Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.957
Filtrer
1.
Proc Natl Acad Sci U S A ; 121(34): e2321686121, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39141352

RÉSUMÉ

To broadly measure the spectrum of cellular self-antigens for natural killer T cells (NKT), we developed a sensitive lipidomics system to analyze lipids trapped between CD1d and NKT T cell receptors (TCRs). We captured diverse antigen complexes formed in cells from natural endogenous lipids, with or without inducing endoplasmic reticulum (ER) stress. After separating protein complexes with no, low, or high CD1d-TCR interaction, we eluted lipids to establish the spectrum of self-lipids that facilitate this interaction. Although this unbiased approach identified fifteen molecules, they clustered into only two related groups: previously known phospholipid antigens and unexpected neutral lipid antigens. Mass spectrometry studies identified the neutral lipids as ceramides, deoxyceramides, and diacylglycerols, which can be considered headless lipids because they lack polar headgroups that usually form the TCR epitope. The crystal structure of the TCR-ceramide-CD1d complex showed how the missing headgroup allowed the TCR to predominantly contact CD1d, supporting a model of CD1d autoreactivity. Ceramide and related headless antigens mediated physiological TCR binding affinity, weak NKT cell responses, and tetramer binding to polyclonal human and mouse NKT cells. Ceramide and sphingomyelin are oppositely regulated components of the "sphingomyelin cycle" that are altered during apoptosis, transformation, and ER stress. Thus, the unique molecular link of ceramide to NKT cell response, along with the recent identification of sphingomyelin blockers of NKT cell activation, provide two mutually reinforcing links for NKT cell response to sterile cellular stress conditions.


Sujet(s)
Antigène CD1d , Lipidomique , Cellules T tueuses naturelles , Récepteurs aux antigènes des cellules T , Cellules T tueuses naturelles/immunologie , Cellules T tueuses naturelles/métabolisme , Antigène CD1d/immunologie , Antigène CD1d/métabolisme , Animaux , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Souris , Lipidomique/méthodes , Humains , Autoantigènes/immunologie , Autoantigènes/métabolisme , Céramides/métabolisme , Céramides/immunologie , Lipides/composition chimique , Lipides/immunologie , Stress du réticulum endoplasmique/immunologie
2.
Brief Bioinform ; 25(5)2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39129361

RÉSUMÉ

Effective clustering of T-cell receptor (TCR) sequences could be used to predict their antigen-specificities. TCRs with highly dissimilar sequences can bind to the same antigen, thus making their clustering into a common antigen group a central challenge. Here, we develop TouCAN, a method that relies on contrastive learning and pretrained protein language models to perform TCR sequence clustering and antigen-specificity predictions. Following training, TouCAN demonstrates the ability to cluster highly dissimilar TCRs into common antigen groups. Additionally, TouCAN demonstrates TCR clustering performance and antigen-specificity predictions comparable to other leading methods in the field.


Sujet(s)
Récepteurs aux antigènes des cellules T , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/génétique , Analyse de regroupements , Humains , Antigènes/immunologie , Biologie informatique/méthodes , Algorithmes , Apprentissage machine
3.
Immunity ; 57(8): 1893-1907.e6, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39096910

RÉSUMÉ

Naive CD4+ T cells in specific pathogen-free (SPF) mice are characterized by transcriptional heterogeneity and subpopulations distinguished by the expression of quiescence, the extracellular matrix (ECM) and cytoskeleton, type I interferon (IFN-I) response, memory-like, and T cell receptor (TCR) activation genes. We demonstrate that this constitutive heterogeneity, including the presence of the IFN-I response cluster, is commensal independent insofar as being identical in germ-free and SPF mice. By contrast, Nippostrongylus brasiliensis infection altered this constitutive heterogeneity. Naive T cell-intrinsic transcriptional changes acquired during helminth infection correlated with and accounted for decreased immunization response to an unrelated antigen. These compositional and functional changes were dependent variables of helminth infection, as they disappeared at the established time point of its clearance in mice. Collectively, our results indicate that the naive T cell pool is subject to dynamic transcriptional changes in response to certain environmental cues, which in turn permutes the magnitude of the immune response.


Sujet(s)
Lymphocytes T CD4+ , Nippostrongylus , Animaux , Souris , Lymphocytes T CD4+/immunologie , Nippostrongylus/immunologie , Infections à Strongylida/immunologie , Infections à Strongylida/parasitologie , Organismes exempts d'organismes pathogènes spécifiques , Transcription génétique , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Helminthiase/immunologie , Interféron de type I/métabolisme , Interféron de type I/immunologie , Souris de lignée C57BL , Activation des lymphocytes/immunologie
4.
Semin Immunopathol ; 46(5): 12, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39150566

RÉSUMÉ

Biomedical research has witnessed significant strides in manufacturing chimeric antigen receptor T cell (CAR-T) therapies, marking a transformative era in cellular immunotherapy. Nevertheless, existing manufacturing methods for autologous cell therapies still pose several challenges related to cost, immune cell source, safety risks, and scalability. These challenges have motivated recent efforts to optimize process development and manufacturing for cell therapies using automated closed-system bioreactors and models created using artificial intelligence. Simultaneously, non-viral gene transfer methods like mRNA, CRISPR genome editing, and transposons are being applied to engineer T cells and other immune cells like macrophages and natural killer cells. Alternative sources of primary immune cells and stem cells are being developed to generate universal, allogeneic therapies, signaling a shift away from the current autologous paradigm. These multifaceted innovations in manufacturing underscore a collective effort to propel this therapeutic approach toward broader clinical adoption and improved patient outcomes in the evolving landscape of cancer treatment. Here, we review current CAR immune cell manufacturing strategies and highlight recent advancements in cell therapy scale-up, automation, process development, and engineering.


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Humains , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/immunologie , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Animaux , Tumeurs/thérapie , Tumeurs/immunologie , Thérapie cellulaire et tissulaire/méthodes , Édition de gène , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/métabolisme
5.
Sci Adv ; 10(33): eado4313, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39141734

RÉSUMÉ

αß T cell receptors (TCRs) principally recognize aberrant peptides bound to major histocompatibility complex molecules (pMHCs) on unhealthy cells, amplifying specificity and sensitivity through physical load placed on the TCR-pMHC bond during immunosurveillance. To understand this mechanobiology, TCRs stimulated by abundantly and sparsely arrayed epitopes (NP366-374/Db and PA224-233/Db, respectively) following in vivo influenza A virus infection were studied with optical tweezers. While certain NP repertoire CD8 T lymphocytes require many ligands for activation, others are digital, needing just few. Conversely, all PA TCRs perform digitally, exhibiting pronounced bond lifetime increases through sustained, energizing volleys of structural transitioning. Optimal digital performance is superior in vivo, correlating with ERK phosphorylation, CD3 loss, and activation marker up-regulation in vitro. Given neoantigen array paucity, digital TCRs are likely critical for immunotherapies.


Sujet(s)
Lymphocytes T CD8+ , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Souris , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Récepteur lymphocytaire T antigène, alpha-bêta/métabolisme , Récepteur lymphocytaire T antigène, alpha-bêta/composition chimique , Virus de la grippe A/immunologie , Humains , Activation des lymphocytes/immunologie , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Pinces optiques
6.
Oncotarget ; 15: 550-561, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39102218

RÉSUMÉ

Overexpression of the secretory protein renalase-1 negatively impacts the survival of melanoma and pancreatic cancer patients, while inhibition of renalase-1 signaling drives tumor rejection by promoting T-cell activation. Thus, we investigated the chemical complementarity between melanoma-resident, T-cell receptor (TCR) complementarity-determining region 3 (CDR3) amino acid sequences (AAs) and the renalase-1 protein. Increasing complementarity of TCR CDR3s to renalase-1 AAs, as assessed by a chemical complementarity scoring algorithm, was associated with improved overall survival (OS) in melanoma patients. The expression levels of several immune signature genes were significantly, positively correlated with increasing TCR CDR3-renalase-1 complementarity scores. Additionally, the survival association observed with high complementarity of TCR CDR3s to renalase-1 AAs was more robust in cases with low renalase-1 gene expression levels. Mapping of TCR CDR3-renalase-1 in silico interaction sites identified major epitope candidates including RP220, the signaling module of the renalase-1 protein, consistent with the fact that a monoclonal antibody to RP220 is a potent inhibitor of melanoma growth. These findings indicate that renalase-1 is a potential antigen for TCR recognition in melanoma and could be considered as a target for immunotherapy.


Sujet(s)
Régions déterminant la complémentarité , Mélanome , Récepteurs aux antigènes des cellules T , Humains , Mélanome/immunologie , Mélanome/génétique , Mélanome/mortalité , Mélanome/anatomopathologie , Mélanome/métabolisme , Régions déterminant la complémentarité/génétique , Régions déterminant la complémentarité/immunologie , Régions déterminant la complémentarité/composition chimique , Régions déterminant la complémentarité/métabolisme , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/génétique , Amidohydrolases/métabolisme , Amidohydrolases/génétique , Pronostic , Femelle , Monoamine oxidase
7.
Int J Biol Sci ; 20(10): 3942-3955, 2024.
Article de Anglais | MEDLINE | ID: mdl-39113701

RÉSUMÉ

T cells play important roles in antitumor immunity. However, given that the hepatocellular carcinoma (HCC) tumor microenvironment confers resistance to T cell-based immunotherapies, novel strategies to boost T cell-mediated antitumor efficacy are urgently needed for the treatment of HCC. Here, we show that high proprotein convertase subtilisin/kexin type9 (PCSK9) expression was negatively associated with HCC patient's overall survival and markers of CD8+ T cells. Pharmacological inhibition of PCSK9 enhanced tumor-specific killing and downregulated PD-1 expression of AFP-specific TCR-T. Inhibition of PCSK9 significantly enhances the anti-HCC efficacy of TCR-T cells and anti-PD-1 immunotherapy in vivo. Moreover, PCSK9 inhibitor suppressed HCC growth dependent on CD8+ T cells. Mechanically, pharmacological inhibition of PCSK9 promoted low-density lipoprotein receptor (LDLR)-mediated activation of mTORC1 signaling in CD8+ T cells. LDLR deficiency was shown to impair cellular mTORC1 signaling and the anti-HCC function of CD8 T cells. On the basis of our findings in this study, we propose a potential metabolic intervention strategy that could be used to enhance the antitumor effects of immunotherapy for HCC.


Sujet(s)
Lymphocytes T CD8+ , Carcinome hépatocellulaire , Immunothérapie , Tumeurs du foie , Proprotéine convertase 9 , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/immunologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/immunologie , Tumeurs du foie/métabolisme , Proprotéine convertase 9/métabolisme , Humains , Animaux , Immunothérapie/méthodes , Souris , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Microenvironnement tumoral , Inhibiteurs de PCSK9 , Souris de lignée C57BL , Récepteurs aux antigènes des cellules T/métabolisme , Mâle
8.
Front Immunol ; 15: 1411957, 2024.
Article de Anglais | MEDLINE | ID: mdl-39114656

RÉSUMÉ

Introduction: CD8+ cytotoxic T lymphocytes (CTLs) are highly effective in defending against viral infections and tumours. They are activated through the recognition of peptide-MHC-I complex by the T-cell receptor (TCR) and co-stimulation. This cognate interaction promotes the organisation of intimate cell-cell connections that involve cytoskeleton rearrangement to enable effector function and clearance of the target cell. This is key for the asymmetric transport and mobilisation of lytic granules to the cell-cell contact, promoting directed secretion of lytic mediators such as granzymes and perforin. Mitochondria play a role in regulating CTL function by controlling processes such as calcium flux, providing the necessary energy through oxidative phosphorylation, and its own protein translation on 70S ribosomes. However, the effect of acute inhibition of cytosolic translation in the rapid response after TCR has not been studied in mature CTLs. Methods: Here, we investigated the importance of cytosolic protein synthesis in human CTLs after early TCR activation and CD28 co-stimulation for the dynamic reorganisation of the cytoskeleton, mitochondria, and lytic granules through short-term chemical inhibition of 80S ribosomes by cycloheximide and 80S and 70S by puromycin. Results: We observed that eukaryotic ribosome function is required to allow proper asymmetric reorganisation of the tubulin cytoskeleton and mitochondria and mTOR pathway activation early upon TCR activation in human primary CTLs. Discussion: Cytosolic protein translation is required to increase glucose metabolism and degranulation capacity upon TCR activation and thus to regulate the full effector function of human CTLs.


Sujet(s)
Lymphocytes T CD8+ , Cytosol , Activation des lymphocytes , Mitochondries , Biosynthèse des protéines , Récepteurs aux antigènes des cellules T , Humains , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Activation des lymphocytes/immunologie , Cytosol/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Mitochondries/métabolisme , Mitochondries/immunologie , Cytosquelette/métabolisme , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/métabolisme , Ribosomes/métabolisme , Transduction du signal , Sérine-thréonine kinases TOR/métabolisme
9.
Sci Signal ; 17(846): eadp8569, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39042728

RÉSUMÉ

Chimeric antigen receptor (CAR) T cells have been used to successfully treat various blood cancers, but adverse effects have limited their potential. Here, we developed chimeric adaptor proteins (CAPs) and CAR tyrosine kinases (CAR-TKs) in which the intracellular ζ T cell receptor (TCRζ) chain was replaced with intracellular protein domains to stimulate signaling downstream of the TCRζ chain. CAPs contain adaptor domains and the kinase domain of ZAP70, whereas CAR-TKs contain only ZAP70 domains. We hypothesized that CAPs and CAR-TKs would be more potent than CARs because they would bypass both the steps that define the signaling threshold of TCRζ and the inhibitory regulation of upstream molecules. CAPs were too potent and exhibited high tonic signaling in vitro. In contrast, CAR-TKs exhibited high antitumor efficacy and significantly enhanced long-term tumor clearance in leukemia-bearing NSG mice as compared with the conventional CD19-28ζ-CAR-T cells. CAR-TKs were activated in a manner independent of the kinase Lck and displayed slower phosphorylation kinetics and prolonged signaling compared with the 28ζ-CAR. Lck inhibition attenuated CAR-TK cell exhaustion and improved long-term function. The distinct signaling properties of CAR-TKs may therefore be harnessed to improve the in vivo efficacy of T cells engineered to express an antitumor chimeric receptor.


Sujet(s)
Récepteurs aux antigènes des cellules T , Récepteurs chimériques pour l'antigène , Transduction du signal , Lymphocytes T , Animaux , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/génétique , Humains , Transduction du signal/immunologie , Souris , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/génétique , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , ZAP-70 Protein-tyrosine kinase/métabolisme , ZAP-70 Protein-tyrosine kinase/génétique , ZAP-70 Protein-tyrosine kinase/immunologie , Immunothérapie adoptive/méthodes , Souris de lignée NOD , Lignée cellulaire tumorale , Phosphorylation
10.
Sci Rep ; 14(1): 15053, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38956389

RÉSUMÉ

Targeting intracellular inhibiting proteins has been revealed to be a promising strategy to improve CD8+ T cell anti-tumor efficacy. Here, we are focusing on intracellular inhibiting proteins specific to TCR signaling: DOK1 and DOK2 expressed in T cells. We hypothesized that depletion of intracellular inhibition checkpoint DOK1 and DOK2 could improve CD8+ T-cell based cancer therapies. To evaluate the role of DOK1 and DOK2 depletion in physiology and effector function of CD8+ T lymphocytes and in cancer progression, we established a transgenic T cell receptor mouse model specific to melanoma antigen hgp100 (pmel-1 TCR Tg) in WT and Dok1/Dok2 DKO (double KO) mice. We showed that both DOK1 and DOK2 depletion in CD8+ T cells after an in vitro pre-stimulation induced a higher percentage of effector memory T cells as well as an up regulation of TCR signaling cascade- induced by CD3 mAbs, including the increased levels of pAKT and pERK, two major phosphoproteins involved in T cell functions. Interestingly, this improved TCR signaling was not observed in naïve CD8+ T cells. Despite this enhanced TCR signaling essentially shown upon stimulation via CD3 mAbs, pre-stimulated Dok1/Dok2 DKO CD8+ T cells did not show any increase in their activation or cytotoxic capacities against melanoma cell line expressing hgp100 in vitro. Altogether we demonstrate here a novel aspect of the negative regulation by DOK1 and DOK2 proteins in CD8+ T cells. Indeed, our results allow us to conclude that DOK1 and DOK2 have an inhibitory role following long term T cell stimulations.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Lymphocytes T CD8+ , Protéines de liaison à l'ADN , Mémoire immunologique , Souris knockout , Phosphoprotéines , Protéines de liaison à l'ARN , Récepteurs aux antigènes des cellules T , Transduction du signal , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Souris , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Phosphoprotéines/métabolisme , Phosphoprotéines/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Récepteurs aux antigènes des cellules T/métabolisme , Lignée cellulaire tumorale , Souris transgéniques
11.
Nat Commun ; 15(1): 6201, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39043656

RÉSUMÉ

CD4+ T cells recognising citrullinated self-epitopes presented by HLA-DRB1 bearing the shared susceptibility epitope (SE) are implicated in rheumatoid arthritis (RA). However, the underlying T cell receptor (TCR) determinants of epitope specificity towards distinct citrullinated peptide antigens, including vimentin-64cit59-71 and α-enolase-15cit10-22 remain unclear. Using HLA-DR4-tetramers, we examine the T cell repertoire in HLA-DR4 transgenic mice and observe biased TRAV6 TCR gene usage across these two citrullinated epitopes which matches with TCR bias previously observed towards the fibrinogen ß-74cit69-81 epitope. Moreover, shared TRAV26-1 gene usage is evident in four α-enolase-15cit10-22 reactive T cells in three human samples. Crystal structures of mouse TRAV6+ and human TRAV26-1+ TCR-HLA-DR4 complexes presenting vimentin-64cit59-71 and α-enolase-15cit10-22, respectively, show three-way interactions between the TCR, SE, citrulline, and the basis for the biased selection of TRAV genes. Position 2 of the citrullinated epitope is a key determinant underpinning TCR specificity. Accordingly, we provide a molecular basis of TCR specificity towards citrullinated epitopes.


Sujet(s)
Polyarthrite rhumatoïde , Lymphocytes T CD4+ , Antigène HLA-DR4 , Souris transgéniques , Vimentine , Humains , Antigène HLA-DR4/immunologie , Antigène HLA-DR4/génétique , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/génétique , Souris , Animaux , Vimentine/immunologie , Vimentine/métabolisme , Vimentine/génétique , Lymphocytes T CD4+/immunologie , Citrullination , Enolase/immunologie , Enolase/génétique , Enolase/métabolisme , Déterminants antigéniques des lymphocytes T/immunologie , Citrulline/métabolisme , Citrulline/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Épitopes/immunologie , Cristallographie aux rayons X , Récepteur lymphocytaire T antigène, alpha-bêta/génétique , Récepteur lymphocytaire T antigène, alpha-bêta/immunologie , Récepteur lymphocytaire T antigène, alpha-bêta/métabolisme
12.
Nat Commun ; 15(1): 5577, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38956082

RÉSUMÉ

Recent advances in single-cell immune profiling have enabled the simultaneous measurement of transcriptome and T cell receptor (TCR) sequences, offering great potential for studying immune responses at the cellular level. However, integrating these diverse modalities across datasets is challenging due to their unique data characteristics and technical variations. Here, to address this, we develop the multimodal generative model mvTCR to fuse modality-specific information across transcriptome and TCR into a shared representation. Our analysis demonstrates the added value of multimodal over unimodal approaches to capture antigen specificity. Notably, we use mvTCR to distinguish T cell subpopulations binding to SARS-CoV-2 antigens from bystander cells. Furthermore, when combined with reference mapping approaches, mvTCR can map newly generated datasets to extensive T cell references, facilitating knowledge transfer. In summary, we envision mvTCR to enable a scalable analysis of multimodal immune profiling data and advance our understanding of immune responses.


Sujet(s)
COVID-19 , Récepteurs aux antigènes des cellules T , SARS-CoV-2 , Analyse sur cellule unique , Transcriptome , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/immunologie , Analyse sur cellule unique/méthodes , Humains , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , COVID-19/immunologie , COVID-19/virologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Analyse de profil d'expression de gènes/méthodes , Antigènes viraux/immunologie , Antigènes viraux/génétique
13.
Oncoimmunology ; 13(1): 2371556, 2024.
Article de Anglais | MEDLINE | ID: mdl-38952674

RÉSUMÉ

Isolation of tumor-specific T cells and their antigen receptors (TCRs) from malignant pleural effusions (MPE) may facilitate the development of TCR-transduced adoptive cellular immunotherapy products for advanced lung cancer patients. However, the characteristics and markers of tumor-specific T-cells in MPE are largely undefined. To this end, to establish the phenotypes and antigen specificities of CD8+ T cells, we performed single-cell RNA and TCR sequencing of samples from three advanced lung cancer patients. Dimensionality reduction on a total of 4,983 CD8+ T cells revealed 10 clusters including naïve, memory, and exhausted phenotypes. We focused particularly on exhausted T cell clusters and tested their TCR reactivity against neoantigens predicted from autologous cancer cell lines. Four different TCRs specific for the same neoantigen and one orphan TCR specific for the autologous cell line were identified from one of the patients. Differential gene expression analysis in tumor-specific T cells relative to the other T cells identified CXCL13, as a candidate gene expressed by tumor-specific T cells. In addition to expressing CXCL13, tumor-specific T cells were present in a higher proportion of T cells co-expressing PDCD1(PD-1)/TNFRSF9(4-1BB). Furthermore, flow cytometric analyses in advanced lung cancer patients with MPE documented that those with high PD-1/4-1BB expression have a better prognosis in the subset of 57 adenocarcinoma patients (p = .039). These data suggest that PD-1/4-1BB co-expression might identify tumor-specific CD8+ T cells in MPE, which are associated with patients' prognosis. (233 words).


Sujet(s)
Lymphocytes T CD8+ , Tumeurs du poumon , Épanchement pleural malin , Récepteurs aux antigènes des cellules T , Analyse sur cellule unique , Humains , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Épanchement pleural malin/immunologie , Épanchement pleural malin/anatomopathologie , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/génétique , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Antigènes néoplasiques/immunologie
14.
Brief Bioinform ; 25(4)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-39007599

RÉSUMÉ

The interaction between T-cell receptors (TCRs) and peptides (epitopes) presented by major histocompatibility complex molecules (MHC) is fundamental to the immune response. Accurate prediction of TCR-epitope interactions is crucial for advancing the understanding of various diseases and their prevention and treatment. Existing methods primarily rely on sequence-based approaches, overlooking the inherent topology structure of TCR-epitope interaction networks. In this study, we present $GTE$, a novel heterogeneous Graph neural network model based on inductive learning to capture the topological structure between TCRs and Epitopes. Furthermore, we address the challenge of constructing negative samples within the graph by proposing a dynamic edge update strategy, enhancing model learning with the nonbinding TCR-epitope pairs. Additionally, to overcome data imbalance, we adapt the Deep AUC Maximization strategy to the graph domain. Extensive experiments are conducted on four public datasets to demonstrate the superiority of exploring underlying topological structures in predicting TCR-epitope interactions, illustrating the benefits of delving into complex molecular networks. The implementation code and data are available at https://github.com/uta-smile/GTE.


Sujet(s)
Récepteurs aux antigènes des cellules T , Récepteurs aux antigènes des cellules T/composition chimique , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Humains , Déterminants antigéniques des lymphocytes T/immunologie , Déterminants antigéniques des lymphocytes T/composition chimique , , Biologie informatique/méthodes , Liaison aux protéines , Épitopes/composition chimique , Épitopes/immunologie , Algorithmes , Logiciel
15.
Semin Immunopathol ; 46(3-4): 8, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39060547

RÉSUMÉ

Adoptive cell therapy (ACT) using Chimeric Antigen Receptor (CAR) and T Cell Receptor (TCR) engineered T cells represents an innovative therapeutic approach for the treatment of hematological malignancies, yet its application for solid tumors is still suboptimal. The tumor microenvironment (TME) places several challenges to overcome for a satisfactory therapeutic effect, such as physical barriers (fibrotic capsule and stroma), and inhibitory signals impeding T cell function. Some of these obstacles can be faced by combining ACT with other anti-tumor approaches, such as chemo/radiotherapy and checkpoint inhibitors. On the other hand, cutting edge technological tools offer the opportunity to overcome and, in some cases, take advantage of TME intrinsic characteristics to boost ACT efficacy. These include: the exploitation of chemokine gradients and integrin expression for preferential T-cell homing and extravasation; metabolic changes that have direct or indirect effects on TCR-T and CAR-T cells by increasing antigen presentation and reshaping T cell phenotype; introduction of additional synthetic receptors on TCR-T and CAR-T cells with the aim of increasing T cells survival and fitness.


Sujet(s)
Immunothérapie adoptive , Tumeurs , Récepteurs chimériques pour l'antigène , Lymphocytes T , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Immunothérapie adoptive/méthodes , Tumeurs/thérapie , Tumeurs/immunologie , Animaux , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/génétique
16.
Zhonghua Bing Li Xue Za Zhi ; 53(7): 667-671, 2024 Jul 08.
Article de Chinois | MEDLINE | ID: mdl-38955696

RÉSUMÉ

Objective: To investigate the clinicopathological features and differential diagnosis of primary mucosal CD30-positive T-cell lymphoproliferative disorders (pmCD30+TLPD). Methods: Eight cases of pmCD30+TLPD diagnosed from 2013 to 2023 at the Department of Pathology, Beijing Friendship Hospital Affiliated to Capital Medical University and Beijing Ludaopei Hospital were retrospectively collected. The immunophenotype, EBV infection status and T-cell receptor (TCR) clonability of tumor cells were examined. The clinicopathological features were analyzed and related literatures were reviewed. Results: There were 5 females and 3 males, aged 28 to 73 years, without B symptoms, lack of trauma and autoimmune diseases. Seven cases occurred in oral mucosa and one in anal canal mucosa. Submucosal nodules with ulcerations were presented in all cases except one, which only submucosal nodule. Morphologically, there was different distribution of allotypic lymphocytes in inflammatory background. Four cases showed "kidney-shaped", "embryonic" and "horseshoe-shaped" cells, and one case resembled Hodgkin and Reed/Sternberg (HRS) cells. Allotypic lymphocytes expressed CD3 (7/8), CD4+/CD8-(7/8) and CD4-/CD8-(1/8). CD30 was uniformly strongly positive while ALK and CD56 were negative. In situ hybridization of EBER was negative in five cases (5/5). Clonal TCR gene rearrangement was positive in two cases. Four patients did not receive radiotherapy or chemotherapy. All the seven patients survived without disease except one died due to concurrent leukopenia. Conclusions: pmCD30+TLPD had a broad morphological spectrum and could be easily confused with primary cutaneous CD30+TLPD and systemic ALK-negative anaplastic large cell lymphoma involving mucosa, which may lead to misdiagnosis. Although the majority of the cases had a favorable prognosis, a few cases relapsed or progressed to lymphoma.


Sujet(s)
Antigènes CD30 , Syndromes lymphoprolifératifs , Humains , Mâle , Femelle , Sujet âgé , Adulte , Syndromes lymphoprolifératifs/anatomopathologie , Syndromes lymphoprolifératifs/métabolisme , Antigènes CD30/métabolisme , Adulte d'âge moyen , Études rétrospectives , Diagnostic différentiel , Lymphocytes T/anatomopathologie , Lymphocytes T/immunologie , Muqueuse de la bouche/anatomopathologie , Cellules de Reed-Sternberg/anatomopathologie , Cellules de Reed-Sternberg/métabolisme , Infections à virus Epstein-Barr , Immunophénotypage , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/génétique
17.
Sci Transl Med ; 16(755): eadg7123, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38985855

RÉSUMÉ

Two types of engineered T cells have been successfully used to treat patients with cancer, one with an antigen recognition domain derived from antibodies [chimeric antigen receptors (CARs)] and the other derived from T cell receptors (TCRs). CARs use high-affinity antigen-binding domains and costimulatory domains to induce T cell activation but can only react against target cells with relatively high amounts of antigen. TCRs have a much lower affinity for their antigens but can react against target cells displaying only a few antigen molecules. Here, we describe a new type of receptor, called a Co-STAR (for costimulatory synthetic TCR and antigen receptor), that combines aspects of both CARs and TCRs. In Co-STARs, the antigen-recognizing components of TCRs are replaced by high-affinity antibody fragments, and costimulation is provided by two modules that drive NF-κB signaling (MyD88 and CD40). Using a TCR-mimic antibody fragment that targets a recurrent p53 neoantigen presented in a common human leukocyte antigen (HLA) allele, we demonstrate that T cells equipped with Co-STARs can kill cancer cells bearing low densities of antigen better than T cells engineered with conventional CARs and patient-derived TCRs in vitro. In mouse models, we show that Co-STARs mediate more robust T cell expansion and more durable tumor regressions than TCRs similarly modified with MyD88 and CD40 costimulation. Our data suggest that Co-STARs may have utility for other peptide-HLA antigens in cancer and other targets where antigen density may limit the efficacy of engineered T cells.


Sujet(s)
Tumeurs , Récepteurs aux antigènes des cellules T , Récepteurs chimériques pour l'antigène , Humains , Animaux , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Tumeurs/immunologie , Tumeurs/thérapie , Souris , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lignée cellulaire tumorale , Antigènes néoplasiques/immunologie , Antigènes néoplasiques/métabolisme , Transduction du signal
18.
Front Immunol ; 15: 1424987, 2024.
Article de Anglais | MEDLINE | ID: mdl-38979423

RÉSUMÉ

Mucosal-associated invariant T (MAIT) cells are a major subset of innate-like T cells that function at the interface between innate and acquired immunity. MAIT cells recognize vitamin B2-related metabolites produced by microbes, through semi-invariant T cell receptor (TCR) and contribute to protective immunity. These foreign-derived antigens are presented by a monomorphic antigen presenting molecule, MHC class I-related molecule 1 (MR1). MR1 contains a malleable ligand-binding pocket, allowing for the recognition of compounds with various structures. However, interactions between MR1 and self-derived antigens are not fully understood. Recently, bile acid metabolites were identified as host-derived ligands for MAIT cells. In this review, we will highlight recent findings regarding the recognition of self-antigens by MAIT cells.


Sujet(s)
Antigènes d'histocompatibilité de classe I , Cellules T invariantes associées aux muqueuses , Cellules T invariantes associées aux muqueuses/immunologie , Cellules T invariantes associées aux muqueuses/métabolisme , Humains , Animaux , Antigènes d'histocompatibilité de classe I/immunologie , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes mineurs d'histocompatibilité/immunologie , Antigènes mineurs d'histocompatibilité/métabolisme , Autoantigènes/immunologie , Présentation d'antigène/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Récepteurs aux antigènes des cellules T/métabolisme
19.
Oncoimmunology ; 13(1): 2373530, 2024.
Article de Anglais | MEDLINE | ID: mdl-38979545

RÉSUMÉ

TCRαß+ CD4- CD8- double-negative T (DNT) cells are minor populations in peripheral blood, and their roles have mostly been discussed in inflammation and autoimmunity. However, the functions of DNT cells in tumor microenvironment remain to be elucidated. We investigated their characteristics, possible origins and functions in colorectal cancer tissues as well as their corresponding tumor-draining lymph nodes. We found a significant enrichment of DNT cells in tumor tissues compared with their corresponding lymph nodes, especially in tumors with lower T cell infiltration. T cell receptor (TCR) sequence analysis of CD4+ T, CD8+ T and DNT cells indicated that TCR sequences detected in DNT cells were found in CD8+ T cells, but rarely in CD4+ T cells, suggesting that a part of DNT cells was likely to be originated from CD8+ T cells. Through a single-cell transcriptomic analysis of DNT cells, we found that a DNT cell cluster, which showed similar phenotypes to central memory CD8+ T cells with low expression of effector and exhaustion markers, revealed some specific gene expression patterns, including higher GZMK expression. Moreover, in flow cytometry analysis, we found that DNT cells lost production of cytotoxic mediators. These findings imply that DNT cells might function as negative regulators of anti-tumor immune responses in tumor microenvironment.


Sujet(s)
Tumeurs colorectales , Noeuds lymphatiques , Microenvironnement tumoral , Humains , Tumeurs colorectales/immunologie , Tumeurs colorectales/anatomopathologie , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/anatomopathologie , Microenvironnement tumoral/immunologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Mâle , Femelle , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Sujet âgé , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/génétique , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Adulte d'âge moyen
20.
Commun Biol ; 7(1): 918, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39080357

RÉSUMÉ

Actin dynamics control early T-cell receptor (TCR) signalling during T-cell activation. However, the precise regulation of initial actin rearrangements is not completely understood. Here, we have investigated the regulatory role of the phosphatase Slingshot-1 (SSH1) in this process. Our data show that SSH1 rapidly polarises to nascent cognate synaptic contacts and later relocalises to peripheral F-actin networks organised at the mature immunological synapse. Knockdown of SSH1 expression by CRISPR/Cas9-mediated genome editing or small interfering RNA reveal a regulatory role for SSH1 in CD3ε conformational change, allowing Nck binding and proper downstream signalling and immunological synapse organisation. TCR triggering induces SSH1-mediated activation of actin dynamics through a mechanism mediated by Limk-1 inactivation. These data suggest that during early TCR activation, SSH1 is required for rapid F-actin rearrangements that mediate initial conformational changes of the TCR, integrin organisation and proximal signalling events for proper synapse organisation. Therefore, the SSH1 and Limk-1 axis is a key regulatory element for full T cell activation.


Sujet(s)
Lim Kinases , Phosphoprotein Phosphatases , Récepteurs aux antigènes des cellules T , Humains , Lim Kinases/métabolisme , Lim Kinases/génétique , Récepteurs aux antigènes des cellules T/métabolisme , Phosphoprotein Phosphatases/métabolisme , Phosphoprotein Phosphatases/génétique , Actines/métabolisme , Actines/génétique , Activation des lymphocytes , Cellules Jurkat , Lymphocytes T/métabolisme , Lymphocytes T/immunologie , Transduction du signal , Synapses immunologiques/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE