Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.087
Filter
1.
Rinsho Ketsueki ; 65(7): 644-651, 2024.
Article in Japanese | MEDLINE | ID: mdl-39098015

ABSTRACT

T cell malignancies pose several unique issues for CAR-T cell therapy that were not significant concerns with CAR-T cells for B-cell malignancies. A general problem to consider in the production of CAR-T cells is "on target-off tumor toxicity." This occurs when the antigen targeted by the CAR-T cells is also expressed on normal cells, not just tumor cells, which causes CAR-T cells to damage these normal cells. In CAR-T cell therapy for T cell tumors, antigens expressed on T cells (such as CD5, CD7, etc.) are the targets, which leads to a problem known as "fratricide," where CAR-T cells kill each other. Other issues include T cell aplasia and contamination of CAR-T cell products with tumor cells. However, several recent clinical trials have shown excellent outcomes for CAR-T cell therapy when genome editing technology is used to overcome these issues by knocking out target antigens or T cell receptors. This review article outlines these challenges and their solutions and discusses the results of recent clinical trials.


Subject(s)
T-Lymphocytes , Humans , T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Neoplasms/therapy , Neoplasms/immunology
2.
Rinsho Ketsueki ; 65(7): 662-667, 2024.
Article in Japanese | MEDLINE | ID: mdl-39098017

ABSTRACT

Chimeric antigen receptor T-cell therapy (CAR-T-cell therapy) has revolutionized the treatment of relapsed and refractory hematological malignancies. Targeting of the CD19 antigen on B cells has yielded high rates of remission induction and sustained remission in patients with acute lymphoblastic leukemia and B-cell lymphomas. Despite these remarkable responses, many escape mechanisms from CAR-T cell therapy have been identified, with the most common being target antigen deficiency. This paper focuses on CD19 CAR-T cell therapies, which are currently the most clinically used, and describes new strategies to overcome resistance using multi-targeted CAR-T cells, such as CD19-CD20 CAR-T cells and CD19-CD22 CAR-T cells, which are being developed in preclinical and clinical trials.


Subject(s)
Antigens, CD19 , Humans , Antigens, CD19/immunology , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/immunology
3.
Best Pract Res Clin Haematol ; 37(2): 101558, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39098804

ABSTRACT

The human adaptive immune repertoire is characterized by specificity and diversity to provide immunity against past and future tasks. Such tasks are mainly infections but also malignant transformations of cells. With its multiple lines of defense, the human immune system contains both, rapid reaction forces and the potential to capture, disassemble and analyze strange structures in order to teach the adaptive immune system and mount a specific immune response. Prevention and mitigation of autoimmunity is of equal importance. In the context of allogeneic hematopoietic cell transplantation (HCT) specific challenges exist with the transfer of cells from the adapted donor immune system to the immunosuppressed recipient. Those challenges are immunogenetic disparity between donor and host, reconstitution of immunity early after HCT by expansion of mature immune effector cells, and impaired thymic function, if the recipient is an adult (as it is the case in most HCTs). The possibility to characterize the adaptive immune repertoire by massively parallel sequencing of T-cell receptor gene rearrangements allows for a much more detailed characterization of the T-cell repertoire. In addition, high-dimensional characterization of immune effector cells based on their immunophenotype and single cell RNA sequencing allow for much deeper insights in adaptive immune responses. We here review, existing - still incomplete - information on immune reconstitution after allogeneic HCT. Building on the technological advances much deeper insights into immune recovery after HCT and adaptive immune responses and can be expected in the coming years.


Subject(s)
Hematopoietic Stem Cell Transplantation , Receptors, Antigen, T-Cell , Humans , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Transplantation, Homologous , Adaptive Immunity , Allografts , T-Lymphocytes/immunology
4.
Oncotarget ; 15: 550-561, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39102218

ABSTRACT

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.


