Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 33
Filtrer
1.
Biomed Pharmacother ; 177: 117057, 2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38976957

RÉSUMÉ

Cyclotides are head-to-tail cyclized peptides with a unique cystine-knot motif. Their structure provides exceptional resistance against enzymatic, chemical, or thermal degradation compared to other peptides. Peptide-based therapeutics promise high specificity, selectivity and lower immunogenicity, making them safer alternatives to small molecules or large biologicals. Cyclotides were researched due to their anti-cancer properties by inducing apoptosis in tumor cells in the past, but the impact of cyclotides on cytotoxic immune cells was poorly studied. Natural Killer (NK) cells are cytotoxic innate lymphoid cells and play an important role in the defense against infected, stressed and transformed cells. NK cells do not need prior sensitization and act in an antigen independent manner, holding promising potential in the field of immunotherapy. To investigate the effect of immunomodulatory cyclotides on NK cells, we evaluated several peptide-enriched plant extracts on NK cell mediated cytotoxicity. We observed that the extract samples derived from Carapichea ipecacuanha (Brot.) L. Andersson augments the killing potential of mouse NK cells against different tumor targets in vitro. Subsequent isolation of cyclotides from C. ipecacuanha extracts led to the identification of a primary candidate that enhances cytotoxicity of both mouse and human NK cells. The augmented killing is facilitated by the increased degranulation capacity of NK cells. In addition, we noted a direct toxic effect of caripe 8 on tumor cells, suggesting a dual therapeutic potential in cancer treatment. This study offers novel insights how natural peptides can influence NK cell cytotoxicity. These pre-clinical findings hold significant promise for advancing current immunotherapeutic approaches.

2.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38876796

RÉSUMÉ

Innate lymphoid cells (ILCs) are critical for intestinal adaptation to microenvironmental challenges, and the gut mucosa is characterized by low oxygen. Adaptation to low oxygen is mediated by hypoxia-inducible transcription factors (HIFs), and the HIF-1α subunit shapes an ILC phenotype upon acute colitis that contributes to intestinal damage. However, the impact of HIF signaling in NKp46+ ILCs in the context of repetitive mucosal damage and chronic inflammation, as it typically occurs during inflammatory bowel disease, is unknown. In chronic colitis, mice lacking the HIF-1α isoform in NKp46+ ILCs show a decrease in NKp46+ ILC1s but a concomitant rise in neutrophils and Ly6Chigh macrophages. Single-nucleus RNA sequencing suggests enhanced interaction of mesenchymal cells with other cell compartments in the colon of HIF-1α KO mice and a loss of mucus-producing enterocytes and intestinal stem cells. This was, furthermore, associated with increased bone morphogenetic pathway-integrin signaling, expansion of fibroblast subsets, and intestinal fibrosis. In summary, this suggests that HIF-1α-mediated ILC1 activation, although detrimental upon acute colitis, protects against excessive inflammation and fibrosis during chronic intestinal damage.


Sujet(s)
Colite , Fibrose , Sous-unité alpha du facteur-1 induit par l'hypoxie , Lymphocytes , Souris knockout , Récepteur-1 de déclenchement de cytotoxicité naturelle , Animaux , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Récepteur-1 de déclenchement de cytotoxicité naturelle/métabolisme , Récepteur-1 de déclenchement de cytotoxicité naturelle/génétique , Souris , Colite/métabolisme , Colite/génétique , Lymphocytes/métabolisme , Lymphocytes/immunologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Inflammation/métabolisme , Souris de lignée C57BL , Maladie chronique , Immunité innée , Transduction du signal , Modèles animaux de maladie humaine , Mâle , Intestins/anatomopathologie , Antigènes Ly
3.
Blood ; 143(24): 2474-2489, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38498036

RÉSUMÉ

ABSTRACT: Patients with T- and natural killer (NK)-cell neoplasms frequently have somatic STAT5B gain-of-function mutations. The most frequent STAT5B mutation is STAT5BN642H, which is known to drive murine T-cell leukemia, although its role in NK-cell malignancies is unclear. Introduction of the STAT5BN642H mutation into human NK-cell lines enhances their potential to induce leukemia in mice. We have generated a mouse model that enables tissue-specific expression of STAT5BN642H and have selectively expressed the mutated STAT5B in hematopoietic cells (N642Hvav/+) or exclusively in NK cells (N642HNK/NK). All N642Hvav/+ mice rapidly develop an aggressive T/NKT-cell leukemia, whereas N642HNK/NK mice display an indolent NK-large granular lymphocytic leukemia (NK-LGLL) that progresses to an aggressive leukemia with age. Samples from patients with NK-cell leukemia have a distinctive transcriptional signature driven by mutant STAT5B, which overlaps with that of murine leukemic N642HNK/NK NK cells. To our knowledge, we have generated the first reliable STAT5BN642H-driven preclinical mouse model that displays an indolent NK-LGLL progressing to aggressive NK-cell leukemia. This novel in vivo tool will enable us to explore the transition from an indolent to an aggressive disease and will thus permit the study of prevention and treatment options for NK-cell malignancies.


Sujet(s)
Cellules tueuses naturelles , Leucémie à grands lymphocytes granuleux , Facteur de transcription STAT-5 , Animaux , Facteur de transcription STAT-5/génétique , Facteur de transcription STAT-5/métabolisme , Souris , Cellules tueuses naturelles/métabolisme , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/anatomopathologie , Humains , Leucémie à grands lymphocytes granuleux/génétique , Leucémie à grands lymphocytes granuleux/anatomopathologie , Modèles animaux de maladie humaine , Lignage cellulaire/génétique , Mutation , Souris transgéniques
4.
Curr Opin Immunol ; 85: 102397, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37931499

RÉSUMÉ

The proper functioning of cytotoxic lymphocytes, such as natural killer and CD8+ T cells, is essential for effective cancer-immunity and immunotherapy responses. The differentiation of these cells is controlled by several transcription factors (TFs), including members of the activator protein (AP)-1 family. The activity of AP-1 family members is regulated by various immune signaling pathways, which can be triggered by activating or inhibitory receptors as well as cytokines. The target genes controlled by AP-1 TFs are central to generate immunity to pathogens or malignancies. Here, we provide an overview of the current understanding of how AP-1 TFs regulate cytotoxic lymphocytes.


Sujet(s)
Antinéoplasiques , Tumeurs , Humains , Facteur de transcription AP-1/métabolisme , Lymphocytes T CD8+ , Cytokines/métabolisme , Lymphocytes T cytotoxiques
5.
PLoS One ; 18(9): e0286256, 2023.
Article de Anglais | MEDLINE | ID: mdl-37713409

RÉSUMÉ

FAM3C/ILEI is an important factor in epithelial-to-mesenchymal transition (EMT) induction, tumor progression and metastasis. Overexpressed in many cancers, elevated ILEI levels and secretion correlate with poor patient survival. Although ILEI's causative role in invasive tumor growth and metastasis has been demonstrated in several cellular tumor models, there are no available transgenic mice to study these effects in the context of a complex organism. Here, we describe the generation and initial characterization of a Tet-ON inducible Fam3c/ILEI transgenic mouse strain. We find that ubiquitous induction of ILEI overexpression (R26-ILEIind) at weaning age leads to a shortened lifespan, reduced body weight and microcytic hypochromic anemia. The anemia was reversible at a young age within a week upon withdrawal of ILEI induction. Vav1-driven overexpression of the ILEIind transgene in all hematopoietic cells (Vav-ILEIind) did not render mice anemic or lower overall fitness, demonstrating that no intrinsic mechanisms of erythroid development were dysregulated by ILEI and that hematopoietic ILEI hyperfunction did not contribute to death. Reduced serum iron levels of R26-ILEIind mice were indicative for a malfunction in iron uptake or homeostasis. Accordingly, the liver, the main organ of iron metabolism, was severely affected in moribund ILEI overexpressing mice: increased alanine transaminase and aspartate aminotransferase levels indicated liver dysfunction, the liver was reduced in size, showed increased apoptosis, reduced cellular iron content, and had a fibrotic phenotype. These data indicate that high ILEI expression in the liver might reduce hepatoprotection and induce liver fibrosis, which leads to liver dysfunction, disturbed iron metabolism and eventually to death. Overall, we show here that the novel Tet-ON inducible Fam3c/ILEI transgenic mouse strain allows tissue specific timely controlled overexpression of ILEI and thus, will serve as a versatile tool to model the effect of elevated ILEI expression in diverse tissue entities and disease conditions, including cancer.


Sujet(s)
Anémie , Longévité , Souris , Animaux , Longévité/génétique , Cirrhose du foie/génétique , Anémie/génétique , Fer , Souris transgéniques
7.
EMBO Rep ; 24(6): e56156, 2023 06 05.
Article de Anglais | MEDLINE | ID: mdl-36987917

RÉSUMÉ

Natural killer (NK) cells are forced to cope with different oxygen environments even under resting conditions. The adaptation to low oxygen is regulated by oxygen-sensitive transcription factors, the hypoxia-inducible factors (HIFs). The function of HIFs for NK cell activation and metabolic rewiring remains controversial. Activated NK cells are predominantly glycolytic, but the metabolic programs that ensure the maintenance of resting NK cells are enigmatic. By combining in situ metabolomic and transcriptomic analyses in resting murine NK cells, our study defines HIF-1α as a regulator of tryptophan metabolism and cellular nicotinamide adenine dinucleotide (NAD+ ) levels. The HIF-1α/NAD+ axis prevents ROS production during oxidative phosphorylation (OxPhos) and thereby blocks DNA damage and NK cell apoptosis under steady-state conditions. In contrast, in activated NK cells under hypoxia, HIF-1α is required for glycolysis, and forced HIF-1α expression boosts glycolysis and NK cell performance in vitro and in vivo. Our data highlight two distinct pathways by which HIF-1α interferes with NK cell metabolism. While HIF-1α-driven glycolysis is essential for NK cell activation, resting NK cell homeostasis relies on HIF-1α-dependent tryptophan/NAD+ metabolism.


Sujet(s)
NAD , Tryptophane , Souris , Animaux , Tryptophane/métabolisme , Cellules tueuses naturelles , Glycolyse/génétique , Hypoxie/métabolisme , Hypoxie cellulaire , Oxygène/métabolisme , Homéostasie , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme
8.
J Leukoc Biol ; 113(1): 27-40, 2023 01 10.
Article de Anglais | MEDLINE | ID: mdl-36822164

RÉSUMÉ

Natural killer (NK) cells are innate lymphocytes capable of mediating immune responses without prior sensitization. NK cells express Fc-gamma receptors (FcγRs) that engage the Fc region of IgG. Studies investigating the role of FcγRs on mouse NK cells have been limited due to lack specific reagents. In this study, we characterize the expression and biological consequences of activating mouse NK cells through their FcγRs. We demonstrate that most NK cells express the activating CD16 receptor, and a subset of NK cells also expresses the inhibitory CD32b receptor. Critically, these FcγRs are functional on mouse NK cells and can modulate antibody-mediated responses. We also characterized mice with conditional knockout alleles of Fcgr3 (CD16) or Fcgr2b (CD32b) in the NK and innate lymphoid cell (ILC) lineage. NK cells in these mice did not reveal any developmental defects and were responsive to cross-linking activating NK receptors, cytokine stimulation, and killing of YAC-1 targets. Importantly, CD16-deficient NK cells failed to induce antibody-directed cellular cytotoxicity of antibody-coated B-cell lymphomas in in vitro assays. In addition, we demonstrate the important role of CD16 on NK cells using an in vivo model of cancer immunotherapy using anti-CD20 antibody treatment of B-cell lymphomas.


Sujet(s)
Immunité innée , Lymphome B , Souris , Animaux , Récepteurs du fragment Fc des IgG/métabolisme , Cytotoxicité immunologique , Cellules tueuses naturelles , Anticorps/métabolisme
9.
Cell Stem Cell ; 29(10): 1459-1474.e9, 2022 Oct 06.
Article de Anglais | MEDLINE | ID: mdl-36113462

RÉSUMÉ

Fibrosis is the final path of nearly every form of chronic disease, regardless of the pathogenesis. Upon chronic injury, activated, fibrogenic fibroblasts deposit excess extracellular matrix, and severe tissue fibrosis can occur in virtually any organ. However, antifibrotic therapies that target fibrogenic cells, while sparing homeostatic fibroblasts in healthy tissues, are limited. We tested whether specific immunization against endogenous proteins, strongly expressed in fibrogenic cells but highly restricted in quiescent fibroblasts, can elicit an antigen-specific cytotoxic T cell response to ameliorate organ fibrosis. In silico epitope prediction revealed that activation of the genes Adam12 and Gli1 in profibrotic cells and the resulting "self-peptides" can be exploited for T cell vaccines to ablate fibrogenic cells. We demonstrate the efficacy of a vaccination approach to mount CD8+ T cell responses that reduce fibroblasts and fibrosis in the liver and lungs in mice. These results provide proof of principle for vaccination-based immunotherapies to treat fibrosis.


Sujet(s)
Fibroblastes , Poumon , Animaux , Épitopes/métabolisme , Fibroblastes/métabolisme , Fibrose , Immunothérapie , Foie/anatomopathologie , Poumon/métabolisme , Souris , Vaccination , Protéine à doigt de zinc GLI1/métabolisme
10.
Front Immunol ; 13: 909275, 2022.
Article de Anglais | MEDLINE | ID: mdl-35784374

RÉSUMÉ

Multiple sclerosis (MS) is a highly debilitating autoimmune disease affecting millions of individuals worldwide. Although classically viewed as T-cell mediated disease, the role of innate lymphoid cells (ILC) such as natural killer (NK) cells and ILC 1-3s has become a focal point as several findings implicate them in the disease pathology. The role of ILCs in MS is still not completely understood as controversial findings have been reported assigning them either a protective or disease-accelerating role. Recent findings in experimental autoimmune encephalomyelitis (EAE) suggest that ILCs infiltrate the central nervous system (CNS), mediate inflammation, and have a disease exacerbating role by influencing the recruitment of autoreactive T-cells. Elucidating the detailed role of ILCs and altered signaling pathways in MS is essential for a more complete picture of the disease pathology and novel therapeutic targets. We here review the current knowledge about ILCs in the development and progression of MS and preclinical models of MS and discuss their potential for therapeutic applications.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Sclérose en plaques , Animaux , Humains , Immunité innée , Cellules tueuses naturelles/anatomopathologie , Sclérose en plaques/anatomopathologie , Lymphocytes T/anatomopathologie
11.
Front Immunol ; 13: 909270, 2022.
Article de Anglais | MEDLINE | ID: mdl-35812461

RÉSUMÉ

Natural killer (NK) cells play an important role in recognizing and killing pathogen-infected or malignant cells. Changes in their numbers or activation can contribute to several diseases and pathologies including systemic sclerosis (SSc), an autoimmune disease characterized by inflammation and tissue remodeling. In these patients, increased expression of the AP-1 transcription factor, Fra-2 was reported. In mice ectopic overexpression of Fra-2 (TG) leads to SSc with strong pulmonary fibrosis, pulmonary hypertension, and inflammation. Analysis of the underlying immune cell profile in the lungs of young TG mice, which do not yet show any signs of lung disease, revealed increased numbers of eosinophils and T cells but strongly reduced NK numbers. Therefore, we aimed to identify the cause of the absence of NK cells in the lungs of these mice and to determine the potential role of Fra-2 in NK development. Examination of inflammatory cell distribution in TG mice revealed similar NK deficiencies in the spleen, blood, and bone marrow. Deeper analysis of the WT and TG bone marrow revealed a potential NK cell developmental defect beginning at the preNKP stage. To determine whether this defect was cell-intrinsic or extrinsic, mixed bone marrow chimera and in vitro differentiation experiments were performed. Both experiments showed that the defect caused by Fra-2 was primarily cell-intrinsic and minimally dependent on the environment. Closer examination of surface markers and transcription factors required for NK development, revealed the expected receptor distribution but changes in transcription factor expression. We found a significant reduction in Nfil3, which is essential for the transition of common lymphoid cells to NK committed precursor cells and an AP-1 binding site in the promotor of this gene. In Summary, our data demonstrates that regulation of Fra-2 is essential for NK development and maturation, and suggests that the early NK dysfunction plays an important role in the pathogenesis of systemic sclerosis.


Sujet(s)
Sclérodermie systémique , Facteur de transcription AP-1 , Animaux , Antigène-2 apparenté à fos/génétique , Antigène-2 apparenté à fos/métabolisme , Inflammation/métabolisme , Cellules tueuses naturelles , Souris , Sclérodermie systémique/anatomopathologie , Facteur de transcription AP-1/métabolisme
12.
J Exp Med ; 219(2)2022 02 07.
Article de Anglais | MEDLINE | ID: mdl-35024767

RÉSUMÉ

Gut innate lymphoid cells (ILCs) show remarkable phenotypic diversity, yet microenvironmental factors that drive this plasticity are incompletely understood. The balance between NKp46+, IL-22-producing, group 3 ILCs (ILC3s) and interferon (IFN)-γ-producing group 1 ILCs (ILC1s) contributes to gut homeostasis. The gut mucosa is characterized by physiological hypoxia, and adaptation to low oxygen is mediated by hypoxia-inducible transcription factors (HIFs). However, the impact of HIFs on ILC phenotype and gut homeostasis is not well understood. Mice lacking the HIF-1α isoform in NKp46+ ILCs show a decrease in IFN-γ-expressing, T-bet+, NKp46+ ILC1s and a concomitant increase in IL-22-expressing, RORγt+, NKp46+ ILC3s in the gut mucosa. Single-cell RNA sequencing revealed HIF-1α as a driver of ILC phenotypes, where HIF-1α promotes the ILC1 phenotype by direct up-regulation of T-bet. Loss of HIF-1α in NKp46+ cells prevents ILC3-to-ILC1 conversion, increases the expression of IL-22-inducible genes, and confers protection against intestinal damage. Taken together, our results suggest that HIF-1α shapes the ILC phenotype in the gut.


Sujet(s)
Antigènes Ly/métabolisme , Plasticité cellulaire/immunologie , Tube digestif/physiologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Immunité innée , Lymphocytes intra-épithéliaux/immunologie , Lymphocytes intra-épithéliaux/métabolisme , Récepteur-1 de déclenchement de cytotoxicité naturelle/métabolisme , Animaux , Marqueurs biologiques , Prédisposition aux maladies , Expression des gènes , Analyse de profil d'expression de gènes , Homéostasie , Immunité muqueuse , Immunophénotypage , Muqueuse intestinale/immunologie , Muqueuse intestinale/métabolisme , Sous-populations de lymphocytes , Souris , Souris knockout , Microbiote , Analyse sur cellule unique
13.
Cell Death Dis ; 12(11): 991, 2021 10 23.
Article de Anglais | MEDLINE | ID: mdl-34689158

RÉSUMÉ

Triple-negative breast cancer (TNBC) is an aggressive malignant disease that is responsible for approximately 15% of breast cancers. The standard of care relies on surgery and chemotherapy but the prognosis is poor and there is an urgent need for new therapeutic strategies. Recent in silico studies have revealed an inverse correlation between recurrence-free survival and the level of cyclin-dependent kinase 8 (CDK8) in breast cancer patients. CDK8 is known to have a role in natural killer (NK) cell cytotoxicity, but its function in TNBC progression and immune cell recognition or escape has not been investigated. We have used a murine model of orthotopic breast cancer to study the tumor-intrinsic role of CDK8 in TNBC. Knockdown of CDK8 in TNBC cells impairs tumor regrowth upon surgical removal and prevents metastasis. In the absence of CDK8, the epithelial-to-mesenchymal transition (EMT) is impaired and immune-mediated tumor-cell clearance is facilitated. CDK8 drives EMT in TNBC cells in a kinase-independent manner. In vivo experiments have confirmed that CDK8 is a crucial regulator of NK-cell-mediated immune evasion in TNBC. The studies also show that CDK8 is involved in regulating the checkpoint inhibitor programmed death-ligand 1 (PD-L1). The CDK8-PD-L1 axis is found in mouse and human TNBC cells, underlining the importance of CDK8-driven immune cell evasion in these highly aggressive breast cancer cells. Our data link CDK8 to PD-L1 expression and provide a rationale for investigating the possibility of CDK8-directed therapy for TNBC.


Sujet(s)
Cyclin-Dependent Kinase 8/métabolisme , Cellules tueuses naturelles/métabolisme , Tumeurs du sein triple-négatives/génétique , Animaux , Humains , Souris , Tumeurs du sein triple-négatives/anatomopathologie
14.
Nat Commun ; 12(1): 4700, 2021 08 04.
Article de Anglais | MEDLINE | ID: mdl-34349124

RÉSUMÉ

During skin injury, immune response and repair mechanisms have to be coordinated for rapid skin regeneration and the prevention of microbial infections. Natural Killer (NK) cells infiltrate hypoxic skin lesions and Hypoxia-inducible transcription factors (HIFs) mediate adaptation to low oxygen. We demonstrate that mice lacking the Hypoxia-inducible factor (HIF)-1α isoform in NK cells show impaired release of the cytokines Interferon (IFN)-γ and Granulocyte Macrophage - Colony Stimulating Factor (GM-CSF) as part of a blunted immune response. This accelerates skin angiogenesis and wound healing. Despite rapid wound closure, bactericidal activity and the ability to restrict systemic bacterial infection are impaired. Conversely, forced activation of the HIF pathway supports cytokine release and NK cell-mediated antibacterial defence including direct killing of bacteria by NK cells despite delayed wound closure. Our results identify, HIF-1α in NK cells as a nexus that balances antimicrobial defence versus global repair in the skin.


Sujet(s)
Cellules tueuses naturelles/immunologie , Peau/immunologie , Peau/microbiologie , Cicatrisation de plaie , Animaux , Hypoxie cellulaire , Cytokines/métabolisme , Hypoxie , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Cellules tueuses naturelles/métabolisme , Cellules tueuses naturelles/anatomopathologie , Souris , Néovascularisation physiologique , Peau/vascularisation , Dermatoses bactériennes/prévention et contrôle
15.
Front Immunol ; 12: 650977, 2021.
Article de Anglais | MEDLINE | ID: mdl-34248938

RÉSUMÉ

The cyclin-dependent kinase 6 (CDK6) regulates the transition through the G1-phase of the cell cycle, but also acts as a transcriptional regulator. As such CDK6 regulates cell survival or cytokine secretion together with STATs, AP-1 or NF-κB. In the hematopoietic system, CDK6 regulates T cell development and promotes leukemia and lymphoma. CDK4/6 kinase inhibitors are FDA approved for treatment of breast cancer patients and have been reported to enhance T cell-mediated anti-tumor immunity. The involvement of CDK6 in T cell functions remains enigmatic. We here investigated the role of CDK6 in CD8+ T cells, using previously generated CDK6 knockout (Cdk6-/-) and kinase-dead mutant CDK6 (Cdk6K43M) knock-in mice. RNA-seq analysis indicated a role of CDK6 in T cell metabolism and interferon (IFN) signaling. To investigate whether these CDK6 functions are T cell-intrinsic, we generated a T cell-specific CDK6 knockout mouse model (Cdk6fl/fl CD4-Cre). T cell-intrinsic loss of CDK6 enhanced mitochondrial respiration in CD8+ T cells, but did not impact on cytotoxicity and production of the effector cytokines IFN-γ and TNF-α by CD8+ T cells in vitro. Loss of CDK6 in peripheral T cells did not affect tumor surveillance of MC38 tumors in vivo. Similarly, while we observed an impaired induction of early responses to type I IFN in CDK6-deficient CD8+ T cells, we failed to observe any differences in the response to LCMV infection upon T cell-intrinsic loss of CDK6 in vivo. This apparent contradiction might at least partially be explained by the reduced expression of Socs1, a negative regulator of IFN signaling, in CDK6-deficient CD8+ T cells. Therefore, our data are in line with a dual role of CDK6 in IFN signaling; while CDK6 promotes early IFN responses, it is also involved in the induction of a negative feedback loop. These data assign CDK6 a role in the fine-tuning of cytokine responses.


Sujet(s)
Antiviraux/immunologie , Lymphocytes T CD8+/immunologie , Kinase-6 cycline-dépendante/immunologie , Cytotoxicité immunologique/immunologie , Interférons/immunologie , Tumeurs expérimentales/immunologie , Animaux , Antiviraux/métabolisme , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD8+/métabolisme , Lymphocytes T CD8+/virologie , Lignée cellulaire , Lignée cellulaire tumorale , Kinase-6 cycline-dépendante/génétique , Kinase-6 cycline-dépendante/métabolisme , Humains , Interférons/métabolisme , Virus de la chorioméningite lymphocytaire/immunologie , Virus de la chorioméningite lymphocytaire/physiologie , Mâle , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Tumeurs expérimentales/métabolisme , Transduction du signal/immunologie
16.
Eur J Immunol ; 50(6): 880-890, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32052406

RÉSUMÉ

NK cells are innate lymphocytes responsible for lysis of pathogen-infected and transformed cells. One of the major activating receptors required for target cell recognition is the NK group 2D (NKG2D) receptor. Numerous reports show the necessity of NKG2D for effective tumor immune surveillance. Further studies identified NKG2D as a key element allowing tumor immune escape. We here use a mouse model with restricted deletion of NKG2D in mature NKp46+ cells (NKG2DΔNK ). NKG2DΔNK NK cells develop normally, have an unaltered IFN-γ production but kill tumor cell lines expressing NKG2D ligands (NKG2DLs) less efficiently. However, upon long-term stimulation with IL-2, NKG2D-deficient NK cells show increased levels of the lytic molecule perforin. Thus, our findings demonstrate a dual function of NKG2D for NK cell cytotoxicity; while NKG2D is a crucial trigger for cytotoxicity of tumor cells expressing activating ligands it is also capable to limit perforin production in IL-2 activated NK cells.


Sujet(s)
Interleukine-2/pharmacologie , Cellules tueuses naturelles/immunologie , Sous-famille K des récepteurs de cellules NK de type lectine/immunologie , Perforines/immunologie , Animaux , Lignée cellulaire tumorale , Immunité cellulaire/effets des médicaments et des substances chimiques , Immunité cellulaire/génétique , Interféron gamma/génétique , Interféron gamma/immunologie , Cellules tueuses naturelles/anatomopathologie , Souris , Souris knockout , Sous-famille K des récepteurs de cellules NK de type lectine/génétique , Perforines/génétique
17.
Front Immunol ; 10: 2590, 2019.
Article de Anglais | MEDLINE | ID: mdl-31781102

RÉSUMÉ

Natural Killer (NK) cells are cytotoxic lymphocytes of the innate immune system and play a critical role in anti-viral and anti-tumor responses. NK cells develop in the bone marrow from hematopoietic stem cells (HSCs) that differentiate through common lymphoid progenitors (CLPs) to NK lineage-restricted progenitors (NKPs). The orchestrated action of multiple cytokines is crucial for NK cell development and maturation. Many of these cytokines such as IL-2, IL-7, IL-12, IL-15, IL-21, IL-27, and interferons (IFNs) signal via the Janus Kinase / Signal Transducer and Activator of Transcription (JAK/STAT) pathway. We here review the current knowledge about these cytokines and the downstream signaling involved in the development and maturation of conventional NK cells and their close relatives, innate lymphoid cells type 1 (ILC1). We further discuss the role of suppressor of cytokine signaling (SOCS) proteins in NK cells and highlight their potential for therapeutic application.


Sujet(s)
Différenciation cellulaire , Cytokines/métabolisme , Janus kinases/métabolisme , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Facteurs de transcription STAT/métabolisme , Transduction du signal , Marqueurs biologiques , Régulation de l'expression des gènes , Humains , Cellules tueuses naturelles/cytologie
18.
Cancer Immunol Res ; 6(4): 458-466, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-29386186

RÉSUMÉ

Cyclin-dependent kinase 8 (CDK8) is a member of the transcription-regulating CDK family. CDK8 activates or represses transcription by associating with the mediator complex or by regulating transcription factors. Oncogenic activity of CDK8 has been demonstrated in several cancer types. Targeting CDK8 represents a potential therapeutic strategy. Because knockdown of CDK8 in a natural killer (NK) cell line enhances cytotoxicity and NK cells provide the first line of immune defense against transformed cells, we asked whether inhibiting CDK8 would improve NK-cell antitumor responses. In this study, we investigated the role of CDK8 in NK-cell function in vivo using mice with conditional ablation of CDK8 in NKp46+ cells (Cdk8fl/flNcr1Cre). Regardless of CDK8 expression, NK cells develop and mature normally in bone marrow and spleen. However, CDK8 deletion increased expression of the lytic molecule perforin, which correlated with enhanced NK-cell cytotoxicity in vitro This translates into improved NK cell-mediated tumor surveillance in vivo in three independent models: B16F10 melanoma, v-abl+ lymphoma, and a slowly developing oncogene-driven leukemia. Our results thereby define a suppressive effect of CDK8 on NK-cell activity. Therapies that target CDK8 in cancer patients may enhance NK-cell responses against tumor cells. Cancer Immunol Res; 6(4); 458-66. ©2018 AACR.


Sujet(s)
Cyclin-Dependent Kinase 8/génétique , Délétion de gène , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Tumeurs/génétique , Tumeurs/immunologie , Animaux , Différenciation cellulaire/génétique , Lignée cellulaire tumorale , Cytotoxicité immunologique , Modèles animaux de maladie humaine , Immunité innée , Cellules tueuses naturelles/cytologie , Mélanome expérimental , Souris , Souris transgéniques , Tumeurs/anatomopathologie
19.
Nat Commun ; 8(1): 1597, 2017 11 17.
Article de Anglais | MEDLINE | ID: mdl-29150606

RÉSUMÉ

Productive angiogenesis, a prerequisite for tumour growth, depends on the balanced release of angiogenic and angiostatic factors by different cell types within hypoxic tumours. Natural killer (NK) cells kill cancer cells and infiltrate hypoxic tumour areas. Cellular adaptation to low oxygen is mediated by Hypoxia-inducible factors (HIFs). We found that deletion of HIF-1α in NK cells inhibited tumour growth despite impaired tumour cell killing. Tumours developing in these conditions were characterised by a high-density network of immature vessels, severe haemorrhage, increased hypoxia, and facilitated metastasis due to non-productive angiogenesis. Loss of HIF-1α in NK cells increased the bioavailability of the major angiogenic cytokine vascular endothelial growth factor (VEGF) by decreasing the infiltration of NK cells that express angiostatic soluble VEGFR-1. In summary, this identifies the hypoxic response in NK cells as an inhibitor of VEGF-driven angiogenesis, yet, this promotes tumour growth by allowing the formation of functionally improved vessels.


Sujet(s)
Sous-unité alpha du facteur-1 induit par l'hypoxie/déficit , Cellules tueuses naturelles/métabolisme , Tumeurs expérimentales/métabolisme , Néovascularisation pathologique/métabolisme , Animaux , Hypoxie cellulaire , Lignée cellulaire tumorale , Cellules cultivées , Hypoxie , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Souris de lignée C57BL , Souris knockout , Tumeurs expérimentales/vascularisation , Tumeurs expérimentales/génétique , Néovascularisation pathologique/génétique , Facteur de croissance endothéliale vasculaire de type A/déficit , Facteur de croissance endothéliale vasculaire de type A/génétique
20.
J Immunol ; 199(6): 1967-1972, 2017 09 15.
Article de Anglais | MEDLINE | ID: mdl-28784848

RÉSUMÉ

Priming of human NK cells with IL-2 is necessary to render them functionally competent upon NKG2D engagement. We examined the underlying mechanisms that control NKG2D responsiveness in NK cells and found that IL-2 upregulates expression of the amino acid transporters SLC1A5 and CD98. Using specific inhibitors to block SLC1A5 and CD98 function, we found that production of IFN-γ and degranulation by CD56bright and CD56dim NK cells following NKG2D stimulation were dependent on both transporters. IL-2 priming increased the activity of mTORC1, and inhibition of mTORC1 abrogated the ability of the IL-2-primed NK cells to produce IFN-γ in response to NKG2D-mediated stimulation. This study identifies a series of IL-2-induced cellular changes that regulates the NKG2D responsiveness in human NK cells.


Sujet(s)
Système ASC de transport d'acides aminés/métabolisme , Antigènes CD98/métabolisme , Cellules tueuses naturelles/physiologie , Antigènes mineurs d'histocompatibilité/métabolisme , Système ASC de transport d'acides aminés/génétique , Antigènes CD56/métabolisme , Cellules cultivées , Cytotoxicité immunologique , Humains , Interféron gamma/métabolisme , Interleukine-2/immunologie , Interleukine-2/métabolisme , Activation des lymphocytes , Complexe-1 cible mécanistique de la rapamycine , Antigènes mineurs d'histocompatibilité/génétique , Complexes multiprotéiques/antagonistes et inhibiteurs , Sous-famille K des récepteurs de cellules NK de type lectine/métabolisme , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...