Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 353
Filter
1.
Nature ; 629(8011): 426-434, 2024 May.
Article in English | MEDLINE | ID: mdl-38658764

ABSTRACT

Expansion of antigen-experienced CD8+ T cells is critical for the success of tumour-infiltrating lymphocyte (TIL)-adoptive cell therapy (ACT) in patients with cancer1. Interleukin-2 (IL-2) acts as a key regulator of CD8+ cytotoxic T lymphocyte functions by promoting expansion and cytotoxic capability2,3. Therefore, it is essential to comprehend mechanistic barriers to IL-2 sensing in the tumour microenvironment to implement strategies to reinvigorate IL-2 responsiveness and T cell antitumour responses. Here we report that prostaglandin E2 (PGE2), a known negative regulator of immune response in the tumour microenvironment4,5, is present at high concentrations in tumour tissue from patients and leads to impaired IL-2 sensing in human CD8+ TILs via the PGE2 receptors EP2 and EP4. Mechanistically, PGE2 inhibits IL-2 sensing in TILs by downregulating the IL-2Rγc chain, resulting in defective assembly of IL-2Rß-IL2Rγc membrane dimers. This results in impaired IL-2-mTOR adaptation and PGC1α transcriptional repression, causing oxidative stress and ferroptotic cell death in tumour-reactive TILs. Inhibition of PGE2 signalling to EP2 and EP4 during TIL expansion for ACT resulted in increased IL-2 sensing, leading to enhanced proliferation of tumour-reactive TILs and enhanced tumour control once the cells were transferred in vivo. Our study reveals fundamental features that underlie impairment of human TILs mediated by PGE2 in the tumour microenvironment. These findings have therapeutic implications for cancer immunotherapy and cell therapy, and enable the development of targeted strategies to enhance IL-2 sensing and amplify the IL-2 response in TILs, thereby promoting the expansion of effector T cells with enhanced therapeutic potential.


Subject(s)
CD8-Positive T-Lymphocytes , Cell Proliferation , Dinoprostone , Interleukin-2 , Lymphocytes, Tumor-Infiltrating , Mitochondria , Signal Transduction , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dinoprostone/metabolism , Down-Regulation , Ferroptosis , Interleukin Receptor Common gamma Subunit/biosynthesis , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/metabolism , Interleukin-2/antagonists & inhibitors , Interleukin-2/immunology , Interleukin-2/metabolism , Interleukin-2 Receptor beta Subunit/metabolism , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mitochondria/metabolism , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Microenvironment/immunology
2.
Int Immunopharmacol ; 133: 112071, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38636374

ABSTRACT

Microglia play a pivotal role in the neuroinflammatory response after brain injury, and their proliferation is dependent on colony-stimulating factors. In the present study, we investigated the effect of inhibiting microglia proliferation on neurological damage post intracerebral hemorrhage (ICH) in a mouse model, an aspect that has never been studied before. Using a colony-stimulating factor-1 receptor antagonist (GW2580), we observed that inhibition of microglia proliferation significantly ameliorated neurobehavioral deficits, attenuated cerebral edema, and reduced hematoma volume after ICH. This intervention was associated with a decrease in pro-inflammatory factors in microglia and an increased infiltration of peripheral regulatory CD8 + CD122+ T cells into the injured brain tissue. The CXCR3/CXCL10 axis is the mechanism of brain homing of regulatory CD8 + CD122+ T cells, and the high expression of IL-10 is the hallmark of their synergistic anti-inflammatory effect with microglia. And activated astrocytes around the insult site are a prominent source of CXCL10. Thus, inhibition of microglial proliferation offers a new perspective for clinical translation. The cross-talk between multiple cells involved in the regulation of the inflammatory response highlights the comprehensive nature of neuroimmunomodulation.


Subject(s)
Brain , Cell Proliferation , Cerebral Hemorrhage , Chemokine CXCL10 , Mice, Inbred C57BL , Microglia , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor , Animals , Microglia/drug effects , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/immunology , Cell Proliferation/drug effects , Male , Mice , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Brain/pathology , Brain/drug effects , Brain/metabolism , Brain/immunology , Chemokine CXCL10/metabolism , Disease Models, Animal , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Receptors, CXCR3/metabolism , Receptors, CXCR3/antagonists & inhibitors , Interleukin-2 Receptor beta Subunit/metabolism , Interleukin-10/metabolism , Anisoles , Pyrimidines
3.
Exp Cell Res ; 437(2): 114012, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38565343

ABSTRACT

Ovarian cancer is one of the most common gynecological tumors worldwide. Despite the availability of multiple treatments for ovarian cancer, its resistance to chemotherapy remains a significant challenge. miRNAs play crucial roles in the initiation and progression of cancer by affecting processes such as differentiation, proliferation, and chemoresistance. According to microarray and qPCR analyses, miR-7704 is significantly downregulated in cisplatin-resistant cells compared to parental cells. In this study, we found that miR-7704 inhibited the proliferation and promoted cisplatin sensitivity of ovarian cancer cells in vitro and in vivo. Moreover, ectopic expression of miR-7704 had the same effect as IL2RB knockdown. Further mechanistic studies revealed that miR-7704 played an inhibitory role by regulating IL2RB expression to inactivate the AKT signaling pathway. Furthermore, IL2RB reversed the miR-7704 mediated resistance to cisplatin in ovarian cancer. Based on these findings, miR-7704 and IL2RB show the potential as novel therapeutic targets for ovarian cancer.


Subject(s)
MicroRNAs , Ovarian Neoplasms , Female , Humans , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Feedback , Gene Expression Regulation, Neoplastic , Interleukin-2 Receptor beta Subunit/metabolism , Interleukin-2 Receptor beta Subunit/pharmacology , Interleukin-2 Receptor beta Subunit/therapeutic use , MicroRNAs/metabolism , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism
4.
Commun Biol ; 6(1): 828, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37558752

ABSTRACT

Interleukin-2 (IL-2) engineered versions, with biased immunological functions, have emerged from yeast display and rational design. Here we reshaped the human IL-2 interface with the IL-2 receptor beta chain through the screening of phage-displayed libraries. Multiple beta super-binders were obtained, having increased receptor binding ability and improved developability profiles. Selected variants exhibit an accumulation of negatively charged residues at the interface, which provides a better electrostatic complementarity to the beta chain, and faster association kinetics. These findings point to mechanistic differences with the already reported superkines, characterized by a conformational switch due to the rearrangement of the hydrophobic core. The molecular bases of the favourable developability profile were tracked to a single residue: L92. Recombinant Fc-fusion proteins including our variants are superior to those based on H9 superkine in terms of expression levels in mammalian cells, aggregation resistance, stability, in vivo enhancement of immune effector responses, and anti-tumour effect.


Subject(s)
Directed Molecular Evolution , Interleukin-2 Receptor beta Subunit , Interleukin-2 , Peptide Library , Humans , Interleukin-2 Receptor beta Subunit/chemistry , Interleukin-2/chemistry , Interleukin-2/genetics , Interleukin-2/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Directed Molecular Evolution/methods , Protein Domains , Animals , Mice , Cell Line, Tumor
5.
Allergol Immunopathol (Madr) ; 51(3): 1-7, 2023.
Article in English | MEDLINE | ID: mdl-37169553

ABSTRACT

BACKGROUND: Immune dysfunction is a common and serious complication of sepsis. This study finds key genes linked to immunity in sepsis. METHODS: The "Limma package" was used to analyze GSE154918 datasets for differentially expressed genes. The differentially expressed genes were then enriched for Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, and interleukin 2 receptor subunit Beta (IL2RB) protein coding gene was chosen for investigation. IL2RB expression in peripheral blood mononuclear cells (PBMC) was assessed by polymerase chain reaction. White blood cells of septic patients and healthy controls were collected from hospitals and linked with acute physiology and chronic health evaluation (APACHE) II, sequential organ failure assessment (SOFA), C-reactive protein (CRP), and procalcitonin (PCT) of septic patients using Pearson's correlation analysis. PBMC cells were transfected with IL2RB, and the effect of transfection was observed on cellular interferon gamma (IFN-γ), interleukin (IL)-12, IL-4, IL-10, and IL-17A. RESULTS: A total of 686 differential genes, comprising 446 upregulated and 240 down regulated genes, were identified. The enrichment of KEGG pathway revealed that the majority of differential genes were enriched in the T helper (Th1)/Th2 cell and Th17 cell differentiation pathways. In patients with sepsis, correlation analysis revealed a negative correlation between IL2RB and APACHE II score, SOFA score, CRP, and PCT. IFN-γ and IL-12 levels were elevated in PBMC of septic patients after IL2RB transfection, but IL-4, IL-10, and IL-17A levels were lowered. CONCLUSION: Sepsis-induced immunological dysfunction is improved by IL2RB, which also balances Th1/Th2 responses and prevents Th17 activation. © 2023 Codon Publications. Published by Codon Publications.


Subject(s)
Leukocytes, Mononuclear , Sepsis , Humans , C-Reactive Protein , Interleukin-10 , Interleukin-12 , Interleukin-17 , Interleukin-2 Receptor beta Subunit , Interleukin-4 , Sepsis/genetics , Sepsis/immunology , Th1 Cells , Th17 Cells , Th2 Cells
6.
Nature ; 610(7930): 173-181, 2022 10.
Article in English | MEDLINE | ID: mdl-36171288

ABSTRACT

Combination therapy with PD-1 blockade and IL-2 is highly effective during chronic lymphocytic choriomeningitis virus infection1. Here we examine the underlying basis for this synergy. We show that PD-1 + IL-2 combination therapy, in contrast to PD-1 monotherapy, substantially changes the differentiation program of the PD-1+TCF1+ stem-like CD8+ T cells and results in the generation of transcriptionally and epigenetically distinct effector CD8+ T cells that resemble highly functional effector CD8+ T cells seen after an acute viral infection. The generation of these qualitatively superior CD8+ T cells that mediate viral control underlies the synergy between PD-1 and IL-2. Our results show that the PD-1+TCF1+ stem-like CD8+ T cells, also referred to as precursors of exhausted CD8+ T cells, are not fate-locked into the exhaustion program and their differentiation trajectory can be changed by IL-2 signals. These virus-specific effector CD8+ T cells emerging from the stem-like CD8+ T cells after combination therapy expressed increased levels of the high-affinity IL-2 trimeric (CD25-CD122-CD132) receptor. This was not seen after PD-1 blockade alone. Finally, we show that CD25 engagement with IL-2 has an important role in the observed synergy between IL-2 cytokine and PD-1 blockade. Either blocking CD25 with an antibody or using a mutated version of IL-2 that does not bind to CD25 but still binds to CD122 and CD132 almost completely abrogated the synergistic effects observed after PD-1 + IL-2 combination therapy. There is considerable interest in PD-1 + IL-2 combination therapy for patients with cancer2,3, and our fundamental studies defining the underlying mechanisms of how IL-2 synergizes with PD-1 blockade should inform these human translational studies.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-2 , Programmed Cell Death 1 Receptor , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Drug Therapy, Combination , Humans , Interleukin Receptor Common gamma Subunit , Interleukin-2/immunology , Interleukin-2/pharmacology , Interleukin-2/therapeutic use , Interleukin-2 Receptor alpha Subunit , Interleukin-2 Receptor beta Subunit , Lymphocytic Choriomeningitis/drug therapy , Lymphocytic Choriomeningitis/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T Cell Transcription Factor 1
7.
Biomed Res Int ; 2022: 2114699, 2022.
Article in English | MEDLINE | ID: mdl-35924269

ABSTRACT

Aims: The clinical diagnosis of Kawasaki disease (KD) is not easy because of many atypical manifestations. This study is aimed at finding potential diagnostic markers and therapeutic targets for KD and analysing their correlation with immune cell infiltrations. Methods: First, we downloaded the KD dataset from the Gene Expression Omnibus (GEO) database and used R software to identify differentially expressed genes (DEGs) and perform functional correlation analysis. Then, CIBERSORT algorithm was used to evaluate immune cell infiltrations in samples. Coexpression analysis between DEGs and infiltrating immune cells was performed to screen the main infiltrating immune cells. Subsequently, the least absolute shrinkage and selection operator (LASSO) logistic regression analysis was used to screen the core genes related to KD. Finally, correlation analysis between the core genes and the main infiltrating immune cells was performed. Results: 327 DEGs were screened out in this study. Among them, 72 shared genes were the category of genes most likely to be disease-causing for they did not change before and after treatment. After analysis, it was found that expression level of IL2RB in KD tissues was significantly upregulated, the number of resting CD4+ memory T cells was decreased, and the decrease was significantly negatively correlated with the upregulated expression of IL2RB. Therefore, it was speculated that the upregulated expression of IL2RB disrupted Th1/Th2 cell differentiation balance, which led to a decrease of resting CD4+ memory T cells and finally caused disorder of immune microenvironment in patients with KD. Conclusions: Upregulated expression of IL2RB leads to disorder of immune microenvironment in patients with KD and eventually causes the occurrence and development of KD. Therefore, IL2RB may serve as a diagnostic marker and potential therapeutic target for KD.


Subject(s)
Gene Regulatory Networks , Mucocutaneous Lymph Node Syndrome , CD4-Positive T-Lymphocytes , Gene Expression Profiling , Humans , Interleukin-2 Receptor beta Subunit/genetics , Mucocutaneous Lymph Node Syndrome/genetics
8.
Gene ; 827: 146472, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35381314

ABSTRACT

BACKGROUND: Abdominal aortic aneurysm (AAA) is potentially life threatening and characterized by immune-inflammatory cell infiltration and extracellular matrix degradation. Currently, pharmacotherapy mainly aims to control risk factors without reversion of the dilated aorta. This study analyzed the immune-inflammatory response and identified the immune-related hub genes of AAA. METHOD: Gene Expression Omnibus datasets (GSE57691, GSE47472 and GSE7084) were downloaded. After identification of GSE57691 differentially expressed genes (DEGs), weighted gene co-expression network analysis of the DEGs was performed. Through enrichment analysis of each module and screening in Immunology Database and Analysis Portal, immune-related hub genes were identified via protein-protein interaction (PPI) network construction and lasso regression. CIBERSORT was utilized to analyze AAA immune infiltration. The correlations between the immune-related hub genes and infiltrating immune cells were investigated. Receiver operating characteristic (ROC) curve analysis was performed to determine immune-related hub gene cutoff values, which were validated in GSE47472 and GSE7084. RESULT: In GSE57691, 1,018 DEGs were identified. Five modules were identified in the co-expression network. The blue and green modules were found to be related to immune-inflammatory responses, and 61 immune-related genes were identified. PPI and lasso regression analyses identified FOS, IL-6 and IL2RB as AAA immune-related hub genes. CIBERSORT analysis indicated significantly increased infiltration of naive B cells, memory activated CD4 T cells, follicular helper T cells, monocytes and M1 macrophages and significantly decreased infiltration of M2 macrophages in AAA compared with normal samples. IL2RB was more strongly associated with immune infiltration in AAA than were FOS and IL6. The IL2RB area under the ROC curve (AUC) value was > 0.9 in both the training and validation set, demonstrating its strong, stable diagnostic value in AAA. CONCLUSION: AAA and normal samples had different immune infiltration statuses. IL2RB was identified as an immune-related hub gene and a potential hub gene with significant diagnostic value in AAA.


Subject(s)
Aortic Aneurysm, Abdominal , Gene Regulatory Networks , Interleukin-2 Receptor beta Subunit/genetics , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Computational Biology , Gene Expression Profiling , Humans , Receptors, Interleukin-2/genetics
9.
Cancer Sci ; 112(11): 4478-4489, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34545658

ABSTRACT

IL-2 is a pleiotropic cytokine that regulates immune cell homeostasis. Its immunomodulatory function has been used clinically as an active immunotherapy agent for metastatic cancers. However, severe adverse effects, including the vascular leak syndrome and the preferential stimulation of anti-immunogenic Treg rather than effector T cells, have been obstacles. We newly designed a mutein IL-2, Mutakine-6 (MK-6), with reduced IL-2Rα-binding capability. MK-6 induced comparable cell growth potential toward IL-2Rßγ-positive T cells but was far less efficient in in vitro Treg proliferation and STAT5 activation. Unlike IL-2, in vivo administration of MK-6 produced minimal adverse effects. Using CT26 and B16F10-syngeneic tumor models, we found MK-6 was highly efficacious on tumor regression. Serum albumin conjugation to MK-6 prolonged in vivo half-life and accumulated in CT26 tumors, showing enhanced antitumor effect. Tumor-infiltrating leukocytes analysis revealed that albumin-fused MK-6 increased the ratio of effector CD8+ T cells to CD4+ Treg cells. These results demonstrated that MK-6 is an efficient immunomodulator potentially used for improved immunotherapy with decreased adverse effects and attenuated Treg stimulation.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Immunologic Factors/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Disease Models, Animal , Female , Half-Life , Immunity, Cellular , Interleukin Receptor Common gamma Subunit/metabolism , Interleukin-2/adverse effects , Interleukin-2/metabolism , Interleukin-2/therapeutic use , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2 Receptor beta Subunit/metabolism , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phosphorylation , STAT5 Transcription Factor/metabolism , Serum Albumin/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/physiology , Tumor Suppressor Proteins/metabolism
10.
Front Immunol ; 12: 708874, 2021.
Article in English | MEDLINE | ID: mdl-34484208

ABSTRACT

Regulatory T cells (Treg) are essential to maintain immune homeostasis and prevent autoimmune disorders. While the function and molecular regulation of Foxp3+CD4+ Tregs are well established, much of CD8+ Treg biology remains to be revealed. Here, we will review the heterogenous subsets of CD8+ T cells have been named "CD8+ Treg" and mainly focus on CD122hiLy49+CD8+ Tregs present in naïve mice. CD122hiLy49+CD8+ Tregs, which depends on transcription factor Helios and homeostatic cytokine IL-15, have been established as a non-redundant regulator of germinal center (GC) reaction. Recently, we have demonstrated that TGF-ß (Transforming growth factor-ß) and transcription factor Eomes (Eomesodermin) are essential for the function and homeostasis of CD8+ Tregs. In addition, we will discuss several open questions regarding the differentiation, function and true identity of CD8+ Tregs as well as a brief comparison between two regulatory T cell subsets critical to control GC reaction, namely CD4+ TFR (follicular regulatory T cells) and CD8+ Tregs.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD8-Positive T-Lymphocytes/classification , Germinal Center/immunology , Humans , Interleukin-2 Receptor beta Subunit/analysis , Mice , NK Cell Lectin-Like Receptor Subfamily A/analysis , T-Lymphocytes, Regulatory/classification , Transforming Growth Factor beta/physiology
11.
J Clin Invest ; 131(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34375310

ABSTRACT

NKTR-255 is a PEG conjugate of recombinant human IL-15 (rhIL-15) being examined as a potential cancer immunotherapeutic. Since IL-15 responses can be mediated by trans or cis presentation via IL-15Rα or soluble IL-15/IL-15Rα complexes, we investigated the role of IL-15Rα in driving NKTR-255 responses using defined naive and memory OVA-specific CD8+ T cells (OT-I) and NK cells in mice. NKTR-255 induced a 2.5- and 2.0-fold expansion of CD8+ T and NK cells, respectively, in WT mice. In adoptive transfer studies, proliferation of naive and memory WT OT-I T cells in response to NKTR-255 was not impaired in IL-15Rα-/- mice, suggesting trans presentation was not utilized by NKTR-255. Interestingly, naive IL-15Rα-/- OT-I cells had deficient responses to NKTR-255, while memory IL-15Rα-/- OT-I cell responses were partially impaired, suggesting that naive CD8+ T cells are more dependent on cis presentation of NKTR-255 than memory CD8+ T cells. In bone marrow chimera studies, IL-15Rα-/- and WT NK cells present in WT recipients had similar responses to NKTR-255, suggesting that cis presentation is not utilized by NK cells. NKTR-255 could form soluble complexes with IL-15Rα; binding to murine IL-15Rα generated superagonists that preferentially stimulated NK cells, showing that conversion to IL-15Rß agonist biases the response toward NK cells. These findings highlight the ability of NKTR-255 to utilize IL-15Rα for cis presentation and act as an IL-15Rαß agonist on CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Interleukin-15/pharmacology , Killer Cells, Natural/drug effects , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Immunologic Memory , Interleukin-15/chemistry , Interleukin-15 Receptor alpha Subunit/physiology , Interleukin-2 Receptor beta Subunit/agonists , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Polyethylene Glycols/chemistry
12.
Front Immunol ; 12: 642856, 2021.
Article in English | MEDLINE | ID: mdl-34054809

ABSTRACT

Invariant NKT (iNKT) cells are thymus-generated innate-like T cells, comprised of three distinct subsets with divergent effector functions. The molecular mechanism that drives the lineage trifurcation of immature iNKT cells into the NKT1, NKT2, and NKT17 subsets remains a controversial issue that remains to be resolved. Because cytokine receptor signaling is necessary for iNKT cell generation, cytokines are proposed to contribute to iNKT subset differentiation also. However, the precise roles and requirements of cytokines in these processes are not fully understood. Here, we show that IL-2Rß, a nonredundant component of the IL-15 receptor complex, plays a critical role in both the development and differentiation of thymic iNKT cells. While the induction of IL-2Rß expression on postselection thymocytes is necessary to drive the generation of iNKT cells, surprisingly, premature IL-2Rß expression on immature iNKT cells was detrimental to their development. Moreover, while IL-2Rß is necessary for NKT1 generation, paradoxically, we found that the increased abundance of IL-2Rß suppressed NKT1 generation without affecting NKT2 and NKT17 cell differentiation. Thus, the timing and abundance of IL-2Rß expression control iNKT lineage fate and development, thereby establishing cytokine receptor expression as a critical regulator of thymic iNKT cell differentiation.


Subject(s)
Interleukin-2 Receptor beta Subunit/physiology , Natural Killer T-Cells/physiology , Thymus Gland/immunology , Animals , Cell Differentiation , Interleukin-15/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Natural Killer T-Cells/classification , Natural Killer T-Cells/cytology , STAT5 Transcription Factor/physiology
13.
Sci Rep ; 11(1): 10592, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34011961

ABSTRACT

The use of recombinant interleukin-2 (IL-2) as a therapeutic protein has been limited by significant toxicities despite its demonstrated ability to induce durable tumor-regression in cancer patients. The adverse events and limited efficacy of IL-2 treatment are due to the preferential binding of IL-2 to cells that express the high-affinity, trimeric receptor, IL-2Rαßγ such as endothelial cells and T-regulatory cells, respectively. Here, we describe a novel bispecific heavy-chain only antibody which binds to and activates signaling through the heterodimeric IL-2Rßγ receptor complex that is expressed on resting T-cells and NK cells. By avoiding binding to IL-2Rα, this molecule circumvents the preferential T-reg activation of native IL-2, while maintaining the robust stimulatory effects on T-cells and NK-cells in vitro. In vivo studies in both mice and cynomolgus monkeys confirm the molecule's in vivo biological activity, extended pharmacodynamics due to the Fc portion of the molecule, and enhanced safety profile. Together, these results demonstrate that the bispecific antibody is a safe and effective IL-2R agonist that harnesses the benefits of the IL-2 signaling pathway as a potential anti-cancer therapy.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Interleukin Receptor Common gamma Subunit/agonists , Interleukin-2 Receptor beta Subunit/agonists , Lymphocytes/drug effects , Animals , CHO Cells , Cricetulus , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Interleukin Receptor Common gamma Subunit/immunology , Interleukin-2 Receptor beta Subunit/immunology , Macaca fascicularis , Male , Mice, Inbred BALB C
14.
Front Immunol ; 12: 646159, 2021.
Article in English | MEDLINE | ID: mdl-33953717

ABSTRACT

IL-15 is one of the important biologics considered for vaccine adjuvant and treatment of cancer. However, a short half-life and poor bioavailability limit its therapeutic potential. Herein, we have structured IL-15 into a chimeric protein to improve its half-life enabling greater bioavailability for longer periods. We have covalently linked IL-15 with IgG2 base to make the IL-15 a stable chimeric protein, which also increased its serum half-life by 40 fold. The dimeric structure of this kind of IgG based biologics has greater stability, resistance to proteolytic cleavage, and less frequent dosing schedule with minimum dosage for achieving the desired response compared to that of their monomeric forms. The structured chimeric IL-15 naturally forms a dimer, and retains its affinity for binding to its receptor, IL-15Rß. Moreover, with the focused action of the structured chimeric IL-15, antigen-presenting cells (APC) would transpresent chimeric IL-15 along with antigen to the T cell, that will help the generation of quantitatively and qualitatively better antigen-specific memory T cells. In vitro and in vivo studies demonstrate the biological activity of chimeric IL-15 with respect to its ability to induce IL-15 signaling and modulating CD8+ T cell response in favor of memory generation. Thus, a longer half-life, dimeric nature, and anticipated focused transpresentation by APCs to the T cells will make chimeric IL-15 a super-agonist for memory CD8+ T cell responses.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Interleukin-15/immunology , Recombinant Fusion Proteins/immunology , Animals , CHO Cells , Cricetulus , Female , Genes, Immunoglobulin Heavy Chain , Humans , Immunoglobulin G/chemistry , Interleukin-15/chemistry , Interleukin-15/genetics , Interleukin-15/metabolism , Interleukin-2 Receptor beta Subunit/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Multimerization , Protein Stability
15.
Biomed Res Int ; 2021: 6624702, 2021.
Article in English | MEDLINE | ID: mdl-33954185

ABSTRACT

INTRODUCTION: Acute lung injury (ALI) induced by sepsis is a process related to inflammatory reactions, which involves lung cell apoptosis and production of inflammatory cytokine. Here, lipopolysaccharide (LPS) was applied to stimulate the mouse or human normal lung epithelial cell line (BEAS-2B) to construct a sepsis model in vivo and in vitro, and we also investigated the effect of miR-497-5p on sepsis-induced ALI. Material and Methods. Before LPS treatment, miR-497-5p antagomir was injected intravenously into mice to inhibit miR-497-5p expression in vivo. Similarly, miR-497-5p was knocked down in BEAS-2B cells. Luciferase reporter assay was applied to predict and confirm the miR-497-5p target gene. Cell viability, apoptosis, the levels of miR-497-5p, IL2RB, SP1, inflammatory cytokine, and lung injury were assessed. RESULTS: In BEAS-2B cells, a significant increase of apoptosis and inflammatory cytokine was shown after LPS stimulation. In septic mice, increased inflammatory cytokine production and apoptosis in lung cells and pulmonary morphological abnormalities were shown. The miR-497-5p inhibitor transfection showed antiapoptotic and anti-inflammatory effects on BEAS-2B cells upon LPS stimulation. In septic mice, the miR-497-5p antagomir injection also alleviated ALI, apoptosis, and inflammation caused by sepsis. The downregulation of IL2RB in BEAS-2B cells reversed the protective effects of the miR-497-5p inhibitor against ALI. CONCLUSION: In conclusion, downregulation of miR-497-5p reduced ALI caused by sepsis through targeting IL2RB, indicating the potential effect of miR-497-5p for improving ALI caused by sepsis.


Subject(s)
Acute Lung Injury/etiology , Down-Regulation/genetics , Interleukin-2 Receptor beta Subunit/metabolism , MicroRNAs/genetics , Sepsis/complications , Animals , Apoptosis , Base Sequence , Cell Line , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Promoter Regions, Genetic/genetics , Sp1 Transcription Factor/metabolism , Up-Regulation/genetics
16.
Nat Commun ; 12(1): 2715, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976157

ABSTRACT

Efficient immune responses rely on heterogeneity, which in CD8+ T cells, amongst other mechanisms, is achieved by asymmetric cell division (ACD). Here we find that ageing, known to negatively impact immune responses, impairs ACD in murine CD8+ T cells, and that this phenotype can be rescued by transient mTOR inhibition. Increased ACD rates in mitotic cells from aged mice restore the expansion and memory potential of their cellular progenies. Further characterization of the composition of CD8+ T cells reveals that virtual memory cells (TVM cells), which accumulate during ageing, have a unique proliferation and metabolic profile, and retain their ability to divide asymmetrically, which correlates with increased memory potential. The opposite is observed for naive CD8+ T cells from aged mice. Our data provide evidence on how ACD modulation contributes to long-term survival and function of T cells during ageing, offering new insights into how the immune system adapts to ageing.


Subject(s)
Aging/genetics , Asymmetric Cell Division/genetics , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/genetics , TOR Serine-Threonine Kinases/genetics , Aging/immunology , Animals , Asymmetric Cell Division/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation , Immunity, Innate , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymphocyte Activation , Mice , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/immunology
17.
Atherosclerosis ; 326: 1-10, 2021 06.
Article in English | MEDLINE | ID: mdl-33945906

ABSTRACT

BACKGROUND AND AIMS: The relationship between inflammation and lipid metabolism is complex and bidirectional. Lymphocyte-driven inflammation has been shown to modulate both atherosclerotic plaque development and cholesterol levels, but the mechanisms are incompletely understood. METHODS: The cardiometabolic effects of IL-2Rßγ signalling in atherosclerotic Apoe-/- mice were investigated by treatment with an agonistic IL-2Rßγ-targeting IL-2/anti-IL-2 complex or a monoclonal anti-CD122 (IL-2Rß) blocking antibody. RESULTS: Administration of IL-2Rßγ agonistic IL-2/anti-IL-2 complexes to Apoe-/- mice augmented opposing arms of the adaptive immune system. Expansion of effector/memory T cells and increased levels of circulating pro-inflammatory cytokines were observed along with elevated levels of regulatory T cells and IL-10. Notably, IL-2/anti-IL-2 treatment did not affect plaque size but decreased levels of plasma cholesterol. The cholesterol lowering effect of IL-2Rßγ agonism was not affected by anti-CD8 or anti-NK1.1 depleting antibody treatment but was contingent on the presence of adaptive immunity. Expression of multiple liver X receptor (LXR)-related genes, including Pltp and Srebp1c in the liver, was decreased by IL-2/anti-IL-2 treatment. Although IL-2Rßγ agonism lowered cholesterol levels, blocking IL-2Rßγ signalling using an anti-CD122 monoclonal antibody did not impact cholesterol levels or plaque burden in Apoe-/- mice. CONCLUSIONS: Elevated IL-2Rßγ signalling results in activation of both inflammatory and regulatory lymphocytes with a net zero effect on atherosclerosis and decreased plasma cholesterol levels. Changes in cholesterol levels were associated with reductions in hepatic LXR-related gene expression. Further studies are needed to investigate the clinical significance of IL-2 mediated modulation of hepatic LXR signalling in inflammatory disorders.


Subject(s)
Atherosclerosis , Interleukin-2 Receptor beta Subunit , Plaque, Atherosclerotic , Animals , Cholesterol , Inflammation , Lymphocytes , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE
18.
Science ; 372(6543)2021 05 14.
Article in English | MEDLINE | ID: mdl-33986151

ABSTRACT

Reactive oxygen species (ROS) increase in activated T cells because of metabolic activity induced to support T cell proliferation and differentiation. We show that these ROS trigger an oxidative stress response that leads to translation repression. This response is countered by Schlafen 2 (SLFN2), which directly binds transfer RNAs (tRNAs) to protect them from cleavage by the ribonuclease angiogenin. T cell-specific SLFN2 deficiency results in the accumulation of tRNA fragments, which inhibit translation and promote stress-granule formation. Interleukin-2 receptor ß (IL-2Rß) and IL-2Rγ fail to be translationally up-regulated after T cell receptor stimulation, rendering SLFN2-deficient T cells insensitive to interleukin-2's mitogenic effects. SLFN2 confers resistance against the ROS-mediated translation-inhibitory effects of oxidative stress normally induced by T cell activation, permitting the robust protein synthesis necessary for T cell expansion and immunity.


Subject(s)
Cell Cycle Proteins/metabolism , Immunity, Cellular , Oxidative Stress , RNA, Transfer/metabolism , T-Lymphocytes/immunology , Animals , Cell Cycle Proteins/genetics , Cell Proliferation , Female , Gene Deletion , Herpesviridae Infections/immunology , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/metabolism , Interleukin-2/metabolism , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/metabolism , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muromegalovirus , Protein Binding , Protein Biosynthesis , Reactive Oxygen Species/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/metabolism , Signal Transduction
19.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34006646

ABSTRACT

Colorectal cancer (CRC) represents the third most common malignancy and the second leading cause of cancer-related deaths worldwide. Although immunotherapy has taken center stage in mainstream oncology, it has shown limited clinical efficacy in CRC, generating an urgent need for discovery of new biomarkers and potential therapeutic targets. Galectin-1 (Gal-1), an endogenous glycan-binding protein, induces tolerogenic programs and contributes to tumor cell evasion of immune responses. Here, we investigated the relevance of Gal-1 in CRC and explored its modulatory activity within the CD8+ regulatory T cell (Treg) compartment. Mice lacking Gal-1 (Lgals1-/- ) developed a lower number of tumors and showed a decreased frequency of a particular population of CD8+CD122+PD-1+ Tregs in the azoxymethane-dextran sodium sulfate model of colitis-associated CRC. Moreover, silencing of tumor-derived Gal-1 in the syngeneic CT26 CRC model resulted in reduced number and attenuated immunosuppressive capacity of CD8+CD122+PD-1+ Tregs, leading to slower tumor growth. Moreover, stromal Gal-1 also influenced the fitness of CD8+ Tregs, highlighting the contribution of both tumor and stromal-derived Gal-1 to this immunoregulatory effect. Finally, bioinformatic analysis of a colorectal adenocarcinoma from The Cancer Genome Atlas dataset revealed a particular signature characterized by high CD8+ Treg score and elevated Gal-1 expression, which delineates poor prognosis in human CRC. Our findings identify CD8+CD122+PD-1+ Tregs as a target of the immunoregulatory activity of Gal-1, suggesting a potential immunotherapeutic strategy for the treatment of CRC.


Subject(s)
Adenocarcinoma/genetics , CD8-Positive T-Lymphocytes/immunology , Colitis/genetics , Colorectal Neoplasms/genetics , Galectin 1/genetics , T-Lymphocytes, Regulatory/immunology , Adenocarcinoma/immunology , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Animals , Atlases as Topic , Azoxymethane/administration & dosage , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Colitis/chemically induced , Colitis/immunology , Colitis/mortality , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Computational Biology , Dextran Sulfate/administration & dosage , Disease Models, Animal , Galectin 1/deficiency , Galectin 1/immunology , Gene Expression Regulation, Neoplastic , Humans , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/immunology , Mice , Mice, Knockout , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction , Survival Analysis , T-Lymphocytes, Regulatory/pathology , Tumor Burden
20.
BMC Immunol ; 22(1): 30, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33980160

ABSTRACT

BACKGROUND: Triggering receptor expressed on myeloid cells 2 (TREM2) signaling is considered to regulate anti-inflammatory responses in macrophages, dendritic cell maturation, osteoclast development, induction of obesity, and Alzheimer's disease pathogenesis. However, little is known regarding the effect of TREM2 on natural killer (NK) cells. RESULTS: Here, we demonstrated for the first time that CD3-CD122+NK1.1+ precursor NK (pNK) cells expressed TREM2 and their population increased in TREM2-overexpressing transgenic (TREM2-TG) mice compared with that in female C57BL/6 J wild type (WT) mice. Both NK cell-activating receptors and NK cell-associated genes were expressed at higher levels in various tissues of TREM2-TG mice than in WT mice. In addition, bone marrow-derived hematopoietic stem cells (HSCs) of TREM2-TG mice (TG-HSCs) successfully differentiated into NK cells in vitro, with a higher yield from TG-HSCs than from WT-HSCs. In contrast, TREM2 signaling inhibition by TREM2-Ig or a phosphatidylinositol 3-kinase (PI3K) inhibitor affected the expression of the NK cell receptor repertoire and decreased the expression levels of NK cell-associated genes, resulting in significant impairment of NK cell differentiation. Moreover, in melanoma-bearing WT mice, injection of bone marrow cells from TREM2-TG mice exerted greater antitumor effects than that with cells from WT control mice. CONCLUSIONS: Collectively, our data clearly showed that TREM2 promoted NK cell development and tumor regression, suggesting TREM2 as a new candidate for cancer immunotherapy.


Subject(s)
Bone Marrow Cells/immunology , Killer Cells, Natural/immunology , Melanoma/immunology , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Animals , Bone Marrow Transplantation , CD3 Complex/metabolism , Cell Differentiation , Female , Humans , Immunotherapy, Adoptive , Interleukin-2 Receptor beta Subunit/metabolism , Melanoma/therapy , Melanoma, Experimental , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental , Receptors, Immunologic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...