Subject(s)
Complementarity Determining Regions , Melanoma , Receptors, Antigen, T-Cell , Humans , Melanoma/immunology , Melanoma/genetics , Melanoma/mortality , Melanoma/pathology , Melanoma/metabolism , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Amidohydrolases/metabolism , Amidohydrolases/genetics , Prognosis , Female , Monoamine Oxidase
5.
Front Immunol ; 15: 1333150, 2024.
Article in English | MEDLINE | ID: mdl-39091493

ABSTRACT

Immunotherapy has made significant strides in cancer treatment with strategies like checkpoint blockade antibodies and adoptive T cell transfer. Chimeric antigen receptor T cells (CAR-T) have emerged as a promising approach to combine these strategies and overcome their limitations. This review explores CAR-T cells as a living drug for cancer treatment. CAR-T cells are genetically engineered immune cells designed to target and eliminate tumor cells by recognizing specific antigens. The study involves a comprehensive literature review on CAR-T cell technology, covering structure optimization, generations, manufacturing processes, and gene therapy strategies. It examines CAR-T therapy in haematologic cancers and solid tumors, highlighting challenges and proposing a suicide gene-based mechanism to enhance safety. The results show significant advancements in CAR-T technology, particularly in structure optimization and generation. The manufacturing process has improved for broader clinical application. However, a series of inherent challenges and side effects still need to be addressed. In conclusion, CAR-T cells hold great promise for cancer treatment, but ongoing research is crucial to improve efficacy and safety for oncology patients. The proposed suicide gene-based mechanism offers a potential solution to mitigate side effects including cytokine release syndrome (the most common toxic side effect of CAR-T therapy) and the associated neurotoxicity.


Subject(s)
Genes, Transgenic, Suicide , Immunotherapy, Adoptive , Neoplasms , Receptors, Chimeric Antigen , T-Lymphocytes , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/genetics , T-Lymphocytes/immunology , Animals , Genetic Therapy/adverse effects , Genetic Therapy/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
6.
Front Immunol ; 15: 1411614, 2024.
Article in English | MEDLINE | ID: mdl-39091495

ABSTRACT

T cell receptors (TCRs) play crucial roles in regulating T cell response by rapidly and accurately recognizing foreign and non-self antigens. The process involves multiple molecules and regulatory mechanisms, forming a complex network to achieve effective antigen recognition. Mathematical modeling techniques can help unravel the intricate network of TCR signaling and identify key regulators that govern it. In this review, we introduce and briefly discuss relevant mathematical models of TCR initial triggering, with a focus on kinetic proofreading (KPR) models with different modified structures. We compare the topology structures, biological hypotheses, parameter choices, and simulation performance of each model, and summarize the advantages and limitations of them. Further studies on TCR modeling design, aiming for an optimized balance of specificity and sensitivity, are expected to contribute to the development of new therapeutic strategies.


Subject(s)
Receptors, Antigen, T-Cell , Signal Transduction , Receptors, Antigen, T-Cell/immunology , Humans , Animals , Signal Transduction/immunology , T-Lymphocytes/immunology , Models, Immunological , Lymphocyte Activation/immunology , Models, Theoretical
7.
Emerg Microbes Infect ; 13(1): 2387448, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39109538

ABSTRACT

Therapeutics for eradicating hepatitis B virus (HBV) infection are still limited and current nucleos(t)ide analogs (NAs) and interferon are effective in controlling viral replication and improving liver health, but they cannot completely eradicate the hepatitis B virus and only a very small number of patients are cured of it. The TCR-like antibodies recognizing viral peptides presented on human leukocyte antigens (HLA) provide possible tools for targeting and eliminating HBV-infected hepatocytes. Here, we generated three TCR-like antibodies targeting three different HLA-A2.1-presented peptides derived from HBV core and surface proteins. Bispecific antibodies (BsAbs) were developed by fuzing variable fragments of these TCR-like mAbs with an anti-CD3ϵ antibody. Our data demonstrate that the BsAbs could act as T cell engagers, effectively redirecting and activating T cells to target HBV-infected hepatocytes in vitro and in vivo. In HBV-persistent mice expressing human HLA-A2.1, two infusions of BsAbs induced marked and sustained suppression in serum HBsAg levels and also reduced the numbers of HBV-positive hepatocytes. These findings highlighted the therapeutic potential of TCR-like BsAbs as a new strategy to cure hepatitis B.


Subject(s)
Antibodies, Bispecific , Disease Models, Animal , Hepatitis B virus , Hepatitis B , Hepatocytes , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Hepatocytes/virology , Hepatocytes/immunology , Mice , Humans , Hepatitis B virus/immunology , Hepatitis B virus/genetics , Hepatitis B/immunology , Hepatitis B/virology , HLA-A2 Antigen/immunology , Hepatitis B Surface Antigens/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology
8.
Front Immunol ; 15: 1411957, 2024.
Article in English | MEDLINE | ID: mdl-39114656

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes , Cytosol , Lymphocyte Activation , Mitochondria , Protein Biosynthesis , Receptors, Antigen, T-Cell , Humans , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Lymphocyte Activation/immunology , Cytosol/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mitochondria/metabolism , Mitochondria/immunology , Cytoskeleton/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Ribosomes/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
9.
Brief Bioinform ; 25(5)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39129361

ABSTRACT

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.


Subject(s)
Receptors, Antigen, T-Cell , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Cluster Analysis , Humans , Antigens/immunology , Computational Biology/methods , Algorithms , Machine Learning
10.
Immunity ; 57(8): 1893-1907.e6, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39096910

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes , Nippostrongylus , Animals , Mice , CD4-Positive T-Lymphocytes/immunology , Nippostrongylus/immunology , Strongylida Infections/immunology , Strongylida Infections/parasitology , Specific Pathogen-Free Organisms , Transcription, Genetic , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Helminthiasis/immunology , Interferon Type I/metabolism , Interferon Type I/immunology , Mice, Inbred C57BL , Lymphocyte Activation/immunology
11.
BMC Cancer ; 24(1): 984, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39123197

ABSTRACT

PURPOSE: Chimeric antigen receptor T-cell (CART) therapy has shown clinical efficacy in refractory and relapsed large B-cell lymphomas, but is associated with serious acute and long-term toxicities. To understand the patient perspective, we measured a patient-reported outcome (PRO), specifically, health-related quality of life (HRQoL), at multiple time points over one year. METHODS: This was a prospective feasibility study of a cohort of patients who were eligible for standard of care CART therapy, tisagenlecleucel. Demographic data and disease characteristics were collected. HRQoL was measured using FACT-Lym at baseline, and months 1, 3, 6 and 12. FACT-Lym includes FACT-G (physical, social, emotional and functional well-being domains), plus a lymphoma subscale. RESULTS: Thirty-four of 35 patients approached, consented to participate. Two of them did not receive their infusion due to progressive disease. 50% were female and median age was 62 (23-77). Twenty-nine patients (91%) completed baseline FACT-Lym and 20 of 21 (95%) eligible patients completed 12-month FACT-Lym. 52% completed all 4 post-baseline FACT-Lym measures. Exploratory analyses for changes in FACT-Lym scores are reported. CONCLUSION: It is feasible to measure longitudinal PROs in patients who receive CART therapy. This study will inform future studies in evaluating the patient perspective on CART therapy.


Subject(s)
Feasibility Studies , Immunotherapy, Adoptive , Lymphoma, Large B-Cell, Diffuse , Patient Reported Outcome Measures , Quality of Life , Humans , Female , Male , Middle Aged , Aged , Adult , Lymphoma, Large B-Cell, Diffuse/therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Prospective Studies , Young Adult , Receptors, Chimeric Antigen/therapeutic use , Receptors, Chimeric Antigen/immunology , Receptors, Antigen, T-Cell/therapeutic use , Receptors, Antigen, T-Cell/immunology , Longitudinal Studies , Neoplasm Recurrence, Local/immunology , Treatment Outcome
12.
Sci Adv ; 10(33): eado4313, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39141734

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Influenza A virus/immunology , Humans , Lymphocyte Activation/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Optical Tweezers
13.
Proc Natl Acad Sci U S A ; 121(34): e2321686121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39141352

ABSTRACT

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.


Subject(s)
Antigens, CD1d , Lipidomics , Natural Killer T-Cells , Receptors, Antigen, T-Cell , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Animals , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Mice , Lipidomics/methods , Humans , Autoantigens/immunology , Autoantigens/metabolism , Ceramides/metabolism , Ceramides/immunology , Lipids/chemistry , Lipids/immunology , Endoplasmic Reticulum Stress/immunology
14.
Sci Transl Med ; 16(755): eadg7123, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38985855

ABSTRACT

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.


Subject(s)
Neoplasms , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen , Humans , Animals , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Neoplasms/immunology , Neoplasms/therapy , Mice , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line, Tumor , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Signal Transduction
15.
Front Immunol ; 15: 1424987, 2024.
Article in English | MEDLINE | ID: mdl-38979423

ABSTRACT

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.


Subject(s)
Histocompatibility Antigens Class I , Mucosal-Associated Invariant T Cells , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/metabolism , Humans , Animals , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Minor Histocompatibility Antigens/immunology , Minor Histocompatibility Antigens/metabolism , Autoantigens/immunology , Antigen Presentation/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism
16.
Sci Immunol ; 9(97): eado5295, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38996008

ABSTRACT

αß T cell receptor (TCR) V(D)J genes code for billions of TCR combinations. However, only some appear on peripheral T cells in any individual because, to mature, thymocytes must react with low affinity but not high affinity with thymus expressed major histocompatibility (MHC)/peptides. MHC proteins are very polymorphic. Different alleles bind different peptides. Therefore, any individual might express many different MHC alleles to ensure that some peptides from an invader are bound to MHC and activate T cells. However, most individuals express limited numbers of MHC alleles. To explore this, we compared the TCR repertoires of naïve CD4 T cells in mice expressing one or two MHC alleles. Unexpectedly, the TCRs in heterozygotes were less diverse that those in the sum of their MHC homozygous relatives. Our results suggest that thymus negative selection cancels out the advantages of increased thymic positive selection in the MHC heterozygotes.


Subject(s)
CD4-Positive T-Lymphocytes , Heterozygote , Animals , Mice , CD4-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Major Histocompatibility Complex/immunology , Major Histocompatibility Complex/genetics , Mice, Inbred C57BL , Thymus Gland/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Mice, Transgenic
17.
Nat Commun ; 15(1): 5577, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38956082

ABSTRACT

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.


Subject(s)
COVID-19 , Receptors, Antigen, T-Cell , SARS-CoV-2 , Single-Cell Analysis , Transcriptome , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Single-Cell Analysis/methods , Humans , SARS-CoV-2/immunology , SARS-CoV-2/genetics , COVID-19/immunology , COVID-19/virology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Gene Expression Profiling/methods , Antigens, Viral/immunology , Antigens, Viral/genetics
18.
Sci Signal ; 17(846): eadp8569, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39042728

ABSTRACT

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.


Subject(s)
Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen , Signal Transduction , T-Lymphocytes , Animals , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/genetics , Humans , Signal Transduction/immunology , Mice , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/immunology , Immunotherapy, Adoptive/methods , Mice, Inbred NOD , Cell Line, Tumor , Phosphorylation
19.
Front Immunol ; 15: 1440499, 2024.
Article in English | MEDLINE | ID: mdl-39021567

ABSTRACT

The tyrosine kinase Lck is mandatory for initiating signaling responses downstream the antigenic T cell receptor (TCR). Numerous studies have shown that a prerequisite for efficient and well-balanced Lck regulation and function is its finely orchestrated spatial distribution pattern, especially at the plane of the plasma membrane. There is a wealth of knowledge on Lck localization sites, preference for specialized lipid microenvironments and colocalization partners. However, several questions concerning the spatial organization of its differentially phosphorylated conformers and the dynamics of their juxtaposition in relation to ligated and non-ligated TCRs remain elusive. In this brief report we introduce a non-invasive nanobody-based approach for mapping Lck subcellular allocation with high precision. Our initial data using this methodology, provide insight into the topology of Lck in resting T cells and its confined localization in a strictly delimited environment within the plane of the plasma membrane.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , Single-Domain Antibodies , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Humans , Single-Domain Antibodies/immunology , Cell Membrane/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Jurkat Cells , Phosphorylation , Signal Transduction
20.
Sci Adv ; 10(30): eadl0796, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39047099

ABSTRACT

Naive T cells recirculate between the spleen and lymph nodes where they mount immune responses when meeting dendritic cells presenting foreign antigen. As this may happen anywhere, naive T cells ought to visit all lymph nodes. Here, deep sequencing almost-complete TCR repertoires led to a comparison of different lymph nodes within and between individual mice. We find strong evidence for a deterministic CD4/CD8 lineage choice and a consistent spatial structure. Specifically, some T cells show a preference for one or multiple lymph nodes, suggesting that their TCR interacts with locally presented (self-)peptides. These findings are mirrored in TCR-transgenic mice showing localized CD69 expression, retention, and cell division. Thus, naive T cells intermittently sense antigenically dissimilar niches, which is expected to affect their homeostatic competition.


Subject(s)
Lymph Nodes , Mice, Transgenic , Receptors, Antigen, T-Cell , Animals , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Antigens, CD/metabolism , Antigens, CD/genetics , Lectins, C-Type/metabolism , Lectins, C-Type/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Differentiation, T-Lymphocyte/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL