique/anatomopathologie , Récepteurs CXCR6/immunologie , Acétates/pharmacologie , Animaux , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/anatomopathologie , Mort cellulaire/effets des médicaments et des substances chimiques , Mort cellulaire/immunologie , Alimentation riche en graisse/effets indésirables , Modèles animaux de maladie humaine , Humains , Interleukine-15/immunologie , Interleukine-15/pharmacologie , Foie/effets des médicaments et des substances chimiques , Mâle , Souris , Souris de lignée C57BL
15.
Adv Immunol ; 147: 61-88, 2020.
Article de Anglais | MEDLINE | ID: mdl-32981635

RÉSUMÉ

Natural killer (NK) cells are innate immune cells critically involved in the control of cancer. Their important role in cancer immunity reflects the ability of NK cells to recognize malignant cells through an array of germline-encoded receptors expressed on their surface, enabling NK cells to detect and rapidly kill tumor cells through targeted cytotoxicity. In addition to their cytotoxic activity, NK cells fulfill a fundamental and often underappreciated role in the local orchestration of cancer immunity through their ability to communicate with innate and adaptive immune cells within the tumor microenvironment (TME), which is achieved through the secretion of multiple chemokines, cytokines, and growth factors. Within tumor tissue, NK cells regulate the recruitment, survival and functional activity of various immune cells including monocytes, granulocytes, dendritic cells and T cells, thereby shaping intratumoral immune cell composition and functionality. Emerging evidence further suggest a role of NK cells in the regulation of stromal cells within the TME. Here, we discuss key aspects of NK cell communication with other intratumoral cell types and its role for cancer immunity. Strategies aimed at boosting anti-cancer immunity by enhancing NK cell communication and functionality within tumor tissue provide attractive new ways for treatment of cancer patients.


Sujet(s)
Immunothérapie adoptive/méthodes , Tumeurs/immunologie , Cellules stromales/immunologie , Animaux , Communication cellulaire , Cytokines/métabolisme , Humains , Immunité innée , Surveillance immunologique , Microenvironnement tumoral
16.
J Clin Invest ; 130(6): 3270-3286, 2020 06 01.
Article de Anglais | MEDLINE | ID: mdl-32191641

RÉSUMÉ

Single-nucleotide polymorphisms and locus amplification link the NF-κB transcription factor c-Rel to human autoimmune diseases and B cell lymphomas, respectively. However, the functional consequences of enhanced c-Rel levels remain enigmatic. Here, we overexpressed c-Rel specifically in mouse B cells from BAC-transgenic gene loci and demonstrate that c-Rel protein levels linearly dictated expansion of germinal center B (GCB) cells and isotype-switched plasma cells. c-Rel expression in B cells of otherwise c-Rel-deficient mice fully rescued terminal B cell differentiation, underscoring its critical B cell-intrinsic roles. Unexpectedly, in GCB cells transcription-independent regulation produced the highest c-Rel protein levels among B cell subsets. In c-Rel-overexpressing GCB cells this caused enhanced nuclear translocation, a profoundly altered transcriptional program, and increased proliferation. Finally, we provide a link between c-Rel gain and autoimmunity by showing that c-Rel overexpression in B cells caused autoantibody production and renal immune complex deposition.


Sujet(s)
Production d'anticorps , Autoanticorps/immunologie , Centre germinatif/immunologie , Plasmocytes/immunologie , Polymorphisme de nucléotide simple , Protéines proto-oncogènes c-rel/immunologie , Animaux , Autoanticorps/génétique , Centre germinatif/anatomopathologie , Souris , Souris transgéniques , Plasmocytes/anatomopathologie , Protéines proto-oncogènes c-rel/génétique
17.
Immunity ; 52(2): 313-327.e7, 2020 02 18.
Article de Anglais | MEDLINE | ID: mdl-32049052

RÉSUMÉ

T cell responses upon infection display a remarkably reproducible pattern of expansion, contraction, and memory formation. If the robustness of this pattern builds entirely on signals derived from other cell types or if activated T cells themselves contribute to the orchestration of these population dynamics-akin to bacterial quorum regulation-is unclear. Here, we examined this question using time-lapse microscopy, genetic perturbation, bioinformatic predictions, and mathematical modeling. We found that ICAM-1-mediated cell clustering enabled CD8+ T cells to collectively regulate the balance between proliferation and apoptosis. Mechanistically, T cell expressed CD80 and CD86 interacted with the receptors CD28 and CTLA-4 on neighboring T cells; these interactions fed two nested antagonistic feedback circuits that regulated interleukin 2 production in a manner dependent on T cell density as confirmed by in vivo modulation of this network. Thus, CD8+ T cell-population-intrinsic mechanisms regulate cellular behavior, thereby promoting robustness of population dynamics.


Sujet(s)
Antigène CD28/métabolisme , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Antigène CTLA-4/métabolisme , Animaux , Antigène CD80/métabolisme , Antigène CD86/métabolisme , Lymphocytes T CD8+/métabolisme , Communication cellulaire , Numération cellulaire , Lignée cellulaire , Survie cellulaire , Suivi cellulaire , Cellules dendritiques/immunologie , Molécule-1 d'adhérence intercellulaire/métabolisme , Interleukine-2/métabolisme , Activation des lymphocytes , Souris , Souris de lignée C57BL , Souris transgéniques , Modèles théoriques
18.
Trends Cancer ; 4(11): 784-792, 2018 11.
Article de Anglais | MEDLINE | ID: mdl-30352680

RÉSUMÉ

Dendritic cells (DCs) are key orchestrators of immune responses. A specific DC subset, conventional type 1 DCs (cDC1s), has been recently associated with human cancer patient survival and, in preclinical models, is critical for the spontaneous rejection of immunogenic cancers and for the success of T cell-based immunotherapies. The unique role of cDC1 reflects the ability to initiate de novo T cell responses after migrating to tumor-draining lymph nodes, as well as to attract T cells, secrete cytokines, and present tumor antigens within the tumor microenvironment, enhancing local cytotoxic T cell function. Strategies aimed at increasing cDC1 abundance in tumors and enhancing their functionality provide attractive new avenues to boost anti-tumor immunity and overcome resistance to cancer immunotherapies.


Sujet(s)
Cellules dendritiques/immunologie , Tumeurs/immunologie , Animaux , Antigènes néoplasiques/immunologie , Humains , Noeuds lymphatiques/immunologie , Lymphocytes T/immunologie
19.
Cell Rep ; 24(2): 419-428, 2018 07 10.
Article de Anglais | MEDLINE | ID: mdl-29996102

RÉSUMÉ

Conventional type 1 DCs (cDC1s) excel at cross-presentation of dead cell-associated antigens partly because they express DNGR-1, a receptor that recognizes exposed actin filaments on dead cells. In vitro polymerized F-actin can be used as a synthetic ligand for DNGR-1. However, cellular F-actin is decorated with actin-binding proteins, which could affect DNGR-1 recognition. Here, we demonstrate that myosin II, an F-actin-associated motor protein, greatly potentiates the binding of DNGR-1 to F-actin. Latex beads coated with F-actin and myosin II are taken up by DNGR-1+ cDC1s, and antigen associated with those beads is efficiently cross-presented to CD8+ T cells. Myosin II-deficient necrotic cells are impaired in their ability to stimulate DNGR-1 or to serve as substrates for cDC1 cross-presentation to CD8+ T cells. These results provide insights into the nature of the DNGR-1 ligand and have implications for understanding immune responses to cell-associated antigens and for vaccine design.


Sujet(s)
Actines/métabolisme , Antigènes/métabolisme , Cross-priming/immunologie , Lectines de type C/métabolisme , Myosine de type II/métabolisme , Récepteurs immunologiques/métabolisme , Animaux , Mort cellulaire , Lignée cellulaire , Immunisation , Mélanome expérimental/immunologie , Mélanome expérimental/anatomopathologie , Souris de lignée C57BL , Chaînes lourdes de myosine/métabolisme , Phagocytose , Liaison aux protéines , Lymphocytes T cytotoxiques/immunologie
20.
Cell ; 172(5): 1022-1037.e14, 2018 02 22.
Article de Anglais | MEDLINE | ID: mdl-29429633

RÉSUMÉ

Conventional type 1 dendritic cells (cDC1) are critical for antitumor immunity, and their abundance within tumors is associated with immune-mediated rejection and the success of immunotherapy. Here, we show that cDC1 accumulation in mouse tumors often depends on natural killer (NK) cells that produce the cDC1 chemoattractants CCL5 and XCL1. Similarly, in human cancers, intratumoral CCL5, XCL1, and XCL2 transcripts closely correlate with gene signatures of both NK cells and cDC1 and are associated with increased overall patient survival. Notably, tumor production of prostaglandin E2 (PGE2) leads to evasion of the NK cell-cDC1 axis in part by impairing NK cell viability and chemokine production, as well as by causing downregulation of chemokine receptor expression in cDC1. Our findings reveal a cellular and molecular checkpoint for intratumoral cDC1 recruitment that is targeted by tumor-derived PGE2 for immune evasion and that could be exploited for cancer therapy.


Sujet(s)
Cellules dendritiques/immunologie , Cellules tueuses naturelles/immunologie , Tumeurs/immunologie , Tumeurs/anatomopathologie , Microenvironnement tumoral/immunologie , Animaux , Lignée cellulaire tumorale , Chimiokine CCL5/métabolisme , Chimiokines C/métabolisme , Cyclooxygenase 1/métabolisme , Cyclooxygenase 2/métabolisme , Dinoprostone/métabolisme , Régulation de l'expression des gènes tumoraux , Humains , Mélanome/génétique , Mélanome/anatomopathologie , Souris , Mutation/génétique , Pronostic , Protéines proto-oncogènes B-raf/génétique , Analyse de survie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE