Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 493
Filter
1.
Nat Commun ; 15(1): 8609, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39366979

ABSTRACT

Immune evasion represents a crucial milestone in the progression of cancer and serves as the theoretical foundation for tumor immunotherapy. In this study, we reveal a negative association between Human Papillomavirus (HPV)-encoded circular RNA, circE7, and the infiltration of CD8+ T cells in head and neck squamous cell carcinoma (HNSCC). Both in vitro and in vivo experiments demonstrate that circE7 suppresses the function and activity of T cells by downregulating the transcription of LGALS9, which encodes the galectin-9 protein. The molecular mechanism involves circE7 binding to acetyl-CoA carboxylase 1 (ACC1), promoting its dephosphorylation and thereby activating ACC1. Activated ACC1 reduces H3K27 acetylation at the LGALS9 gene promoter, leading to decreased galectin-9 expression. Notably, galectin-9 interacts with immune checkpoint molecules TIM-3 and PD-1, inhibiting the secretion of cytotoxic cytokines by T cells and promoting T cell apoptosis. Here, we demonstrate a mechanism by which HPV promotes immune evasion in HNSCC through a circE7-driven epigenetic modification and propose a potential immunotherapy strategy for HNSCC that involves the combined use of anti-PD-1 and anti-TIM-3 inhibitors.


Subject(s)
CD8-Positive T-Lymphocytes , Galectins , Head and Neck Neoplasms , Immune Evasion , RNA, Circular , Squamous Cell Carcinoma of Head and Neck , Humans , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/virology , Squamous Cell Carcinoma of Head and Neck/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/virology , Head and Neck Neoplasms/genetics , Animals , Galectins/genetics , Galectins/metabolism , Galectins/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Circular/immunology , Immune Evasion/genetics , Mice , Papillomaviridae/genetics , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Papillomavirus Infections/genetics , Gene Expression Regulation, Neoplastic , Tumor Escape/genetics , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Male , Apoptosis/genetics , Female , Epigenesis, Genetic , RNA, Viral/genetics , RNA, Viral/immunology , Human Papillomavirus Viruses
2.
Oncol Res ; 32(10): 1575-1587, 2024.
Article in English | MEDLINE | ID: mdl-39308517

ABSTRACT

Background: Immune checkpoint ligand-receptor interactions appear to be associated with multiple myeloma (MM) progression. Simultaneously, previous studies showed the possibility of PD-1 and TIM-3 expression on T cells upon stimulation with common γ-chain family cytokines in vitro and during homeostatic proliferation. The aim of the present work was to study the impact of homeostatic proliferation on the expansion of certain T cell subsets up-regulating PD-1 and TIM-3 checkpoint molecules. Methods: The expression of CD25, CD122, CD127 common γ-chain cytokine receptors, phosphorylated signal transducer and activator of transcription-5 (pSTAT5) and eomesodermin (EOMES) was comparatively assessed with flow cytometry in PD-1- and TIM-3-negative and positive T cells before the conditioning and during the first post-transplant month in peripheral blood samples of MM patients. Results: Substantial proportions of PD-1- and TIM-3-positive T lymphocytes expressed common γ-chain cytokine receptors and pSTAT5. Frequencies of cytokine receptor expressing cells were significantly higher within TIM-3+ T cells compared to PD-1+TIM-3- subsets. Considerable proportions of both PD-1-/TIM-3-negative and positive CD8+ T cells express EOMES, while only moderate frequencies of CD4+ PD-1+/TIM-3+ T cells up-regulate this transcription factor. Besides, the surface presence of CD25 and intranuclear expression of EOMES in CD4+ T cells were mutually exclusive regardless of PD-1 and TIM-3 expression. The stimulation with common γ-chain cytokines up-regulates PD-1 and TIM-3 during the proliferation of initially PD-1/TIM-3-negative T cells but fails to expand initially PD-1+ and TIM-3+ T cell subsets in vitro. Conclusions: Both PD-1 and TIM-3 expressing T cells appear to be able to respond to homeostatic cytokine stimulation. Differences in common γ-chain cytokine receptor expression between PD-1+ and TIM-3+ T cells may reflect functional dissimilarity of these cell subsets. Checkpoint blockade appears to alleviate lymphopenia-induced proliferation of PD-1+ T cells but may raise the possibility of immune-mediated adverse events.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Multiple Myeloma , Programmed Cell Death 1 Receptor , Humans , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Hepatitis A Virus Cellular Receptor 2/metabolism , Programmed Cell Death 1 Receptor/metabolism , Middle Aged , Male , Female , Aged , Interleukin-7/metabolism , Interleukin-15/pharmacology , Interleukin-15/metabolism , Up-Regulation , Adult , Receptors, Cytokine/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
Cell Mol Life Sci ; 81(1): 399, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39264416

ABSTRACT

Regulatory T cells (Tregs) play a key role in suppressing systemic effector immune responses, thereby preventing autoimmune diseases but also potentially contributing to tumor progression. Thus, there is great interest in clinically manipulating Tregs, but the precise mechanisms governing in vitro-induced Treg (iTreg) differentiation are not yet fully understood. Here, we used multiparametric mass cytometry to phenotypically profile human iTregs during the early stages of in vitro differentiation at single-cell level. A panel of 25 metal-conjugated antibodies specific to markers associated with human Tregs was used to characterize these immunomodulatory cells. We found that iTregs highly express the transcription factor FOXP3, as well as characteristic Treg-associated surface markers (e.g. CD25, PD1, CD137, CCR4, CCR7, CXCR3, and CD103). Expression of co-inhibitory factors (e.g. TIM3, LAG3, and TIGIT) increased slightly at late stages of iTreg differentiation. Further, CD103 was upregulated on a subpopulation of iTregs with greater suppressive capacity than their CD103- counterparts. Using mass-spectrometry-based proteomics, we showed that sorted CD103+ iTregs express factors associated with immunosuppression. Overall, our study highlights that during early stages of differentiation, iTregs resemble memory-like Treg features with immunosuppressive activity, and provides opportunities for further investigation into the molecular mechanisms underlying Treg function.


Subject(s)
Antigens, CD , Cell Differentiation , Forkhead Transcription Factors , Integrin alpha Chains , T-Lymphocytes, Regulatory , Humans , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Antigens, CD/metabolism , Antigens, CD/immunology , Integrin alpha Chains/metabolism , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/immunology , Phenotype , Hepatitis A Virus Cellular Receptor 2/metabolism , Immune Tolerance , Receptors, Immunologic/metabolism , Proteomics/methods , Receptors, CXCR3/metabolism , Lymphocyte Activation Gene 3 Protein , Cells, Cultured
4.
Commun Biol ; 7(1): 1090, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237613

ABSTRACT

T cell immunoglobulin and mucin-containing molecule 3 (TIM-3) exhibits unique, cell type- and context-dependent characteristics and functions. Here, we report that TIM-3 on myeloid cells plays essential roles in modulating lung inflammation. We found that myeloid cell-specific TIM-3 knock-in (FSF-TIM3/LysM-Cre+) mice have lower body weight and shorter lifespan than WT mice. Intriguingly, the lungs of FSF-TIM3/LysM-Cre+ mice display excessive inflammation and features of disease-associated pathology. We further revealed that galectin-3 levels are notably elevated in TIM-3-overexpressing lung-derived myeloid cells. Furthermore, both TIM-3 blockade and GB1107, a galectin-3 inhibitor, ameliorated lung inflammation in FSF-TIM3/LysM-Cre+/- mice. Using an LPS-induced lung inflammation model with myeloid cell-specific TIM-3 knock-out mice, we demonstrated the association of TIM-3 with both lung inflammation and galectin-3. Collectively, our findings suggest that myeloid TIM-3 is an important regulator in the lungs and that modulation of TIM-3 and galectin-3 could offer therapeutic benefits for inflammation-associated lung diseases.


Subject(s)
Galectin 3 , Hepatitis A Virus Cellular Receptor 2 , Myeloid Cells , Pneumonia , Animals , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/genetics , Galectin 3/metabolism , Galectin 3/genetics , Myeloid Cells/metabolism , Mice , Pneumonia/metabolism , Pneumonia/pathology , Pneumonia/genetics , Mice, Knockout , Mice, Inbred C57BL , Galectins/metabolism , Galectins/genetics , Lung/pathology , Lung/metabolism
5.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273326

ABSTRACT

Due to the genetic diversity between the mother and the fetus, heightened control over the immune system during pregnancy is crucial. Immunological parameters determined by clinicians in women with idiopathic recurrent spontaneous abortion (RSA) include the quantity and activity of Natural Killer (NK) and Natural Killer T (NKT) cells, the quantity of regulatory T lymphocytes, and the ratio of pro-inflammatory cytokines, which indicate imbalances in Th1 and Th2 cell response. The processes are controlled by immune checkpoint proteins (ICPs) expressed on the surface of immune cells. We aim to investigate differences in the expression of ICPs on T cells, T regulatory lymphocytes, NK cells, and NKT cells in peripheral blood samples collected from RSA women, pregnant women, and healthy multiparous women. We aim to discover new insights into the role of ICPs involved in recurrent pregnancy loss. Peripheral blood mononuclear cells (PBMCs) were isolated by gradient centrifugation from blood samples obtained from 10 multiparous women, 20 pregnant women (11-14th week of pregnancy), and 20 RSA women, at maximum of 72 h after miscarriage. The PBMCs were stained for flow cytometry analysis. Standard flow cytometry immunophenotyping of PBMCs was performed using antibodies against classical lymphocyte markers, including CD3, CD4, CD8, CD56, CD25, and CD127. Additionally, ICPs were investigated using antibodies against Programmed Death Protein-1 (PD-1, CD279), T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3, CD366), V-domain Ig suppressor of T cell activation (VISTA), T cell immunoglobulin and ITIM domain (TIGIT), and Lymphocyte activation gene 3 (LAG-3). We observed differences in the surface expression of ICPs in the analyzed subpopulations of lymphocytes between early pregnancy and RSA, after miscarriage, and in women. We noted diminished expression of PD-1 on T lymphocytes (p = 0.0046), T helper cells (CD3CD4 positive cells, p = 0.0165), T cytotoxic cells (CD3CD8 positive cells, p = 0.0046), T regulatory lymphocytes (CD3CD4CD25CD127 low positive cells, p = 0.0106), and NKT cells (CD3CD56/CD16 positive cells, p = 0.0438), as well as LAG-3 on lymphocytes T (p = 0.0225) T helper, p = 0.0426), T cytotoxic cells (p = 0.0458) and Treg (p = 0.0293), and cells from RSA women. Impaired expression of TIM-3 (p = 0.0226) and VISTA (p = 0.0039) on CD8 cytotoxic T and NK (TIM3 p = 0.0482; VISTA p = 0.0118) cells was shown, with an accompanying increased expression of TIGIT (p = 0.0211) on NKT cells. The changes in the expression of surface immune checkpoints indicate their involvement in the regulation of pregnancy. The data might be utilized to develop specific therapies for RSA women based on the modulation of ICP expression.


Subject(s)
Abortion, Habitual , Biomarkers , Immune Checkpoint Proteins , Killer Cells, Natural , Humans , Female , Pregnancy , Abortion, Habitual/immunology , Abortion, Habitual/metabolism , Abortion, Habitual/blood , Adult , Biomarkers/blood , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Immunophenotyping , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Antigens, CD/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Programmed Cell Death 1 Receptor/metabolism
6.
Acta Otorhinolaryngol Ital ; 44(4): 242-251, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39347549

ABSTRACT

Objective: This study evaluated the expression of TIM-3 and its influence on macrophage polarisation in recalcitrant chronic rhinosinusitis with nasal polyps (CRSwNP). Methods: We detected TIM-3 expression in serum and tissue samples of healthy controls (HC), primary CRSwNP, and patients with recurrent CRSwNP. Macrophage markers were detected among three groups, and their correlations with TIM-3 levels were examined. Macrophages from circulating blood were collected and used to examine the impact of TIM-3 on polarisation in vitro. Results: TIM-3 levels were enhanced in the CRSwNP group compared to the HC group. Tissue immunofluorescence revealed elevated TIM-3 expression in patients with CRSwNP, and patients with multiple recurrences exhibited higher TIM-3 levels compared to their first recurrence and baseline levels. Tissue CD163 and CD206 levels were higher in recurrent CRSwNP in comparison with primary cases and HCs, and had a positive correlation with TIM-3 levels. TIM-3 overexpression promoted M2 polarisation and enhanced TGF-ß1 and IL-10 secretion. Conclusions: TIM-3 expression was enhanced in patients with CRSwNP, especially in those undergoing revision surgeries. TIM-3 may be a novel biomarker for recalcitrant CRSwNP. TIM-3-driven M2 polarisation might be involved in the mechanisms of recurrent CRSwNP.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Macrophages , Nasal Polyps , Rhinitis , Sinusitis , Humans , Sinusitis/immunology , Sinusitis/metabolism , Sinusitis/complications , Nasal Polyps/immunology , Nasal Polyps/complications , Nasal Polyps/metabolism , Rhinitis/immunology , Rhinitis/metabolism , Rhinitis/complications , Hepatitis A Virus Cellular Receptor 2/metabolism , Chronic Disease , Male , Female , Macrophages/metabolism , Middle Aged , Adult , Rhinosinusitis
7.
Biomed Pharmacother ; 179: 117388, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39243430

ABSTRACT

CAR T cell therapy for AML remains limited due to the lack of a proper target without on-target off-tumor toxicity. TIM3 is a promising target due to its high expression on AML cells and absence in most normal hematopoietic cells. Previous reports have shown that each CAR component impacts CAR functionality. Here, we optimized TIM-3 targeting CAR T cells for AML therapy. We generated CARs targeting TIM3 with two different non-signaling domains: an IgG2-CH3 spacer with CD28 transmembrane domain (CH3/CD28) and a CD8α spacer with CD8α transmembrane domain (CD8/CD8), and evaluated their characteristics and function. Incorporating the non-signaling CH3/CD28 domain resulted in unstable CAR expression in anti-TIM3 CAR T cells, leading to lower surface CAR expression over time and reduced cytotoxic function compared to anti-TIM3 CARs with the CD8/CD8 domain. Both types of anti-TIM3 CAR T cells transiently exhibited fratricide, which subsided overtime, and both CAR T cells achieved substantial T cell expansion. To further optimize the design, we explored the effects of different costimulatory domains. Compared with CD28 costimulation, 4-1BB and CD27 combined with a CD8/CD8 non-signaling domain showed higher cytokine secretion, superior antitumor activity, and enhanced T-cell persistence after repeated antigen exposure. These findings emphasize the impact of the optimal design of CAR constructs that provide efficient function. In the context of anti-TIM3 CAR T cells, using a CD8α spacer and transmembrane domain with TNFR-based costimulation is a promising CAR design to improve anti-TIM3 CAR T cell function for AML therapy.


Subject(s)
CD8 Antigens , Hepatitis A Virus Cellular Receptor 2 , Immunotherapy, Adoptive , Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/immunology , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/immunology , Animals , Hepatitis A Virus Cellular Receptor 2/metabolism , Immunotherapy, Adoptive/methods , CD8 Antigens/metabolism , CD8 Antigens/immunology , Cell Line, Tumor , Mice , CD28 Antigens/immunology , CD28 Antigens/metabolism , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Mice, Inbred NOD
8.
Cell Rep Med ; 5(8): 101686, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39168104

ABSTRACT

Resistance to PD-1 blockade in onco-immunotherapy greatly limits its clinical application. T cell immunoglobulin and mucin domain containing-3 (Tim-3), a promising immune checkpoint target, is cleaved by ADAM10/17 to produce its soluble form (sTim-3) in humans, potentially becoming involved in anti-PD-1 resistance. Herein, serum sTim-3 upregulation was observed in non-small cell lung cancer (NSCLC) and various digestive tumors. Notably, serum sTim-3 is further upregulated in non-responding patients undergoing anti-PD-1 therapy for NSCLC and anti-PD-1-resistant cholangiocarcinoma patients. Furthermore, sTim-3 overexpression facilitates tumor progression and confers anti-PD-1 resistance in multiple tumor mouse models. Mechanistically, sTim-3 induces terminal T cell exhaustion and attenuates CD8+ T cell response to PD-1 blockade through carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM-1). Moreover, the ADAM10 inhibitor GI254023X, which blocks sTim-3 production, reduces tumor progression in Tim-3 humanized mice and reverses anti-PD-1 resistance in human tumor-infiltrating lymphocytes (TILs). Overall, human sTim-3 holds great predictive and therapeutic potential in onco-immunotherapy.


Subject(s)
Biomarkers, Tumor , CD8-Positive T-Lymphocytes , Drug Resistance, Neoplasm , Hepatitis A Virus Cellular Receptor 2 , Programmed Cell Death 1 Receptor , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Humans , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , Drug Resistance, Neoplasm/drug effects , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Prognosis , Biomarkers, Tumor/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Cell Line, Tumor , Female , Male , Lung Neoplasms/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , ADAM10 Protein/metabolism , Mice, Inbred C57BL , T-Cell Exhaustion
9.
Front Immunol ; 15: 1407403, 2024.
Article in English | MEDLINE | ID: mdl-39206199

ABSTRACT

T cell immunoglobulin and mucin domain-3 (TIM-3), a crucial immune checkpoint following PD1 and CTLA4, is widely found in several immune cells. Nonetheless, its performance in recent clinical trials appears disappointing. Ovarian cancer (OC), a malignant tumor with a high mortality rate in gynecology, faces significant hurdles in immunotherapy. The broad presence of TIM-3 offers a new opportunity for immunotherapy in OC. This study reviews the role of TIM-3 in OC and assesses its potential as a target for immunotherapy. The regulatory effects of TIM-3 on the immune microenvironment in OC are discussed, with a focus on preclinical studies that demonstrate TIM-3's modulation of various immune cells in OC. Additionally, the potential therapeutic advantages and challenges of targeting TIM-3 in OC are examined.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Ovarian Neoplasms , Tumor Microenvironment , Humans , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/immunology , Female , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/therapy , Tumor Microenvironment/immunology , Animals , Immunotherapy/methods
10.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(8): 737-742, 2024 Aug.
Article in Chinese | MEDLINE | ID: mdl-39215671

ABSTRACT

T-cell immunoglobulin and mucin domain 3 (TIM3) is an immune checkpoint protein expressed on various types of immune cells in two forms, namely membrane TIM3 (mTIM3) and soluble TIM3 (sTIM3). mTIM3 and sTIM3 share the same ligands, but less is known about functions and mechanisms of sTIM3 compared to those of mTlM3. Abnormal levels of plasma sTIM3 have been observed in patients with many diseases including cancer. Current research mainly focuses on the potential of sTIM3 as a biomarker for the diagnosis and treatment of cancer. However, there are limited investigations on sTIM3 as a target for immunotherapy. This paper provides a literature review on the recent research progress of the production and biological functions of sTIM3 and its clinical significance in cancer immunotherapy.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Immunotherapy , Neoplasms , Humans , Neoplasms/immunology , Neoplasms/therapy , Hepatitis A Virus Cellular Receptor 2/immunology , Hepatitis A Virus Cellular Receptor 2/metabolism , Immunotherapy/methods , Animals , Biomarkers, Tumor/immunology
11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(8): 754-760, 2024 Aug.
Article in Chinese | MEDLINE | ID: mdl-39215674

ABSTRACT

With the ongoing advancement of immune checkpoint research, targeting tumors through immune checkpoint blockade has emerged as a crucial approach in cancer therapy. T cell immunoglobulin and mucin-domain containing protein 3 (TIM-3) functions as a negative immune checkpoint. It has been demonstrated that the interaction of TIM-3 with its ligand galectin-9 (Gal-9) can promote immune escape in a variety of cancers. In hematologic, digestive, and respiratory tumors, it affects different signaling pathways by blocking TIM-3/Gal-9 interaction, thereby regulating the growth of T cells, B cells, dendritic cells, NK cells, and monocytes/macrophages, and inhibiting regulatory T cells to exert anti-tumor effects. TIM-3 antibodies have potential therapeutic value as immune checkpoint inhibitors in molecularly-targeted anti-tumor therapy. This article reviews the mechanisms of anti-tumor effects of TIM-3/Gal-9 in the tumor microenvironment of various cancers.


Subject(s)
Galectins , Hepatitis A Virus Cellular Receptor 2 , Neoplasms , Humans , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/immunology , Galectins/metabolism , Galectins/immunology , Neoplasms/immunology , Neoplasms/metabolism , Animals , Tumor Microenvironment/immunology , Signal Transduction
12.
Bull Exp Biol Med ; 177(3): 287-292, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39123087

ABSTRACT

T-cell immunoglobulin and mucin domain 3 (TIM-3) belongs to the group of inhibitory checkpoint receptors and has traditionally been of interest in terms of its expression on activated CD4+ and CD8+ T cells. The treatment with TIM-3 inhibitors is considered as a promising strategy in cancer immunotherapy. The review focuses on new data on the expression of TIM-3 on dendritic cells (DCs) that play a key role in initiating the antigen-specific immune response and inducing effector CD8+ T cells. The main hypothesis is that TIM-3 is suggested to act as a negative regulator of DCs. Further studies on TIM-3-mediated DC regulation will improve the effectiveness of current strategies in the treatment of cancer using DCs and checkpoint molecule inhibitors, where the main targets can be not only T cells, but also TIM-3-expressing DCs.


Subject(s)
CD8-Positive T-Lymphocytes , Dendritic Cells , Hepatitis A Virus Cellular Receptor 2 , Dendritic Cells/immunology , Dendritic Cells/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/genetics , Humans , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , Animals , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Immunotherapy/methods , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use
13.
Eur J Oral Sci ; 132(5): e13010, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39090710

ABSTRACT

The aim of this study was to examine the expression of programmed death-ligand 1 (PD-L1) and of T cell immunoglobulin and mucin domain-containing protein (TIM3) in oral epithelial dysplasia and head and neck squamous cell carcinoma (HNSCC). Mouse HNSCC was induced with 4-nitroquinoline-1 oxide (4NQO). Oral epithelial dysplastic lesions, carcinoma in situ and HNSCC lesions were stained with anti-PD-L1 and TIM3 antibodies. The expression of PD-L1 and TIM3 in tumor cells and immune cells was semiquantitatively measured and compared. In parallel, human dysplasia and HNSCC were stained with anti-PD-L1 and anti-TIM3. The expression pattern of PD-L1+ and TIM3+ cells was further compared. In human and mouse samples both PD-L1 and TIM3 were found to be expressed in neoplastic and immune cells in HNSCC, but not in dysplasia. There was no significant difference in PD-L1 and TIM3 expression between metastatic and nonmetastatic HNSCC. We conclude that the 4NQO-induced mouse HNSCC model may be an excellent preclinical model for immune checkpoint therapy.


Subject(s)
B7-H1 Antigen , Head and Neck Neoplasms , Hepatitis A Virus Cellular Receptor 2 , Squamous Cell Carcinoma of Head and Neck , Animals , Hepatitis A Virus Cellular Receptor 2/metabolism , B7-H1 Antigen/metabolism , Mice , Humans , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , 4-Nitroquinoline-1-oxide , Female
14.
JCI Insight ; 9(18)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39146023

ABSTRACT

Pediatric diffuse midline gliomas (DMG) with altered H3-K27M are aggressive brain tumors that arise during childhood. Despite advances in genomic knowledge and the significant number of clinical trials testing new targeted therapies, patient outcomes are still poor. Immune checkpoint blockades with small molecules, such as aptamers, are opening new therapeutic options that represent hope for this orphan disease. Here, we demonstrated that a TIM-3 aptamer (TIM-3 Apt) as monotherapy increased the immune infiltration and elicited a strong specific immune response with a tendency to improve the overall survival of treated DMG-bearing mice. Importantly, combining TIM-3 Apt with radiotherapy increased the overall median survival and led to long-term survivor mice in 2 pediatric DMG orthotopic murine models. Interestingly, TIM-3 Apt administration increased the number of myeloid populations and the proinflammatory CD8-to-Tregs ratios in the tumor microenvironment as compared with nontreated groups after radiotherapy. Importantly, the depletion of T cells led to a major loss of the therapeutic effect achieved by the combination. This work uncovers TIM-3 targeting as an immunotherapy approach to improve the radiotherapy outcome in DMGs and offers a strong foundation for propelling a phase I clinical trial using radiotherapy and TIM-3 blockade combination as a treatment for these tumors.


Subject(s)
Aptamers, Nucleotide , Brain Neoplasms , Glioma , Hepatitis A Virus Cellular Receptor 2 , Animals , Mice , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/genetics , Aptamers, Nucleotide/pharmacology , Aptamers, Nucleotide/therapeutic use , Glioma/radiotherapy , Glioma/pathology , Glioma/mortality , Glioma/drug therapy , Glioma/therapy , Glioma/immunology , Brain Neoplasms/radiotherapy , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Disease Models, Animal , Cell Line, Tumor , Combined Modality Therapy/methods , Female
15.
J Clin Invest ; 134(19)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39137048

ABSTRACT

Despite being the leading cause of cancer-related childhood mortality, pediatric gliomas have been relatively understudied, and the repurposing of immunotherapies has not been successful. Whole-transcriptome sequencing, single-cell sequencing, and sequential multiplex immunofluorescence were used to identify an immunotherapeutic strategy that could be applied to multiple preclinical glioma models. MAPK-driven pediatric gliomas have a higher IFN signature relative to other molecular subgroups. Single-cell sequencing identified an activated and cytotoxic microglia (MG) population designated MG-Act in BRAF-fused, MAPK-activated pilocytic astrocytoma (PA), but not in high-grade gliomas or normal brain. T cell immunoglobulin and mucin domain 3 (TIM3) was expressed on MG-Act and on the myeloid cells lining the tumor vasculature but not normal brain vasculature. TIM3 expression became upregulated on immune cells in the PA microenvironment, and anti-TIM3 reprogrammed ex vivo immune cells from human PAs to a proinflammatory cytotoxic phenotype. In a genetically engineered murine model of MAPK-driven, low-grade gliomas, anti-TIM3 treatment increased median survival over IgG- and anti-PD-1-treated mice. Single-cell RNA-Seq data during the therapeutic window of anti-TIM3 revealed enrichment of the MG-Act population. The therapeutic activity of anti-TIM3 was abrogated in mice on the CX3CR1 MG-KO background. These data support the use of anti-TIM3 in clinical trials of pediatric low-grade, MAPK-driven gliomas.


Subject(s)
Astrocytoma , Brain Neoplasms , Hepatitis A Virus Cellular Receptor 2 , Proto-Oncogene Proteins B-raf , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/immunology , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Animals , Mice , Proto-Oncogene Proteins B-raf/genetics , Astrocytoma/genetics , Astrocytoma/immunology , Astrocytoma/pathology , Astrocytoma/therapy , Astrocytoma/metabolism , Child , Brain Neoplasms/immunology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Female , Tumor Microenvironment/immunology , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Glioma/immunology , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Glioma/therapy
16.
JCI Insight ; 9(16)2024 08 22.
Article in English | MEDLINE | ID: mdl-39171524

ABSTRACT

Decidual regulatory T cells (Tregs) are essential for successful pregnancy outcome. A subset of Tregs, T cell immunoglobulin and mucin domain-containing protein 3-positive regulatory T cells (TregsTim-3+), plays a central role in the acceptance of the fetus during early stages of normal pregnancy. The molecular mechanism regulating the differentiation and function of TregsTim-3+ is unknown. Here, we investigated the role of the transcription factor B lymphocyte-induced maturation protein 1 (Blimp-1) on decidual TregTim-3+ differentiation. We demonstrated that Blimp-1 enhanced the coexpression of negative costimulatory molecules (Tim-3, T cell immunoreceptor with Ig and ITIM domains, and programmed cell death protein 1) on Tregs and improved their immunosuppressive functions, including increased IL-10 secretion, suppression of effector T cell proliferation, and promotion of macrophage polarization toward the M2 phenotype. Furthermore, we showed that IL-27 regulated the expression of Tim-3 and Blimp-1 through the STAT1 signaling pathway and that transfer of TregsBlimp-1+ into an abortion-prone mouse model effectively reduced embryo absorption rate. We postulated that abnormalities in the IL-27/Blimp-1 axis might be associated with recurrent pregnancy loss (RPL). These findings provided insights for developing more efficient immunotherapies for women with RPL.


Subject(s)
Cell Differentiation , Hepatitis A Virus Cellular Receptor 2 , Positive Regulatory Domain I-Binding Factor 1 , T-Lymphocytes, Regulatory , Female , Animals , Pregnancy , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Mice , Cell Differentiation/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Humans , Decidua/immunology , Decidua/metabolism , Decidua/cytology , Interleukin-10/metabolism , Interleukins/metabolism , Interleukins/immunology , Interleukin-27/metabolism
17.
J Cell Mol Med ; 28(14): e18552, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39054581

ABSTRACT

Acute myeloid leukaemia (AML) is a biologically heterogeneous haematological malignancy. This study was performed to identify the potential biomarkers for the prognosis and treatment of AML. We applied weighted gene co-expression network analysis to identify key modules and hub genes related to the prognosis of AML using data from The Cancer Genome Atlas (TCGA). In total, 1581 differentially expressed genes (1096 upregulated and 485 downregulated) were identified between AML patients and healthy controls, with the blue module being the most significant among 14 modules associated with AML morphology. Through functional enrichment analysis, we identified 217 genes in the blue module significantly enriched in 'neutrophil degranulation' and 'neutrophil activation involved in immune response' pathways. The survival analysis revealed six genes (S100A9, S100A8, HK3, CD93, CXCR2 and FGL2) located in the significantly enriched pathway that were notably related to AML survival. We validated the expression of these six genes at gene and single-cell levels and identified methylation loci of each gene, except for S100A8. Finally, in vitro experiments were performed to demonstrate whether the identified hub genes were associated with AML survival. After knockdown of CD93 and FGL2, cell proliferation was significantly reduced in U937 cell line over 5 days. In summary, we identified CD93 and FGL2 as key hub genes related to AML survival, with FGL2 being a novel biomarker for the prognosis and treatment of AML.


Subject(s)
Biomarkers, Tumor , Gene Regulatory Networks , Leukemia, Myeloid, Acute , Receptors, Complement , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/metabolism , Biomarkers, Tumor/genetics , Prognosis , Receptors, Complement/genetics , Receptors, Complement/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Leukemic , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Gene Expression Profiling , Cell Line, Tumor , DNA Methylation/genetics , Survival Analysis , Fibrinogen
18.
Clin Exp Immunol ; 218(1): 55-64, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-38975703

ABSTRACT

Members of the T-cell immunoglobulin and mucin (TIM) family, which is crucial for T-cell function, are implicated in autoimmunity. TIM-1 and -3 play distinct roles in autoimmunity, with TIM-1 acting as a costimulatory molecule and TIM-3 regulating Th1 responses. We investigated the therapeutic potential of anti-TIM-1 (RMT1-10) and anti-TIM-3 (RMT3-23) antibodies in an autoimmune arthritis model. Zymosan A was used to induce arthritis in female SKG mice. The arthritis scores, histology, mRNA expression, cytokine levels, micro-computed tomography, and flow cytometry results were obtained. The application of RMT1-10 reduced the arthritis scores, histological damage, and CD4+ T-cell infiltrations, and it suppressed interleukin (IL)-6 and -17A and reduced TIM-3 mRNA expressions. RMT3-23 also lowered arthritis severity, improved histology, and reduced serum levels of tumor necrosis factor (TNF)-α and IL-17A. RMT3-23 inhibited intracellular TNF-α and IL-6 and early apoptosis. An amelioration of autoimmune arthritis was achieved by blocking the TIM-1 and -3 signaling pathways via RMT1-10 and RMT3-23 administration, leading to a widespread decrease in inflammatory cytokines. Both antibodies exhibited therapeutic effects, suggesting TIM-1 and -3 as potential targets for rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid , Disease Models, Animal , Hepatitis A Virus Cellular Receptor 1 , Hepatitis A Virus Cellular Receptor 2 , Signal Transduction , Animals , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Mice , Hepatitis A Virus Cellular Receptor 1/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Female , Interleukin-6/metabolism , Interleukin-6/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Interleukin-17/metabolism , Interleukin-17/antagonists & inhibitors , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism
19.
Cell Biochem Biophys ; 82(3): 2759-2766, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38995531

ABSTRACT

The immune system plays a pivotal role in controlling chronic myeloid leukemia (CML). CD8+ T cell exhaustion results in reduced effectiveness of T cell-mediated immunity, thereby contributing to disease progression. This study intends to figure out whether the combined blockade of inhibitory molecules TIM-3/PD-1 can affect CD8+ T cell exhaustion in CML. A CML mouse model was established via transplantation of bone marrow cells transduced with BCR-ABL-expressing retrovirus vectors. PD-1 and TIM-3 signaling were blocked using corresponding molecular antibodies. Flow cytometry analysis was conducted to detect cell surface molecules and intracellular cytokines. ELISA was employed for measuring cytokine concentrations in the culture medium. The results showed that TIM-3 and PD-1 were coexpressed on exhausted CD8+ T cells from CML mice. Combined blockade of PD-1/TIM3 synergistically delayed CML progression in mice. Moreover, ex vivo experiments showed that their co-blockade promoted the proliferation and cytokine secretion of CD8+ T cells isolated from CML mice. In conclusion, blocking TIM-3 and PD-1 improves exhausted CD8+ T cell function in CML.


Subject(s)
CD8-Positive T-Lymphocytes , Disease Models, Animal , Hepatitis A Virus Cellular Receptor 2 , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Programmed Cell Death 1 Receptor , Animals , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Mice, Inbred C57BL , Cytokines/metabolism , T-Cell Exhaustion
20.
Allergy ; 79(9): 2435-2447, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39021347

ABSTRACT

BACKGROUND: Galectin-9 (Gal-9) has been implicated in allergic and autoimmune diseases, but its role and relevance in chronic spontaneous urticaria (CSU) are unclear. OBJECTIVES: To characterize the role and relevance of Gal-9 in the pathogenesis of CSU. METHODS: We assessed 60 CSU patients for their expression of Gal-9 on circulating eosinophils and basophils as well as T cell expression of the Gal-9 receptor TIM-3, compared them with 26 healthy controls (HCs), and explored possible links with disease features including disease activity (urticaria activity score, UAS), total IgE, basophil activation test (BAT), and response to omalizumab treatment. We also investigated potential drivers of Gal-9 expression by eosinophils and basophils. RESULTS: Our CSU patients had markedly increased rates of circulating Gal-9+ eosinophils and basophils and high numbers of lesional Gal-9+ cells. High rates of blood Gal-9+ eosinophils/basophils were linked to high disease activity, IgE levels, and BAT negativity. Serum levels of TNF-α were positively correlated with circulating Gal-9+ eosinophils/basophils, and TNF-α markedly upregulated Gal-9 on eosinophils. CSU patients who responded to omalizumab treatment had more Gal-9+ eosinophils/basophils than non-responders, and omalizumab reduced blood levels of Gal-9+ eosinophils/basophils in responders. Gal-9+ eosinophils/basophils were negatively correlated with TIM-3+TH17 cells. CONCLUSION: Our findings demonstrate a previously unrecognized involvement of the Gal-9/TIM-3 pathway in the pathogenesis CSU and call for studies that explore its relevance.


Subject(s)
Anti-Allergic Agents , Basophils , Biomarkers , Chronic Urticaria , Eosinophils , Galectins , Omalizumab , Adult , Female , Humans , Male , Middle Aged , Young Adult , Anti-Allergic Agents/therapeutic use , Anti-Allergic Agents/pharmacology , Basophils/metabolism , Basophils/immunology , Case-Control Studies , Chronic Urticaria/drug therapy , Eosinophils/metabolism , Eosinophils/immunology , Galectins/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Omalizumab/therapeutic use , Omalizumab/pharmacology , Severity of Illness Index , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL