Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.024
Filter
1.
Methods Mol Biol ; 2857: 1-8, 2025.
Article in English | MEDLINE | ID: mdl-39348050

ABSTRACT

Aging is a natural process that compromises the immune system's functionality increasing the risk of infectious, tumors, and autoimmune diseases. The thymus involution is an age-dependent process characterized by decreased cellularity, peripheral lymphocyte infiltration into the perivascular space, and expansion of adipose tissue. All those modifications hamper the functionality of the organ and lead to a decline of naïve T-cell production with a shrinking of the T-cell repertoire. Thymus atrophy is described in several disorders including autoimmune diseases. The quantification of T-cell receptor excision circles (TRECs) in recent thymus emigrants is a standard procedure to investigate the thymic function. In this chapter, we discuss the methodology used to quantify this molecule in peripheral blood mononuclear cells and isolated CD4+ and CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Receptors, Antigen, T-Cell , Thymus Gland , Humans , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Thymus Gland/immunology , Thymus Gland/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology
2.
Methods Mol Biol ; 2857: 127-135, 2025.
Article in English | MEDLINE | ID: mdl-39348061

ABSTRACT

The T-cell receptor (TCR) is the key molecule involved in the adaptive immune response. It is generated by the V(D)J recombination, responsible of the enormous diversity of the TCR repertoire, a crucial feature determining the individual capability to response to antigens and to build immunological memory. A pivotal role in the recognition of antigen is played by the hypervariable complementarity-determining region 3 (CDR3) of the V-beta chain of TCR. Investigating the CDR3 supports the understanding of the adaptive immune system dynamics in physiological processes, such as immune aging, and in disease, especially autoimmune disorders in which T cells are main actors. High-throughput sequencing (HTS) paved the way for a great progress in the investigation of TCR repertoire, enhancing the read depth in the process of library generation of sequencing and the number of samples that can be analyzed simultaneously. Therefore, the leverage of big datasets stressed the need to develop computational approach, by bioinformatics, to unravel the characteristics of the TCR repertoire.


Subject(s)
Complementarity Determining Regions , Computational Biology , High-Throughput Nucleotide Sequencing , Receptors, Antigen, T-Cell , T-Lymphocytes , Workflow , Computational Biology/methods , Humans , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , High-Throughput Nucleotide Sequencing/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Complementarity Determining Regions/genetics , Cell Separation/methods , V(D)J Recombination
3.
Nat Commun ; 15(1): 8722, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379371

ABSTRACT

Stevens-Johnson syndrome and toxic epidermal necrolysis (SJS/TEN) is a rare but life-threatening cutaneous drug reaction mediated by human leukocyte antigen (HLA) class I-restricted CD8+ T cells. For unbiased assessment of cellular immunopathogenesis, here we perform single-cell (sc) transcriptome, surface proteome, and T cell receptor (TCR) sequencing on unaffected skin, affected skin, and blister fluid from 15 SJS/TEN patients. From 109,888 cells, we identify 15 scRNA-defined subsets. Keratinocytes express markers indicating HLA class I-restricted antigen presentation and appear to trigger the proliferation of and killing by cytotoxic CD8+ tissue-resident T cells that express granulysin, granzyme B, perforin, LAG3, CD27, and LINC01871, and signal through the PKM, MIF, TGFß, and JAK-STAT pathways. In affected tissue, cytotoxic CD8+ T cells express private expanded and unexpanded TCRαß that are absent or unexpanded in unaffected skin, and mixed populations of macrophages and fibroblasts express pro-inflammatory markers or those favoring repair. This data identifies putative cytotoxic TCRs and therapeutic targets.


Subject(s)
CD8-Positive T-Lymphocytes , Keratinocytes , Receptors, Antigen, T-Cell , Single-Cell Analysis , Stevens-Johnson Syndrome , Humans , Stevens-Johnson Syndrome/immunology , Stevens-Johnson Syndrome/genetics , Single-Cell Analysis/methods , Keratinocytes/immunology , Keratinocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Skin/immunology , Skin/pathology , T-Lymphocytes, Cytotoxic/immunology , Granzymes/metabolism , Granzymes/genetics , Transcriptome , Male , Perforin/metabolism , Perforin/genetics , Female , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , Macrophages/immunology , Macrophages/metabolism
4.
Proc Natl Acad Sci U S A ; 121(42): e2408696121, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39374400

ABSTRACT

A key challenge in molecular biology is to decipher the mapping of protein sequence to function. To perform this mapping requires the identification of sequence features most informative about function. Here, we quantify the amount of information (in bits) that T cell receptor (TCR) sequence features provide about antigen specificity. We identify informative features by their degree of conservation among antigen-specific receptors relative to null expectations. We find that TCR specificity synergistically depends on the hypervariable regions of both receptor chains, with a degree of synergy that strongly depends on the ligand. Using a coincidence-based approach to measuring information enables us to directly bound the accuracy with which TCR specificity can be predicted from partial matches to reference sequences. We anticipate that our statistical framework will be of use for developing machine learning models for TCR specificity prediction and for optimizing TCRs for cell therapies. The proposed coincidence-based information measures might find further applications in bounding the performance of pairwise classifiers in other fields.


Subject(s)
Receptors, Antigen, T-Cell , T-Lymphocytes , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Humans , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , T-Cell Antigen Receptor Specificity , Amino Acid Sequence
5.
J Transl Med ; 22(1): 897, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39367419

ABSTRACT

TCR-T cell therapy represents a promising advancement in adoptive immunotherapy for cancer treatment. Despite its potential, the development and preclinical testing of TCR-T cells face significant challenges. This review provides a structured overview of the key stages in preclinical testing, including in silico, in vitro, and in vivo methods, within the context of the sequential development of novel therapies. This review aimed to systematically outline the processes for evaluating TCR-T cells at each stage: from in silico approaches used to predict target antigens, assess cross-reactivity, and minimize off-target effects, to in vitro assays designed to measure cell functionality, cytotoxicity, and activation. Additionally, the review discusses the limitations of in vivo testing in animal models, particularly in accurately reflecting the human tumor microenvironment and immune responses. Performed analysis emphasizes the importance of these preclinical stages in the safe and effective development of TCR-T cell therapies. While current models provide valuable insights, we identify critical gaps, particularly in in vivo biodistribution and toxicity assessments, and propose the need for enhanced standardization and the development of more representative models. This structured approach aims to improve the predictability and safety of TCR-T cell therapy as it advances towards clinical application.


Subject(s)
T-Lymphocytes , Humans , Animals , T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Cell- and Tissue-Based Therapy , Immunotherapy, Adoptive/methods , Social Control, Formal , Neoplasms/therapy , Neoplasms/immunology
6.
Sci Adv ; 10(41): eado2134, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39383231

ABSTRACT

The molecular mechanisms responsible for the heightened reactivity of quiescent T cells in human early life remain largely elusive. Our previous research identified that quiescent adult naïve CD4+ T cells express LINE1 (long interspersed nuclear elements 1) spliced in previously unknown isoforms, and their down-regulation marks the transition to activation. Here, we unveil that neonatal naïve T cell quiescence is characterized by enhanced energy production and protein synthesis. This phenotype is associated with the absence of LINE1 expression attributed to tonic T cell receptor/mTOR complex 1 (mTORC1) signaling and (polypyrimidine tract-binding protein 1 (PTBP1)-mediated LINE1 splicing suppression. The absence of LINE1 expression primes these cells for rapid execution of the activation program by directly regulating protein synthesis. LINE1 expression progressively increases in childhood and adults, peaking in elderly individuals, and, by decreasing protein synthesis, contributes to immune senescence in aging. Our study proposes LINE1 as a critical player of human T cell function across the human life span.


Subject(s)
Long Interspersed Nucleotide Elements , Mechanistic Target of Rapamycin Complex 1 , Polypyrimidine Tract-Binding Protein , Protein Biosynthesis , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Signal Transduction , Adult , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/metabolism , Lymphocyte Activation , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , Aging/metabolism , Gene Expression Regulation , Child
7.
Front Immunol ; 15: 1443910, 2024.
Article in English | MEDLINE | ID: mdl-39257583

ABSTRACT

The thymus plays a pivotal role in generating a highly-diverse repertoire of T lymphocytes while preventing autoimmunity. Thymus seeding progenitors (TSPs) are a heterogeneous group of multipotent progenitors that migrate to the thymus via CCR7 and CCR9 receptors. While NOTCH guides thymus progenitors toward T cell fate, the absence or disruption of NOTCH signaling renders the thymus microenvironment permissive to other cell fates. Following T cell commitment, developing T cells undergo multiple selection checkpoints by engaging with the extracellular matrix, and interacting with thymic epithelial cells (TECs) and other immune subsets across the different compartments of the thymus. The different selection checkpoints assess the T cell receptor (TCR) performance, with failure resulting in either repurposing (agonist selection), or cell death. Additionally, environmental cues such as inflammation and endocrine signaling induce acute thymus atrophy, contributing to the demise of most developing T cells during thymic selection. We discuss the occurrence of acute thymus atrophy in response to systemic inflammation. The thymus demonstrates high plasticity, shaping inflammation by abrogating T cell development and undergoing profound structural changes, and facilitating regeneration and restoration of T cell development once inflammation is resolved. Despite the challenges, thymic selection ensures a highly diverse T cell repertoire capable of discerning between self and non-self antigens, ultimately egressing to secondary lymphoid organs where they complete their maturation and exert their functions.


