Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
Plus de filtres










Base de données
Gamme d'année
2.
Science ; 383(6679): eadf6493, 2024 01 12.
Article de Anglais | MEDLINE | ID: mdl-38207030

RÉSUMÉ

Neutrophils are increasingly recognized as key players in the tumor immune response and are associated with poor clinical outcomes. Despite recent advances characterizing the diversity of neutrophil states in cancer, common trajectories and mechanisms governing the ontogeny and relationship between these neutrophil states remain undefined. Here, we demonstrate that immature and mature neutrophils that enter tumors undergo irreversible epigenetic, transcriptional, and proteomic modifications to converge into a distinct, terminally differentiated dcTRAIL-R1+ state. Reprogrammed dcTRAIL-R1+ neutrophils predominantly localize to a glycolytic and hypoxic niche at the tumor core and exert pro-angiogenic function that favors tumor growth. We found similar trajectories in neutrophils across multiple tumor types and in humans, suggesting that targeting this program may provide a means of enhancing certain cancer immunotherapies.


Sujet(s)
Reprogrammation cellulaire , Tumeurs , Néovascularisation pathologique , Granulocytes neutrophiles , Humains , Tumeurs/vascularisation , Tumeurs/immunologie , Granulocytes neutrophiles/immunologie , Protéomique , Reprogrammation cellulaire/génétique , Reprogrammation cellulaire/immunologie , Néovascularisation pathologique/génétique , Néovascularisation pathologique/immunologie , Récepteurs de TRAIL/immunologie , Épigenèse génétique , Hypoxie , Transcription génétique
3.
Immunity ; 56(1): 143-161.e11, 2023 01 10.
Article de Anglais | MEDLINE | ID: mdl-36630913

RÉSUMÉ

Although T cells can exert potent anti-tumor immunity, a subset of T helper (Th) cells producing interleukin-22 (IL-22) in breast and lung tumors is linked to dismal patient outcome. Here, we examined the mechanisms whereby these T cells contribute to disease. In murine models of lung and breast cancer, constitutional and T cell-specific deletion of Il22 reduced metastases without affecting primary tumor growth. Deletion of the IL-22 receptor on cancer cells decreases metastasis to a degree similar to that seen in IL-22-deficient mice. IL-22 induced high expression of CD155, which bound to the activating receptor CD226 on NK cells. Excessive activation led to decreased amounts of CD226 and functionally impaired NK cells, which elevated the metastatic burden. IL-22 signaling was also associated with CD155 expression in human datasets and with poor patient outcomes. Taken together, our findings reveal an immunosuppressive circuit activated by T cell-derived IL-22 that promotes lung metastasis.


Sujet(s)
Interleukines , Tumeurs , Récepteurs viraux , Lymphocytes T auxiliaires , Animaux , Humains , Souris , Antigènes de différenciation des lymphocytes T/métabolisme , Interleukines/génétique , Interleukines/métabolisme , Cellules tueuses naturelles/métabolisme , Tumeurs/métabolisme , Liaison aux protéines , Lymphocytes T auxiliaires/métabolisme ,
5.
Cancer Cell ; 40(12): 1459-1461, 2022 12 12.
Article de Anglais | MEDLINE | ID: mdl-36400019

RÉSUMÉ

Lung cancers are very heterogeneous, a feature that in part emanates from the tumor microenvironment. In this issue, Salcher et al. provide a comprehensive analysis of hundreds of patients with non-small cell lung cancer (NSCLC) at the single-cell level to discover extreme immune diversity and define neutrophil populations associated with treatment outcome.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/anatomopathologie , Microenvironnement tumoral
6.
FEBS J ; 289(13): 3692-3703, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-33999496

RÉSUMÉ

Neutrophils dominate the immunological landscape of multiple types of solid tumours in mice and humans and exert different pro- or antitumoral activity. This functional heterogeneity has prompted a search for different subsets and classifications of tumour-infiltrating neutrophils with the idea of better delineating their specific roles in cancer. In this review, we describe current studies that highlight specific mechanisms by which neutrophils exert pro- or antitumoral function and focus on how distinct tumour types induce unique functional states in neutrophils, co-opt granulopoiesis, modulate neutrophil ageing and prolong the neutrophil life span. In addition, we discuss how the tissue-specific tumour stroma and the stage of the cancer influence the function and number of tumour-infiltrating neutrophils. Finally, we explore different approaches to enhance the therapeutic efficacy in cancer types dominated by neutrophils.


Sujet(s)
Tumeurs , Granulocytes neutrophiles , Animaux , Souris , Tumeurs/anatomopathologie
7.
EMBO Mol Med ; 13(12): e12924, 2021 12 07.
Article de Anglais | MEDLINE | ID: mdl-34762341

RÉSUMÉ

Long-range communication between tumor cells and the lymphatic vasculature defines competency for metastasis in different cancer types, particularly in melanoma. Nevertheless, the discovery of selective blockers of lymphovascular niches has been compromised by the paucity of experimental systems for whole-body analyses of tumor progression. Here, we exploit immunocompetent and immunodeficient mouse models for live imaging of Vegfr3-driven neolymphangiogenesis, as a versatile platform for drug screening in vivo. Spatiotemporal analyses of autochthonous melanomas and patient-derived xenografts identified double-stranded RNA mimics (dsRNA nanoplexes) as potent inhibitors of neolymphangiogenesis, metastasis, and post-surgical disease relapse. Mechanistically, dsRNA nanoplexes were found to exert a rapid dual action in tumor cells and in their associated lymphatic vasculature, involving the transcriptional repression of the lymphatic drivers Midkine and Vegfr3, respectively. This suppressive function was mediated by a cell-autonomous type I interferon signaling and was not shared by FDA-approved antimelanoma treatments. These results reveal an alternative strategy for targeting the tumor cell-lymphatic crosstalk and underscore the power of Vegfr3-lymphoreporters for pharmacological testing in otherwise aggressive cancers.


Sujet(s)
Mélanome , ARN double brin , Animaux , Humains , Mélanome/traitement médicamenteux , Mélanome/anatomopathologie , Souris , Souris nude , Transduction du signal
8.
Adv Drug Deliv Rev ; 175: 113833, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34147531

RÉSUMÉ

Imaging of the lymphatic vasculature has gained great attention in various fields, not only because lymphatic vessels act as a key draining system in the body, but also for their implication in autoimmune diseases, organ transplant, inflammation and cancer. Thus, neolymphangiogenesis, or the generation of new lymphatics, is typically an early event in the development of multiple tumor types, particularly in aggressive ones such as malignant melanoma. Still, the understanding of how lymphatic endothelial cells get activated at distal (pre)metastatic niches and their impact on therapy is still unclear. Addressing these questions is of particular interest in the case of immune modulators, because endothelial cells may favor or halt inflammatory processes depending on the cellular context. Therefore, there is great interest in visualizing the lymphatic vasculature in vivo. Here, we review imaging tools and mouse models used to analyze the lymphatic vasculature during tumor progression. We also discuss therapeutic approaches based on nanomedicines to target the lymphatic system and the potential use of extracellular vesicles to track and target sentinel lymph nodes. Finally, we summarize main pre-clinical models developed to visualize the lymphatic vasculature in vivo, discussing their applications with a particular focus in metastatic melanoma.


Sujet(s)
Vésicules extracellulaires/métabolisme , Système lymphatique/imagerie diagnostique , Système d'administration de médicaments à base de nanoparticules , Animaux , Vésicules extracellulaires/anatomopathologie , Humains , Noeuds lymphatiques/imagerie diagnostique , Noeuds lymphatiques/effets des médicaments et des substances chimiques , Noeuds lymphatiques/anatomopathologie , Système lymphatique/effets des médicaments et des substances chimiques , Système lymphatique/anatomopathologie , Vaisseaux lymphatiques/imagerie diagnostique , Vaisseaux lymphatiques/effets des médicaments et des substances chimiques , Vaisseaux lymphatiques/anatomopathologie , Noeud lymphatique sentinelle/imagerie diagnostique , Noeud lymphatique sentinelle/anatomopathologie
9.
Nat Med ; 26(12): 1865-1877, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-33077955

RÉSUMÉ

An open question in aggressive cancers such as melanoma is how malignant cells can shift the immune system to pro-tumorigenic functions. Here we identify midkine (MDK) as a melanoma-secreted driver of an inflamed, but immune evasive, microenvironment that defines poor patient prognosis and resistance to immune checkpoint blockade. Mechanistically, MDK was found to control the transcriptome of melanoma cells, allowing for coordinated activation of nuclear factor-κB and downregulation of interferon-associated pathways. The resulting MDK-modulated secretome educated macrophages towards tolerant phenotypes that promoted CD8+ T cell dysfunction. In contrast, genetic targeting of MDK sensitized melanoma cells to anti-PD-1/anti-PD-L1 treatment. Emphasizing the translational relevance of these findings, the expression profile of MDK-depleted tumors was enriched in key indicators of a good response to immune checkpoint blockers in independent patient cohorts. Together, these data reveal that MDK acts as an internal modulator of autocrine and paracrine signals that maintain immune suppression in aggressive melanomas.


Sujet(s)
Carcinogenèse/effets des médicaments et des substances chimiques , Mélanome expérimental/thérapie , Midkine/génétique , Microenvironnement tumoral/génétique , Animaux , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/génétique , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/génétique , Thérapie génétique , Humains , Mélanome expérimental/génétique , Mélanome expérimental/anatomopathologie , Souris , Midkine/pharmacologie , Facteur de transcription NF-kappa B/génétique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/génétique , Protéines recombinantes/génétique , Protéines recombinantes/pharmacologie , Transcriptome/génétique
11.
Sci Rep ; 8(1): 3357, 2018 02 20.
Article de Anglais | MEDLINE | ID: mdl-29463868

RÉSUMÉ

In this study, we investigated the role of CD38 in a pristane-induced murine model of lupus. CD38-deficient (Cd38-/-) but not ART2-deficient (Art2-/-) mice developed less severe lupus compared to wild type (WT) mice, and their protective phenotype consisted of (i) decreased IFN-I-stimulated gene expression, (ii) decreased numbers of peritoneal CCR2hiLy6Chi inflammatory monocytes, TNF-α-producing Ly6G+ neutrophils and Ly6Clo monocytes/macrophages, (iii) decreased production of anti-single-stranded DNA and anti-nRNP autoantibodies, and (iv) ameliorated glomerulonephritis. Cd38-/- pristane-elicited peritoneal exudate cells had defective CCL2 and TNF-α secretion following TLR7 stimulation. However, Tnf-α and Cxcl12 gene expression in Cd38-/- bone marrow (BM) cells was intact, suggesting a CD38-independent TLR7/TNF-α/CXCL12 axis in the BM. Chemotactic responses of Cd38-/- Ly6Chi monocytes and Ly6G+ neutrophils were not impaired. However, Cd38-/- Ly6Chi monocytes and Ly6Clo monocytes/macrophages had defective apoptosis-mediated cell death. Importantly, mice lacking the cation channel TRPM2 (Trpm2-/-) exhibited very similar protection, with decreased numbers of PECs, and apoptotic Ly6Chi monocytes and Ly6Clo monocytes/macrophages compared to WT mice. These findings reveal a new role for CD38 in promoting aberrant inflammation and lupus-like autoimmunity via an apoptosis-driven mechanism. Furthermore, given the implications of CD38 in the activation of TRPM2, our data suggest that CD38 modulation of pristane-induced apoptosis is TRPM2-dependent.


Sujet(s)
Antigènes CD38/métabolisme , Apoptose , Immunosuppresseurs/pharmacologie , Lupus érythémateux cutané/induit chimiquement , Lupus érythémateux cutané/anatomopathologie , Glycoprotéines membranaires/métabolisme , Canaux cationiques TRPM/métabolisme , Terpènes/pharmacologie , ADP ribose transferases/déficit , ADP ribose transferases/métabolisme , Antigènes CD38/déficit , Animaux , Modèles animaux de maladie humaine , Prédisposition aux maladies , Facteurs immunologiques/métabolisme , Leucocytes/immunologie , Glycoprotéines membranaires/déficit , Souris
12.
Nature ; 546(7660): 676-680, 2017 06 28.
Article de Anglais | MEDLINE | ID: mdl-28658220

RÉSUMÉ

Cutaneous melanoma is a type of cancer with an inherent potential for lymph node colonization, which is generally preceded by neolymphangiogenesis. However, sentinel lymph node removal does not necessarily extend the overall survival of patients with melanoma. Moreover, lymphatic vessels collapse and become dysfunctional as melanomas progress. Therefore, it is unclear whether (and how) lymphangiogenesis contributes to visceral metastasis. Soluble and vesicle-associated proteins secreted by tumours and/or their stroma have been proposed to condition pre-metastatic sites in patients with melanoma. Still, the identities and prognostic value of lymphangiogenic mediators remain unclear. Moreover, our understanding of lymphangiogenesis (in melanomas and other tumour types) is limited by the paucity of mouse models for live imaging of distal pre-metastatic niches. Injectable lymphatic tracers have been developed, but their limited diffusion precludes whole-body imaging at visceral sites. Vascular endothelial growth factor receptor 3 (VEGFR3) is an attractive 'lymphoreporter' because its expression is strongly downregulated in normal adult lymphatic endothelial cells, but is activated in pathological situations such as inflammation and cancer. Here, we exploit this inducibility of VEGFR3 to engineer mouse melanoma models for whole-body imaging of metastasis generated by human cells, clinical biopsies or endogenously deregulated oncogenic pathways. This strategy revealed early induction of distal pre-metastatic niches uncoupled from lymphangiogenesis at primary lesions. Analyses of the melanoma secretome and validation in clinical specimens showed that the heparin-binding factor midkine is a systemic inducer of neo-lymphangiogenesis that defines patient prognosis. This role of midkine was linked to a paracrine activation of the mTOR pathway in lymphatic endothelial cells. These data support the use of VEGFR3 reporter mice as a 'MetAlert' discovery platform for drivers and inhibitors of metastasis.


Sujet(s)
Cytokines/métabolisme , Vaisseaux lymphatiques/métabolisme , Métastase tumorale/imagerie diagnostique , Métastase tumorale/anatomopathologie , Imagerie du corps entier/méthodes , Animaux , Modèles animaux de maladie humaine , Évolution de la maladie , Cellules endothéliales/métabolisme , Femelle , Gènes rapporteurs , Humains , Lymphangiogenèse , Vaisseaux lymphatiques/anatomopathologie , Mâle , Mélanome/imagerie diagnostique , Mélanome/métabolisme , Mélanome/anatomopathologie , Souris , Midkine , Communication paracrine , Pronostic , Récidive , Reproductibilité des résultats , Sérine-thréonine kinases TOR/métabolisme , Récepteur-3 au facteur croissance endothéliale vasculaire/analyse , Récepteur-3 au facteur croissance endothéliale vasculaire/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
13.
Cancer Cell ; 30(5): 694-707, 2016 Nov 14.
Article de Anglais | MEDLINE | ID: mdl-27908735

RÉSUMÉ

RNA binding proteins (RBPs) modulate cancer progression through poorly understood mechanisms. Here we show that the RBP UNR/CSDE1 is overexpressed in melanoma tumors and promotes invasion and metastasis. iCLIP sequencing, RNA sequencing, and ribosome profiling combined with in silico studies unveiled sets of pro-metastatic factors coordinately regulated by UNR as part of RNA regulons. In addition to RNA steady-state levels, UNR was found to control many of its targets at the level of translation elongation/termination. Key pro-oncogenic targets of UNR included VIM and RAC1, as validated by loss- and gain-of-function studies. Our results identify UNR as an oncogenic modulator of melanoma progression, unravel the underlying molecular mechanisms, and identify potential targets for this therapeutically challenging malignancy.


Sujet(s)
Protéines de liaison à l'ADN/métabolisme , Mélanome/anatomopathologie , Protéines de liaison à l'ARN/métabolisme , Régulation positive , Vimentine/génétique , Protéine G rac1/génétique , Animaux , Protéines de liaison à l'ADN/génétique , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Humains , Mélanome/génétique , Mélanome/métabolisme , Souris , Invasion tumorale , Métastase tumorale , Transplantation tumorale , Maturation post-transcriptionnelle des ARN , Protéines de liaison à l'ARN/génétique , Ribosomes/génétique , Analyse de séquence d'ARN/méthodes
14.
Curr Pharm Des ; 22(41): 6234-6250, 2016.
Article de Anglais | MEDLINE | ID: mdl-27568731

RÉSUMÉ

For more than a century, scientists have tried to exploit the anti-tumoral potential of the immune system to treat cancer. However, clinical success was traditionally limited. Consequently, classical anti-neoplastic treatments such as surgery, radiotherapy, and chemotherapy used to be the first line of treatment. Fortunately, this scenario is changing, particularly with the identification of immune checkpoint inhibitors. Now more than ever, it becomes essential to understand how cancer cells modulate the immune system. In this review, we summarize main mechanisms of antigen presentation in the tumor microenvironment, a main event in the establishment of anti-tumor immunity. Specifically, we focus on recent progress in mechanisms involved in tumor antigen presentation, as well as on strategies cancer cells develop to favor immune suppressive and immune tolerant enironments. In addition, we briefly discuss current therapeutic approaches to overcome tumor immune evasion.


Sujet(s)
Présentation d'antigène/immunologie , Antigènes néoplasiques/immunologie , Tumeurs/thérapie , Humains , Tumeurs/immunologie , Microenvironnement tumoral/immunologie
15.
Cytokine ; 62(2): 232-43, 2013 May.
Article de Anglais | MEDLINE | ID: mdl-23538292

RÉSUMÉ

CD38 is a multifunctional protein possessing ADP-ribosyl cyclase activity responsible for both the synthesis and the degradation of several Ca(2+)-mobilizing second messengers. In mammals, CD38 also functions as a receptor. In this study CD38 expression in CD4(+), CD8(+), or CD25(+) T cells was significantly higher in systemic lupus erythematosus (SLE) patients than in Normal controls. Increased CD38 expression in SLE T cells correlated with plasma levels of Th2 (IL-4, IL-10, IL-13) and Th1 (IL-1ß, IL-12, IFN-γ, TNF-α) cytokines, and was more prevalent in clinically active SLE patients than in Normal controls. In contrast, elevated anti-CD38 IgG autoantibodies were more frequent in clinically quiescent SLE patients (SLEDAI=0) than in Normal controls, and correlated with moderate increased plasma levels of IL-10 and IFN-γ. However, clinically active SLE patients were mainly discriminated from quiescent SLE patients by increased levels of IL-10 and anti-dsDNA antibodies, with odds ratios (ORs) of 3.7 and 4.8, respectively. Increased frequency of anti-CD38 autoantibodies showed an inverse relationship with clinical activity (OR=0.43), and in particular with the frequency of anti-dsDNA autoantibodies (OR=0.21). Increased cell death occurred in CD38(+) Jurkat T cells treated with anti-CD38(+) SLE plasmas, and not in these cells treated with anti-CD38(-) SLE plasmas, or Normal plasmas. This effect did not occur in CD38-negative Jurkat T cells, suggesting that it could be attributed to anti-CD38 autoantibodies. These results support the hypothesis that anti-CD38 IgG autoantibodies or their associated plasma factors may dampen immune activation by affecting the viability of CD38(+) effector T cells and may provide protection from certain clinical SLE features.


Sujet(s)
Antigènes CD38/immunologie , Autoanticorps/sang , Immunoglobuline G/immunologie , Lupus érythémateux disséminé/immunologie , Lupus érythémateux disséminé/métabolisme , Sous-populations de lymphocytes T/immunologie , Antigènes CD38/biosynthèse , Anticorps antinucléaires/sang , Anticorps antinucléaires/immunologie , Autoanticorps/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Cytokines/biosynthèse , Cytokines/sang , Femelle , Humains , Immunoglobuline G/sang , Sous-unité alpha du récepteur à l'interleukine-2/métabolisme , Cellules Jurkat , Lupus érythémateux disséminé/sang , Activation des lymphocytes , Numération des lymphocytes , Mâle , Phénotype , Sous-populations de lymphocytes T/métabolisme
16.
PLoS One ; 7(3): e33534, 2012.
Article de Anglais | MEDLINE | ID: mdl-22438945

RÉSUMÉ

CD38, a type II transmembrane glycoprotein expressed in many cells of the immune system, is involved in cell signaling, migration and differentiation. Studies in CD38 deficient mice (CD38 KO mice) indicate that this molecule controls inflammatory immune responses, although its involvement in these responses depends on the disease model analyzed. Here, we explored the role of CD38 in the control of autoimmune responses using chicken collagen type II (col II) immunized C57BL/6-CD38 KO mice as a model of collagen-induced arthritis (CIA). We demonstrate that CD38 KO mice develop an attenuated CIA that is accompanied by a limited joint induction of IL-1ß and IL-6 expression, by the lack of induction of IFNγ expression in the joints and by a reduction in the percentages of invariant NKT (iNKT) cells in the spleen. Immunized CD38 KO mice produce high levels of circulating IgG1 and low of IgG2a anti-col II antibodies in association with reduced percentages of Th1 cells in the draining lymph nodes. Altogether, our results show that CD38 participates in the pathogenesis of CIA controlling the number of iNKT cells and promoting Th1 inflammatory responses.


Sujet(s)
Antigènes CD38/déficit , Arthrite expérimentale/immunologie , Glycoprotéines membranaires/déficit , Antigènes CD38/génétique , Antigènes CD38/immunologie , Animaux , Anticorps hétérophiles/sang , Arthrite expérimentale/génétique , Arthrite expérimentale/anatomopathologie , Poulets , Collagène de type II/immunologie , Cytokines/génétique , Expression des gènes , Immunoglobuline G/sang , Interféron gamma/biosynthèse , Interféron gamma/génétique , Interleukine-1 bêta/génétique , Interleukine-6/génétique , Mâle , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/immunologie , Souris , Souris de lignée C57BL , Souris knockout , Cellules T tueuses naturelles/immunologie , ARN messager/génétique , ARN messager/métabolisme , Rate/immunologie , Lymphocytes auxiliaires Th1/immunologie , Cellules Th17/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...