Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Blood ; 143(12): 1124-1138, 2024 Mar 21.
Article de Anglais | MEDLINE | ID: mdl-38153903

RÉSUMÉ

ABSTRACT: The CD161 inhibitory receptor is highly upregulated by tumor-infiltrating T cells in multiple human solid tumor types, and its ligand, CLEC2D, is expressed by both tumor cells and infiltrating myeloid cells. Here, we assessed the role of the CD161 receptor in hematological malignancies. Systematic analysis of CLEC2D expression using the Cancer Cell Line Encyclopedia revealed that CLEC2D messenger RNA was most abundant in hematological malignancies, including B-cell and T-cell lymphomas as well as lymphocytic and myelogenous leukemias. CLEC2D protein was detected by flow cytometry on a panel of cell lines representing a diverse set of hematological malignancies. We, therefore, used yeast display to generate a panel of high-affinity, fully human CD161 monoclonal antibodies (mAbs) that blocked CLEC2D binding. These mAbs were specific for CD161 and had a similar affinity for human and nonhuman primate CD161, a property relevant for clinical translation. A high-affinity CD161 mAb enhanced key aspects of T-cell function, including cytotoxicity, cytokine production, and proliferation, against B-cell lines originating from patients with acute lymphoblastic leukemia, diffuse large B-cell lymphoma, and Burkitt lymphoma. In humanized mouse models, this CD161 mAb enhanced T-cell-mediated immunity, resulting in a significant survival benefit. Single cell RNA-seq data demonstrated that CD161 mAb treatment enhanced expression of cytotoxicity genes by CD4 T cells as well as a tissue-residency program by CD4 and CD8 T cells that is associated with favorable survival outcomes in multiple human cancer types. These fully human mAbs, thus, represent potential immunotherapy agents for hematological malignancies.


Sujet(s)
Tumeurs hématologiques , Tumeurs , Animaux , Souris , Humains , Lymphocytes T CD4+ , Immunité cellulaire , Lymphocytes T CD8+ , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Tumeurs hématologiques/génétique , Tumeurs hématologiques/thérapie , Sous-famille B des récepteurs de cellules NK de type lectine/génétique
2.
Cell Rep ; 42(12): 113564, 2023 12 26.
Article de Anglais | MEDLINE | ID: mdl-38100350

RÉSUMÉ

Triple-negative breast cancer (TNBC) is a heterogeneous disease with limited treatment options. To characterize TNBC heterogeneity, we defined transcriptional, epigenetic, and metabolic subtypes and subtype-driving super-enhancers and transcription factors by combining functional and molecular profiling with computational analyses. Single-cell RNA sequencing revealed relative homogeneity of the major transcriptional subtypes (luminal, basal, and mesenchymal) within samples. We found that mesenchymal TNBCs share features with mesenchymal neuroblastoma and rhabdoid tumors and that the PRRX1 transcription factor is a key driver of these tumors. PRRX1 is sufficient for inducing mesenchymal features in basal but not in luminal TNBC cells via reprogramming super-enhancer landscapes, but it is not required for mesenchymal state maintenance or for cellular viability. Our comprehensive, large-scale, multiplatform, multiomics study of both experimental and clinical TNBC is an important resource for the scientific and clinical research communities and opens venues for future investigation.


Sujet(s)
Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/anatomopathologie , Facteurs de transcription/métabolisme , Régulation de l'expression des gènes tumoraux , Protéines à homéodomaine/métabolisme
3.
Cancer Discov ; 13(5): 1186-1209, 2023 05 04.
Article de Anglais | MEDLINE | ID: mdl-36811466

RÉSUMÉ

Tumor heterogeneity is a major barrier to cancer therapy, including immunotherapy. Activated T cells can efficiently kill tumor cells following recognition of MHC class I (MHC-I)-bound peptides, but this selection pressure favors outgrowth of MHC-I-deficient tumor cells. We performed a genome-scale screen to discover alternative pathways for T cell-mediated killing of MHC-I-deficient tumor cells. Autophagy and TNF signaling emerged as top pathways, and inactivation of Rnf31 (TNF signaling) and Atg5 (autophagy) sensitized MHC-I-deficient tumor cells to apoptosis by T cell-derived cytokines. Mechanistic studies demonstrated that inhibition of autophagy amplified proapoptotic effects of cytokines in tumor cells. Antigens from apoptotic MHC-I-deficient tumor cells were efficiently cross-presented by dendritic cells, resulting in heightened tumor infiltration by IFNγ-and TNFα-producing T cells. Tumors with a substantial population of MHC-I-deficient cancer cells could be controlled by T cells when both pathways were targeted using genetic or pharmacologic approaches. SIGNIFICANCE: Tumor heterogeneity is a major barrier to immunotherapy. We show that MHC-I-deficient tumor cells are forced into apoptosis by T cell-derived cytokines when TNF signaling and autophagy pathways are targeted. This approach enables T cell-mediated elimination of tumors with a substantial population of resistant, MHC-I-deficient tumor cells. This article is highlighted in the In This Issue feature, p. 1027.


Sujet(s)
Tumeurs , Lymphocytes T , Humains , Cytokines , Antigènes d'histocompatibilité de classe I/métabolisme , Tumeurs/génétique , Tumeurs/thérapie , Gènes MHC de classe I
4.
Proc Natl Acad Sci U S A ; 120(1): e2213222120, 2023 01 03.
Article de Anglais | MEDLINE | ID: mdl-36577059

RÉSUMÉ

Adoptive T cell transfer (ACT) therapies suffer from a number of limitations (e.g., poor control of solid tumors), and while combining ACT with cytokine therapy can enhance effectiveness, this also results in significant side effects. Here, we describe a nanotechnology approach to improve the efficacy of ACT therapies by metabolically labeling T cells with unnatural sugar nanoparticles, allowing direct conjugation of antitumor cytokines onto the T cell surface during the manufacturing process. This allows local, concentrated activity of otherwise toxic cytokines. This approach increases T cell infiltration into solid tumors, activates the host immune system toward a Type 1 response, encourages antigen spreading, and improves control of aggressive solid tumors and achieves complete blood cancer regression with otherwise noncurative doses of CAR-T cells. Overall, this method provides an effective and easily integrated approach to the current ACT manufacturing process to increase efficacy in various settings.


Sujet(s)
Cytokines , Tumeurs , Humains , Cytokines/métabolisme , Immunothérapie adoptive/méthodes , Récepteurs aux antigènes des cellules T , Lymphocytes T , Tumeurs/anatomopathologie , Thérapie cellulaire et tissulaire
5.
Nat Commun ; 13(1): 7558, 2022 12 07.
Article de Anglais | MEDLINE | ID: mdl-36476730

RÉSUMÉ

Cancer prevention has a profound impact on cancer-associated mortality and morbidity. We previously identified TGFß signaling as a candidate regulator of mammary epithelial cells associated with breast cancer risk. Here, we show that short-term TGFBR inhibitor (TGFBRi) treatment of peripubertal ACI inbred and Sprague Dawley outbred rats induces lasting changes and prevents estrogen- and carcinogen-induced mammary tumors, respectively. We identify TGFBRi-responsive cell populations by single cell RNA-sequencing, including a unique epithelial subpopulation designated secretory basal cells (SBCs) with progenitor features. We detect SBCs in normal human breast tissues and find them to be associated with breast cancer risk. Interactome analysis identifies SBCs as the most interactive cell population and the main source of insulin-IGF signaling. Accordingly, inhibition of TGFBR and IGF1R decrease proliferation of organoid cultures. Our results reveal a critical role for TGFß in regulating mammary epithelial cells relevant to breast cancer and serve as a proof-of-principle cancer prevention strategy.


Sujet(s)
Tumeurs , Rats , Humains , Animaux , Rats de lignée ACI , Rat Sprague-Dawley
6.
Nature ; 606(7916): 992-998, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35614223

RÉSUMÉ

Most cancer vaccines target peptide antigens, necessitating personalization owing to the vast inter-individual diversity in major histocompatibility complex (MHC) molecules that present peptides to T cells. Furthermore, tumours frequently escape T cell-mediated immunity through mechanisms that interfere with peptide presentation1. Here we report a cancer vaccine that induces a coordinated attack by diverse T cell and natural killer (NK) cell populations. The vaccine targets the MICA and MICB (MICA/B) stress proteins expressed by many human cancers as a result of DNA damage2. MICA/B serve as ligands for the activating NKG2D receptor on T cells and NK cells, but tumours evade immune recognition by proteolytic MICA/B cleavage3,4. Vaccine-induced antibodies increase the density of MICA/B proteins on the surface of tumour cells by inhibiting proteolytic shedding, enhance presentation of tumour antigens by dendritic cells to T cells and augment the cytotoxic function of NK cells. Notably, this vaccine maintains efficacy against MHC class I-deficient tumours resistant to cytotoxic T cells through the coordinated action of NK cells and CD4+ T cells. The vaccine is also efficacious in a clinically important setting: immunization following surgical removal of primary, highly metastatic tumours inhibits the later outgrowth of metastases. This vaccine design enables protective immunity even against tumours with common escape mutations.


Sujet(s)
Syndromes myélodysplasiques , Tumeurs , Maladies génétiques de la peau , Vaccins , Antigènes d'histocompatibilité de classe I , Humains , Cellules tueuses naturelles , Sous-famille K des récepteurs de cellules NK de type lectine/métabolisme , Tumeurs/prévention et contrôle
7.
Cancer Discov ; 11(8): 2050-2071, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33707234

RÉSUMÉ

A number of cancer drugs activate innate immune pathways in tumor cells but unfortunately also compromise antitumor immune function. We discovered that inhibition of CARM1, an epigenetic enzyme and cotranscriptional activator, elicited beneficial antitumor activity in both cytotoxic T cells and tumor cells. In T cells, Carm1 inactivation substantially enhanced their antitumor function and preserved memory-like populations required for sustained antitumor immunity. In tumor cells, Carm1 inactivation induced a potent type 1 interferon response that sensitized resistant tumors to cytotoxic T cells. Substantially increased numbers of dendritic cells, CD8 T cells, and natural killer cells were present in Carm1-deficient tumors, and infiltrating CD8 T cells expressed low levels of exhaustion markers. Targeting of CARM1 with a small molecule elicited potent antitumor immunity and sensitized resistant tumors to checkpoint blockade. Targeting of this cotranscriptional regulator thus offers an opportunity to enhance immune function while simultaneously sensitizing resistant tumor cells to immune attack. SIGNIFICANCE: Resistance to cancer immunotherapy remains a major challenge. Targeting of CARM1 enables immunotherapy of resistant tumors by enhancing T-cell functionality and preserving memory-like T-cell populations within tumors. CARM1 inhibition also sensitizes resistant tumor cells to immune attack by inducing a tumor cell-intrinsic type 1 interferon response.This article is highlighted in the In This Issue feature, p. 1861.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs/thérapie , Protein-arginine N-methyltransferases/antagonistes et inhibiteurs , Lignée cellulaire tumorale/effets des médicaments et des substances chimiques , Humains , Immunothérapie , Lymphocytes T/effets des médicaments et des substances chimiques
8.
Cell ; 184(5): 1281-1298.e26, 2021 03 04.
Article de Anglais | MEDLINE | ID: mdl-33592174

RÉSUMÉ

T cells are critical effectors of cancer immunotherapies, but little is known about their gene expression programs in diffuse gliomas. Here, we leverage single-cell RNA sequencing (RNA-seq) to chart the gene expression and clonal landscape of tumor-infiltrating T cells across 31 patients with isocitrate dehydrogenase (IDH) wild-type glioblastoma and IDH mutant glioma. We identify potential effectors of anti-tumor immunity in subsets of T cells that co-express cytotoxic programs and several natural killer (NK) cell genes. Analysis of clonally expanded tumor-infiltrating T cells further identifies the NK gene KLRB1 (encoding CD161) as a candidate inhibitory receptor. Accordingly, genetic inactivation of KLRB1 or antibody-mediated CD161 blockade enhances T cell-mediated killing of glioma cells in vitro and their anti-tumor function in vivo. KLRB1 and its associated transcriptional program are also expressed by substantial T cell populations in other human cancers. Our work provides an atlas of T cells in gliomas and highlights CD161 and other NK cell receptors as immunotherapy targets.


Sujet(s)
Gliome/immunologie , Sous-famille B des récepteurs de cellules NK de type lectine/génétique , Lymphocytes T/immunologie , Animaux , Antigènes néoplasiques , Modèles animaux de maladie humaine , Analyse de profil d'expression de gènes , Gliome/génétique , Cellules tueuses naturelles/immunologie , Lectines de type C/génétique , Lymphocytes TIL/immunologie , Souris , Récepteurs de surface cellulaire/génétique , Analyse sur cellule unique , Sous-populations de lymphocytes T/immunologie , Lymphocytes T/cytologie , Échappement de la tumeur à la surveillance immunitaire
9.
Nat Med ; 27(3): 515-525, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33479501

RÉSUMÉ

Personal neoantigen vaccines have been envisioned as an effective approach to induce, amplify and diversify antitumor T cell responses. To define the long-term effects of such a vaccine, we evaluated the clinical outcome and circulating immune responses of eight patients with surgically resected stage IIIB/C or IVM1a/b melanoma, at a median of almost 4 years after treatment with NeoVax, a long-peptide vaccine targeting up to 20 personal neoantigens per patient ( NCT01970358 ). All patients were alive and six were without evidence of active disease. We observed long-term persistence of neoantigen-specific T cell responses following vaccination, with ex vivo detection of neoantigen-specific T cells exhibiting a memory phenotype. We also found diversification of neoantigen-specific T cell clones over time, with emergence of multiple T cell receptor clonotypes exhibiting distinct functional avidities. Furthermore, we detected evidence of tumor infiltration by neoantigen-specific T cell clones after vaccination and epitope spreading, suggesting on-target vaccine-induced tumor cell killing. Personal neoantigen peptide vaccines thus induce T cell responses that persist over years and broaden the spectrum of tumor-specific cytotoxicity in patients with melanoma.


Sujet(s)
Antigènes néoplasiques/génétique , Vaccins anticancéreux/immunologie , Épitopes/immunologie , Mémoire immunologique , Mélanome/immunologie , Humains , Mélanome/anatomopathologie
10.
Cancer Cell ; 39(1): 54-67.e9, 2021 01 11.
Article de Anglais | MEDLINE | ID: mdl-33385331

RÉSUMÉ

Cancer immunotherapy shows limited efficacy against many solid tumors that originate from epithelial tissues, including triple-negative breast cancer (TNBC). We identify the SOX4 transcription factor as an important resistance mechanism to T cell-mediated cytotoxicity for TNBC cells. Mechanistic studies demonstrate that inactivation of SOX4 in tumor cells increases the expression of genes in a number of innate and adaptive immune pathways important for protective tumor immunity. Expression of SOX4 is regulated by the integrin αvß6 receptor on the surface of tumor cells, which activates TGFß from a latent precursor. An integrin αvß6/8-blocking monoclonal antibody (mAb) inhibits SOX4 expression and sensitizes TNBC cells to cytotoxic T cells. This integrin mAb induces a substantial survival benefit in highly metastatic murine TNBC models poorly responsive to PD-1 blockade. Targeting of the integrin αvß6-TGFß-SOX4 pathway therefore provides therapeutic opportunities for TNBC and other highly aggressive human cancers of epithelial origin.


Sujet(s)
Anticorps monoclonaux/administration et posologie , Antigènes néoplasiques/génétique , Antinéoplasiques immunologiques/usage thérapeutique , Intégrines/génétique , Facteurs de transcription SOX-C/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs du sein triple-négatives/traitement médicamenteux , Échappement de la tumeur à la surveillance immunitaire , Animaux , Anticorps monoclonaux/pharmacologie , Antigènes néoplasiques/métabolisme , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Intégrines/antagonistes et inhibiteurs , Intégrines/métabolisme , Souris , Transplantation tumorale , Facteurs de transcription SOX-C/métabolisme , Analyse de séquence d'ARN , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Lymphocytes T cytotoxiques/métabolisme , Facteur de croissance transformant bêta/génétique , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/immunologie , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Cancer Cell ; 38(1): 44-59.e9, 2020 07 13.
Article de Anglais | MEDLINE | ID: mdl-32663469

RÉSUMÉ

Ependymoma is a heterogeneous entity of central nervous system tumors with well-established molecular groups. Here, we apply single-cell RNA sequencing to analyze ependymomas across molecular groups and anatomic locations to investigate their intratumoral heterogeneity and developmental origins. Ependymomas are composed of a cellular hierarchy initiating from undifferentiated populations, which undergo impaired differentiation toward three lineages of neuronal-glial fate specification. While prognostically favorable groups of ependymoma predominantly harbor differentiated cells, aggressive groups are enriched for undifferentiated cell populations. The delineated transcriptomic signatures correlate with patient survival and define molecular dependencies for targeted treatment approaches. Taken together, our analyses reveal a developmental hierarchy underlying ependymomas relevant to biological and clinical behavior.


Sujet(s)
Tumeurs du système nerveux central/génétique , Épendymome/génétique , Analyse de séquence d'ARN/méthodes , Analyse sur cellule unique/méthodes , Différenciation cellulaire/génétique , Prolifération cellulaire/génétique , Tumeurs du système nerveux central/anatomopathologie , Tumeurs du système nerveux central/thérapie , Enfant , Épendymome/anatomopathologie , Épendymome/thérapie , Génomique/méthodes , Humains , Neurones/métabolisme , Neurones/anatomopathologie , Pronostic , Analyse de survie
12.
Cancer Immunol Res ; 8(6): 769-780, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32209637

RÉSUMÉ

Resistance to cytotoxic T cells is frequently mediated by loss of MHC class I expression or IFNγ signaling in tumor cells, such as mutations of B2M or JAK1 genes. Natural killer (NK) cells could potentially target such resistant tumors, but suitable NK-cell-based strategies remain to be developed. We hypothesized that such tumors could be targeted by NK cells if sufficient activating signals were provided. Human tumors frequently express the MICA and MICB ligands of the activating NKG2D receptor, but proteolytic shedding of MICA/B represents an important immune evasion mechanism in many human cancers. We showed that B2M- and JAK1-deficient metastases were targeted by NK cells following treatment with a mAb that blocks MICA/B shedding. We also demonstrated that the FDA-approved HDAC inhibitor panobinostat and a MICA/B antibody acted synergistically to enhance MICA/B surface expression on tumor cells. The HDAC inhibitor enhanced MICA/B gene expression, whereas the MICA/B antibody stabilized the synthesized protein on the cell surface. The combination of panobinostat and the MICA/B antibody reduced the number of pulmonary metastases formed by a human melanoma cell line in NOD/SCID gamma mice reconstituted with human NK cells. NK-cell-mediated immunity induced by a mAb specific for MICA/B, therefore, provides an opportunity to target tumors with mutations that render them resistant to cytotoxic T cells.


Sujet(s)
Antigènes d'histocompatibilité de classe I/composition chimique , Immunité cellulaire/immunologie , Cellules tueuses naturelles/immunologie , Tumeurs du poumon/thérapie , Mélanome/thérapie , Lymphocytes T cytotoxiques/immunologie , Animaux , Anticorps monoclonaux/pharmacologie , Apoptose , Prolifération cellulaire , Femelle , Régulation de l'expression des gènes tumoraux , Antigènes d'histocompatibilité de classe I/immunologie , Antigènes d'histocompatibilité de classe I/métabolisme , Humains , Tumeurs du poumon/immunologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/secondaire , Mélanome/immunologie , Mélanome/métabolisme , Mélanome/anatomopathologie , Souris , Souris de lignée C57BL , Souris de lignée NOD , Souris knockout , Souris SCID , Sous-famille K des récepteurs de cellules NK de type lectine/métabolisme , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
13.
JCI Insight ; 4(23)2019 12 05.
Article de Anglais | MEDLINE | ID: mdl-31801909

RÉSUMÉ

NK cells contribute to protective antitumor immunity, but little is known about the functional states of NK cells in human solid tumors. To address this issue, we performed single-cell RNA-seq analysis of NK cells isolated from human melanoma metastases, including lesions from patients who had progressed following checkpoint blockade. This analysis identified major differences in the transcriptional programs of tumor-infiltrating compared with circulating NK cells. Tumor-infiltrating NK cells represented 7 clusters with distinct gene expression programs indicative of significant functional specialization, including cytotoxicity and chemokine synthesis programs. In particular, NK cells from 3 clusters expressed high levels of XCL1 and XCL2, which encode 2 chemokines known to recruit XCR1+ cross-presenting DCs into tumors. In contrast, NK cells from 2 other clusters showed a higher level of expression of cytotoxicity genes. These data reveal key features of NK cells in human tumors and identify NK cell populations with specialized gene expression programs.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Cellules tueuses naturelles/immunologie , Lymphocytes TIL/immunologie , Mélanome/génétique , Mélanome/immunologie , Métastase tumorale/immunologie , Lignée cellulaire tumorale , Chimiokines/génétique , Chimiokines/métabolisme , Chimiokines C , Cross-priming , Humains , Immunité innée
14.
J Exp Med ; 215(10): 2617-2635, 2018 10 01.
Article de Anglais | MEDLINE | ID: mdl-30185635

RÉSUMÉ

A number of autoimmunity-associated MHC class II proteins interact only weakly with the invariant chain-derived class II-associated invariant chain peptide (CLIP). CLIP dissociates rapidly from I-Ag7 even in the absence of DM, and this property is related to the type 1 diabetes-associated ß57 polymorphism. We generated knock-in non-obese diabetic (NOD) mice with a single amino acid change in the CLIP segment of the invariant chain in order to moderately slow CLIP dissociation from I-Ag7 These knock-in mice had a significantly reduced incidence of spontaneous type 1 diabetes and diminished islet infiltration by CD4 T cells, in particular T cells specific for fusion peptides generated by covalent linkage of proteolytic fragments within ß cell secretory granules. Rapid CLIP dissociation enhanced the presentation of such extracellular peptides, thus bypassing the conventional MHC class II antigen-processing pathway. Autoimmunity-associated MHC class II polymorphisms therefore not only modify binding of self-peptides, but also alter the biochemistry of peptide acquisition.


Sujet(s)
Présentation d'antigène , Antigènes de différenciation des lymphocytes B/immunologie , Auto-immunité , Lymphocytes T CD4+/immunologie , Antigènes d'histocompatibilité de classe II/immunologie , Animaux , Antigènes de différenciation des lymphocytes B/génétique , Lymphocytes T CD4+/cytologie , Techniques de knock-in de gènes , Antigènes d'histocompatibilité de classe II/génétique , Souris , Souris de lignée NOD , Souris transgéniques
15.
Science ; 359(6383): 1537-1542, 2018 Mar 30.
Article de Anglais | MEDLINE | ID: mdl-29599246

RÉSUMÉ

MICA and MICB are expressed by many human cancers as a result of cellular stress, and can tag cells for elimination by cytotoxic lymphocytes through natural killer group 2D (NKG2D) receptor activation. However, tumors evade this immune recognition pathway through proteolytic shedding of MICA and MICB proteins. We rationally designed antibodies targeting the MICA α3 domain, the site of proteolytic shedding, and found that these antibodies prevented loss of cell surface MICA and MICB by human cancer cells. These antibodies inhibited tumor growth in multiple fully immunocompetent mouse models and reduced human melanoma metastases in a humanized mouse model. Antitumor immunity was mediated mainly by natural killer (NK) cells through activation of NKG2D and CD16 Fc receptors. This approach prevents the loss of important immunostimulatory ligands by human cancers and reactivates antitumor immunity.


Sujet(s)
Anticorps bloquants/usage thérapeutique , Anticorps monoclonaux/usage thérapeutique , Antigènes d'histocompatibilité de classe I/immunologie , Cellules tueuses naturelles/immunologie , Mélanome/thérapie , Animaux , Anticorps bloquants/immunologie , Anticorps monoclonaux/immunologie , Antigènes d'histocompatibilité de classe I/composition chimique , Humains , Immunocompétence , Ligands , Mélanome/immunologie , Mélanome/anatomopathologie , Mélanome expérimental/immunologie , Mélanome expérimental/anatomopathologie , Mélanome expérimental/thérapie , Souris , Souris de lignée C57BL , Sous-famille K des récepteurs de cellules NK de type lectine/immunologie , Métastase tumorale , Domaines protéiques/immunologie , Récepteurs du fragment Fc des IgG/immunologie
17.
Nat Immunol ; 18(7): 791-799, 2017 Jul.
Article de Anglais | MEDLINE | ID: mdl-28530712

RÉSUMÉ

During infection, antigen-specific T cells undergo tightly regulated developmental transitions controlled by transcriptional and post-transcriptional regulation of gene expression. We found that the microRNA miR-31 was strongly induced by activation of the T cell antigen receptor (TCR) in a pathway involving calcium and activation of the transcription factor NFAT. During chronic infection with lymphocytic choriomeningitis virus (LCMV) clone 13, miR-31-deficent mice recovered from clinical disease, while wild-type mice continued to show signs of disease. This disease phenotype was explained by the presence of larger numbers of cytokine-secreting LCMV-specific CD8+ T cells in miR-31-deficent mice than in wild-type mice. Mechanistically, miR-31 increased the sensitivity of T cells to type I interferons, which interfered with effector T cell function and increased the expression of several proteins related to T cell dysfunction during chronic infection. These studies identify miR-31 as an important regulator of T cell exhaustion in chronic infection.


Sujet(s)
Infections à Arenaviridae/immunologie , Lymphocytes T CD8+/immunologie , Cytokines/immunologie , microARN/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Animaux , Anticorps antiviraux/immunologie , Infections à Arenaviridae/génétique , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Calcium/métabolisme , Immunoprécipitation de la chromatine , Cellules dendritiques/immunologie , Test ELISA , Cytométrie en flux , Analyse de profil d'expression de gènes , Immunotransfert , Interféron de type I/pharmacologie , Virus de la chorioméningite lymphocytaire/immunologie , Souris , Souris knockout , microARN/génétique , Facteurs de transcription NFATC/métabolisme , Réaction de polymérisation en chaine en temps réel
18.
Sci Signal ; 9(438): ra75, 2016 07 26.
Article de Anglais | MEDLINE | ID: mdl-27460989

RÉSUMÉ

The T cell costimulatory receptor CD28 is required for the full activation of naïve T cells and for the development and maintenance of Foxp3(+) regulatory T (Treg) cells. We showed that the cytoplasmic domain of CD28 was bound to the plasma membrane in resting cells and that ligand binding to CD28 resulted in its release. Membrane binding by the CD28 cytoplasmic domain required two clusters of basic amino acid residues, which interacted with the negatively charged inner leaflet of the plasma membrane. These same clusters of basic residues also served as interaction sites for Lck, a Src family kinase critical for CD28 function. This signaling complex was further stabilized by the Lck-mediated phosphorylation of CD28 Tyr(207) and the subsequent binding of the Src homology 2 (SH2) domain of Lck to this phosphorylated tyrosine. Mutation of the basic clusters in the CD28 cytoplasmic domain reduced the recruitment to the CD28-Lck complex of protein kinase Cθ (PKCθ), which serves as a key effector kinase in the CD28 signaling pathway. Consequently, mutation of either a basic cluster or Tyr(207) impaired CD28 function in mice as shown by the reduced thymic differentiation of FoxP3(+) Treg cells. On the basis of these results, we propose a previously undescribed model for the initiation of CD28 signaling.


Sujet(s)
Antigène CD28/immunologie , Membrane cellulaire/immunologie , Protéine tyrosine kinase p56(lck) spécifique des lymphocytes/immunologie , Transduction du signal/immunologie , Lymphocytes T régulateurs/immunologie , Animaux , Antigène CD28/génétique , Membrane cellulaire/génétique , Humains , Cellules Jurkat , Protéine tyrosine kinase p56(lck) spécifique des lymphocytes/génétique , Souris , Phosphorylation/génétique , Phosphorylation/immunologie , Domaines protéiques , Protein kinase C-epsilon/génétique , Protein kinase C-epsilon/immunologie , Transduction du signal/génétique
19.
Cell ; 151(7): 1557-68, 2012 Dec 21.
Article de Anglais | MEDLINE | ID: mdl-23260142

RÉSUMÉ

HLA-DR molecules bind microbial peptides in an endosomal compartment and present them on the cell surface for CD4 T cell surveillance. HLA-DM plays a critical role in the endosomal peptide selection process. The structure of the HLA-DM-HLA-DR complex shows major rearrangements of the HLA-DR peptide-binding groove. Flipping of a tryptophan away from the HLA-DR1 P1 pocket enables major conformational changes that position hydrophobic HLA-DR residues into the P1 pocket. These conformational changes accelerate peptide dissociation and stabilize the empty HLA-DR peptide-binding groove. Initially, incoming peptides have access to only part of the HLA-DR groove and need to compete with HLA-DR residues for access to the P2 site and the hydrophobic P1 pocket. This energetic barrier creates a rapid and stringent selection process for the highest-affinity binders. Insertion of peptide residues into the P2 and P1 sites reverses the conformational changes, terminating selection through DM dissociation.


Sujet(s)
Antigènes HLA-D/composition chimique , Antigènes HLA-D/métabolisme , Antigène HLA-DR1/composition chimique , Antigène HLA-DR1/métabolisme , Séquence d'acides aminés , Animaux , Cristallographie aux rayons X , Humains , Interactions hydrophobes et hydrophiles , Modèles moléculaires , Données de séquences moléculaires , Conformation des protéines , Motifs et domaines d'intéraction protéique , Alignement de séquences
20.
J Exp Med ; 208(1): 91-102, 2011 Jan 17.
Article de Anglais | MEDLINE | ID: mdl-21199956

RÉSUMÉ

Self-reactive T cells that escape elimination in the thymus can cause autoimmune pathology, and it is therefore important to understand the structural mechanisms of self-antigen recognition. We report the crystal structure of a T cell receptor (TCR) from a patient with relapsing-remitting multiple sclerosis that engages its self-peptide-major histocompatibility complex (pMHC) ligand in an unusual manner. The TCR is bound in a highly tilted orientation that prevents interaction of the TCR-α chain with the MHC class II ß chain helix. In this structure, only a single germline-encoded TCR loop engages the MHC protein, whereas in most other TCR-pMHC structures all four germline-encoded TCR loops bind to the MHC helices. The tilted binding mode also prevents peptide contacts by the short complementarity-determining region (CDR) 3ß loop, and interactions that contribute to peptide side chain specificity are focused on the CDR3α loop. This structure is the first example in which only a single germline-encoded TCR loop contacts the MHC helices. Furthermore, the reduced interaction surface with the peptide may facilitate TCR cross-reactivity. The structural alterations in the trimolecular complex are distinct from previously characterized self-reactive TCRs, indicating that there are multiple unusual ways for self-reactive TCRs to bind their pMHC ligand.


Sujet(s)
Antigènes d'histocompatibilité/immunologie , Peptides/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Animaux , Antigènes d'histocompatibilité/composition chimique , Humains , Souris , Modèles moléculaires , Peptides/composition chimique , Structure tertiaire des protéines , Récepteurs aux antigènes des cellules T/composition chimique , Similitude structurale de protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...