Subject(s)
Atrophy , T-Lymphocytes , Thymus Gland , Thymus Gland/immunology , Thymus Gland/pathology , Humans , Animals , T-Lymphocytes/immunology , Cell Movement/immunology , Signal Transduction , Cell Differentiation/immunology , Inflammation/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology
8.
Science ; 385(6715): eadd8947, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39298586

ABSTRACT

Humans with monogenic inborn errors responsible for extreme disease phenotypes can reveal essential physiological pathways. We investigated germline mutations in GNAI2, which encodes Gαi2, a key component in heterotrimeric G protein signal transduction usually thought to regulate adenylyl cyclase-mediated cyclic adenosine monophosphate (cAMP) production. Patients with activating Gαi2 mutations had clinical presentations that included impaired immunity. Mutant Gαi2 impaired cell migration and augmented responses to T cell receptor (TCR) stimulation. We found that mutant Gαi2 influenced TCR signaling by sequestering the guanosine triphosphatase (GTPase)-activating protein RASA2, thereby promoting RAS activation and increasing downstream extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)-AKT S6 signaling to drive cellular growth and proliferation.


Subject(s)
GTP-Binding Protein alpha Subunit, Gi2 , Germ-Line Mutation , Receptors, Antigen, T-Cell , T-Lymphocytes , ras GTPase-Activating Proteins , Humans , Cell Movement/genetics , Cell Proliferation , GTP-Binding Protein alpha Subunit, Gi2/genetics , Immunity/genetics , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , ras GTPase-Activating Proteins/genetics , ras Proteins/metabolism , ras Proteins/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Pedigree
9.
J Immunother Cancer ; 12(9)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39266213

ABSTRACT

BACKGROUND: We discovered a novel human endogenous retrovirus (CT-RCC HERV-E) that was selectively expressed in most clear cell renal cell carcinomas (ccRCC) and served as a source of antigens for T cell-mediated killing. Here, we described the cloning of a novel T cell receptor (TCR) targeting a CT-RCC HERV-E-derived antigen specific to ccRCC and characterized antitumor activity of HERV-E TCR-transduced T cells (HERV-E T cells). METHODS: We isolated a CD8+ T cell clone from a patient with immune-mediated regression of ccRCC post-allogeneic stem cell transplant that recognized the CT-RCC-1 HERV-E-derived peptide in an HLA-A11-restricted manner. We used 5'Rapid Amplification of cDNA Ends (RACE) to clone the full length HERV-E TCR and generated retrovirus encoding this TCR for transduction of T cells. We characterized HERV-E T cells for phenotype and function in vitro and in a murine xenograft model. Lastly, we implemented a good manufacturing practice-compliant method for scalable production of HERV-E T cells. RESULTS: The HLA-A11-restricted HERV-E-reactive TCR exhibited a CD8-dependent phenotype and demonstrated specific recognition of the CT-RCC-1 peptide. CD8+ T cells modified to express HERV-E TCR displayed potent antitumor activity against HLA-A11+ ccRCC cells expressing CT-RCC HERV-E compared with unmodified T cells. Killing by HERV-E T cells was lost when cocultured against HERV-E knockout ccRCC cells. HERV-E T cells induced regression of established ccRCC tumors in a murine model and improved survival of tumor-bearing mice. Large-scale production of HERV-E T cells under good manufacturing practice conditions generated from healthy donors retained specific antigen recognition and cytotoxicity against ccRCC. CONCLUSIONS: This is the first report showing that human ccRCC cells can be selectively recognized and killed by TCR-engineered T cells targeting a HERV-derived antigen. These preclinical findings provided the foundation for evaluating HERV-E TCR-transduced T cell infusions in patients with metastatic ccRCC in a clinical trial (NCT03354390).


Subject(s)
Carcinoma, Renal Cell , Endogenous Retroviruses , Kidney Neoplasms , Receptors, Antigen, T-Cell , Humans , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Animals , Mice , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Xenograft Model Antitumor Assays , Cell Line, Tumor , CD8-Positive T-Lymphocytes/immunology
10.
Cancer Med ; 13(17): e70164, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39240157

ABSTRACT

BACKGROUND: T cells, the "superstar" of the immune system, play a crucial role in antitumor immunity. T-cell receptors (TCR) are crucial molecules that enable T cells to identify antigens and start immunological responses. The body has evolved a unique method for rearrangement, resulting in a vast diversity of TCR repertoires. A healthy TCR repertoire is essential for the particular identification of antigens by T cells. METHODS: In this article, we systematically summarized the TCR creation mechanisms and analysis methodologies, particularly focusing on the application of next-generation sequencing (NGS) technology. We explore the TCR repertoire in health and cancer, and discuss the implications of TCR repertoire analysis in understanding carcinogenesis, cancer progression, and treatment. RESULTS: The TCR repertoire analysis has enormous potential for monitoring the emergence and progression of malignancies, as well as assessing therapy response and prognosis. The application of NGS has dramatically accelerated our comprehension of TCR diversity and its role in cancer immunity. CONCLUSIONS: To substantiate the significance of TCR repertoires as biomarkers, more thorough and exhaustive research should be conducted. The TCR repertoire analysis, enabled by advanced sequencing technologies, is poised to become a crucial tool in the future of cancer diagnosis, monitoring, and therapy evaluation.


Subject(s)
High-Throughput Nucleotide Sequencing , Neoplasms , Receptors, Antigen, T-Cell , Humans , Neoplasms/immunology , Neoplasms/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Biomarkers, Tumor/genetics , Prognosis
11.
Sci Rep ; 14(1): 21331, 2024 09 12.
Article in English | MEDLINE | ID: mdl-39266656

ABSTRACT

Programming cell signaling during T-cell activation represents a simple strategy for improving the potency of therapeutic T-cell products. Stim-R technology (Lyell Immunopharma) is a customizable, degradable synthetic cell biomimetic that emulates physiologic, cell-like presentation of signal molecules to control T-cell activation. A breadth of Stim-R formulations with different anti-CD3/anti-CD28 (αCD3/αCD28) antibody densities and stoichiometries were screened for their effects on multiple metrics of T-cell function. We identified an optimized formulation that produced receptor tyrosine kinase-like orphan receptor 1 (ROR1)-targeted chimeric antigen receptor (CAR) T cells with enhanced persistence and polyfunctionality in vitro, as assessed in repeat-stimulation assays, compared with a benchmark product generated using a conventional T-cell-activating reagent. In transcriptomic analyses, CAR T cells activated with Stim-R technology showed downregulation of exhaustion-associated gene sets and retained a unique subset of stem-like cells with effector-associated gene signatures following repeated exposure to tumor cells. Compared with the benchmark product, CAR T cells activated using the optimized Stim-R technology formulation exhibited higher peak expansion, prolonged persistence, and improved tumor control in a solid tumor xenograft model. Enhancing T-cell products with Stim-R technology during T-cell activation may help improve therapeutic efficacy against solid tumors.


Subject(s)
Lymphocyte Activation , Receptors, Chimeric Antigen , Signal Transduction , T-Lymphocytes , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/genetics , Humans , Animals , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Mice , Immunotherapy, Adoptive/methods , Xenograft Model Antitumor Assays , Cell Line, Tumor , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , CD28 Antigens/immunology , CD28 Antigens/metabolism
12.
Cells ; 13(17)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39273050

ABSTRACT

Immunotherapy represents a transformative shift in cancer treatment. Among myriad immune-based approaches, chimeric antigen receptor (CAR) T-cell therapy has shown promising results in treating hematological malignancies. Despite aggressive treatment options, the prognosis for patients with malignant brain tumors remains poor. Research leveraging CAR T-cell therapy for brain tumors has surged in recent years. Pre-clinical models are crucial in evaluating the safety and efficacy of these therapies before they advance to clinical trials. However, current models recapitulate the human tumor environment to varying degrees. Novel in vitro and in vivo techniques offer the opportunity to validate CAR T-cell therapies but also have limitations. By evaluating the strengths and weaknesses of various pre-clinical glioma models, this review aims to provide a roadmap for the development and pre-clinical testing of CAR T-cell therapies for brain tumors.


Subject(s)
Brain Neoplasms , Glioma , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Glioma/therapy , Glioma/immunology , Glioma/pathology , Humans , Animals , Immunotherapy, Adoptive/methods , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , Disease Models, Animal , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology
13.
Sci Adv ; 10(36): eadj4632, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39231214

ABSTRACT

Low antigen sensitivity and a gradual loss of effector functions limit the clinical applicability of chimeric antigen receptor (CAR)-modified T cells and call for alternative antigen receptor designs for effective T cell-based cancer immunotherapy. Here, we applied advanced microscopy to demonstrate that TCR/CD3-based synthetic constructs (TCC) outperform second-generation CAR formats with regard to conveyed antigen sensitivities by up to a thousandfold. TCC-based antigen recognition occurred without adverse nonspecific signaling, which is typically observed in CAR-T cells, and did not depend-unlike sensitized peptide/MHC detection by conventional T cells-on CD4 or CD8 coreceptor engagement. TCC-endowed signaling properties may prove critical when targeting antigens in low abundance and aiming for a durable anticancer response.


Subject(s)
Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , CD3 Complex/metabolism , CD3 Complex/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Immunotherapy, Adoptive/methods , Signal Transduction , Cell Line, Tumor
15.
Cell ; 187(18): 4826-4828, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39241743

ABSTRACT

Early reports suggest that chimeric antigen receptor (CAR)-T therapy has remarkable potential for treating autoimmune disease. Current approaches rely on autologous CAR-T cells, creating a bottleneck to the broad deployment of this therapy. In this issue of Cell, Wang et al.1 report the first use of allogeneic CAR-T cells in three patients with systemic autoimmune disease.


Subject(s)
Autoimmune Diseases , Receptors, Chimeric Antigen , Humans , Autoimmune Diseases/therapy , Autoimmune Diseases/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology
16.
Front Immunol ; 15: 1444424, 2024.
Article in English | MEDLINE | ID: mdl-39234253

ABSTRACT

Lymphocyte activation gene (Lag)-3 is an inhibitory co-receptor and target of immune checkpoint inhibitor (ICI) therapy for cancer. The dynamic behavior of Lag-3 was analyzed at the immune synapse upon T-cell activation to elucidate the Lag-3 inhibitory mechanism. Lag-3 formed clusters and co-localized with T-cell receptor microcluster (TCR-MC) upon T-cell activation similar to PD-1. Lag-3 blocking antibodies (Abs) inhibited the co-localization between Lag-3 and TCR-MC without inhibiting Lag-3 cluster formation. Lag-3 also inhibited MHC-II-independent stimulation and Lag-3 Ab, which did not block MHC-II binding could still block Lag-3's inhibitory function, suggesting that the Lag-3 Ab blocks the Lag-3 inhibitory signal by dissociating the co-assembly of TCR-MC and Lag-3 clusters. Consistent with the combination benefit of PD-1 and Lag-3 Abs to augment T-cell responses, bispecific Lag-3/PD-1 antagonists effectively inhibited both cluster formation and co-localization of PD-1 and Lag-3 with TCR-MC. Therefore, Lag-3 inhibits T-cell activation at TCR-MC, and the target of Lag-3 ICI is to dissociate the co-localization of Lag-3 with TCR-MC.


Subject(s)
Antigens, CD , Immune Checkpoint Inhibitors , Lymphocyte Activation Gene 3 Protein , Lymphocyte Activation , Receptors, Antigen, T-Cell , Immune Checkpoint Inhibitors/pharmacology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Antigens, CD/immunology , Antigens, CD/metabolism , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals
17.
J Immunother Cancer ; 12(9)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39237261

ABSTRACT

The intricate origins, subsets, and characteristics of TCR (T Cell Receptor) s, along with the mechanisms underpinning the antitumor response of tumor-infiltrating T lymphocytes within the tumor microenvironment (TME) remain enigmatic. Recently, the advent of single-cell RNA+TCR-sequencing (scRNA+TCR seq) has revolutionized TME analysis, providing unprecedented insight into the origins, cell subsets, TCR CDR3 compositions, and the expression patterns of response/depletion factors within individual tumor-infiltrating T lymphocytes. Our analysis of the shared scRNA+TCR seq dataset revealed a substantial presence of dual TCR T cells, characterized by clonal hyperplasia and remarkable migratory prowess across various tissues, including blood, normal, peritumoral, and tumor tissues in non-small cell lung cancer patients. Notably, dual TCR CD8+T cells predominantly fell within the CXCL13+subset, displaying potent antitumor activity and a strong preference for tumor tissue residency. Conversely, dual TCR CD4+T cells were predominantly classified as CD5+ or LMNA+subsets, exhibiting a more even distribution across diverse tissue types. By harnessing scRNA+TCR seq and other cutting-edge technologies, we can delve deeper into the effects and mechanisms that regulate the antitumor response or tolerance of dual TCR T cells. This innovative approach holds immense promise in offering fresh perspectives and avenues for advancing research on TIL (Tumor infiltrating lymphocyte)s within the TME.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Receptors, Antigen, T-Cell , Tumor Microenvironment , Humans , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/immunology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Tumor Microenvironment/immunology , Receptors, Antigen, T-Cell/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Single-Cell Analysis/methods
18.
Biol Res ; 57(1): 69, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39342392

ABSTRACT

BACKGROUND: The LYP tyrosine phosphatase presents a SNP (1858C > T) that increases the risk of developing autoimmune diseases such as type I diabetes and arthritis. It remains unclear how this SNP affects LYP function and promotes the development of these diseases. The scarce information about LYP substrates is in part responsible for the poor understanding of LYP function. RESULTS: In this study, we identify in T lymphocytes several adaptor proteins as potential substrates targeted by LYP, including FYB, SLP-76, HS-1, Vav, SKAP1 and SKAP2. We also show that LYP co-localizes with SLP76 in microclusters, upon TCR engagement. CONCLUSIONS: These data indicate that LYP may modulate T cell activation by dephosphorylating several adaptor proteins, such as FYB, SLP-76, HS-1, Vav, SKAP1 and SKAP2 upon TCR engagement.


Subject(s)
Adaptor Proteins, Signal Transducing , Phosphoproteins , Signaling Lymphocytic Activation Molecule Associated Protein , T-Lymphocytes , Humans , Adaptor Proteins, Signal Transducing/metabolism , Jurkat Cells , Lymphocyte Activation , Phosphoproteins/metabolism , Phosphorylation , Protein Tyrosine Phosphatases/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein/genetics , Signaling Lymphocytic Activation Molecule Associated Protein/metabolism
19.
Science ; 385(6714): eadj1979, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39265028

ABSTRACT

T cell receptor (TCR) sensitivity to peptide-major histocompatibility complex (MHC) dictates T cell fate. Canonical models of TCR sensitivity cannot be fully explained by transcriptional regulation. In this work, we identify a posttranscriptional regulatory mechanism of TCR sensitivity that guides alternative splicing of TCR signaling transcripts through an evolutionarily ultraconserved poison exon (PE) in the RNA-binding protein (RBP) TRA2ß in mouse and human. TRA2ß-PE splicing, seen during cancer and infection, was required for TCR-induced effector T cell expansion and function. Tra2ß-PE skipping enhanced T cell response to antigen by increasing TCR sensitivity. As antigen levels decreased, Tra2ß-PE reinclusion allowed T cell survival. Finally, we found that TRA2ß-PE was first included in the genome of jawed vertebrates that were capable of TCR gene rearrangements. We propose that TRA2ß-PE splicing acts as a gatekeeper of TCR sensitivity to shape T cell fate.


Subject(s)
Alternative Splicing , Exons , Receptors, Antigen, T-Cell , Serine-Arginine Splicing Factors , Animals , Humans , Mice , Cell Survival , Conserved Sequence , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Receptors, Antigen, T-Cell/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
20.
Sci Adv ; 10(36): eadn3470, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39231218

ABSTRACT

Regulatory T cells (Treg cells) hold promise for sustainable therapy of immune disorders. Recent advancements in chimeric antigen receptor development and genome editing aim to enhance the specificity and function of Treg cells. However, impurities and functional instability pose challenges for the development of safe gene-edited Treg cell products. Here, we examined different Treg cell subsets regarding their fate, epigenomic stability, transcriptomes, T cell receptor repertoires, and function ex vivo and after manufacturing. Each Treg cell subset displayed distinct features, including lineage stability, epigenomics, surface markers, T cell receptor diversity, and transcriptomics. Earlier-differentiated memory Treg cell populations, including a hitherto unidentified naïve-like memory Treg cell subset, outperformed late-differentiated effector memory-like Treg cells in regulatory function, proliferative capacity, and epigenomic stability. High yields of stable, functional Treg cell products could be achieved by depleting the small effector memory-like Treg cell subset before manufacturing. Considering Treg cell subset composition appears critical to maintain lineage stability in the final cell product.


Subject(s)
Immunologic Memory , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Humans , Phenotype , Memory T Cells/immunology , Memory T Cells/metabolism , Cell Differentiation , Receptors, Antigen, T-Cell/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL