Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 101
1.
Clin Immunol ; 263: 110223, 2024 Jun.
Article En | MEDLINE | ID: mdl-38636890

Idiopathic severe aplastic anemia (SAA) is a disease of bone marrow failure caused by T-cell-induced destruction of hematopoietic stem and progenitor cells (HSPCs), however the mechanism remains unclear. We performed single-cell RNA sequencing of PBMCs and BMMCs from SAA patients and healthy donors and identified a CD8+ T cell subset with a tissue residency phenotype (Trm) in bone marrow that exhibit high IFN-γ and FasL expression and have a higher ability to induce apoptosis in HSPCs in vitro through FasL expression. CD8+ Trm cells were induced by IL-15 presented by IL-15Rα on monocytes, especially CD16+ monocytes, which were increased in SAA patients. CD16+ monocytes contributed to IL-15-induced CD38+CXCR6+ pre-Trm differentiation into CD8+ Trm cells, which can be inhibited by the CD38 inhibitor 78c. Our results demonstrate that IL-15-induced CD8+ Trm cells are pathogenic cells that mediate HSPC destruction in SAA patients and are therapeutic targets for future treatments.


Anemia, Aplastic , CD8-Positive T-Lymphocytes , GPI-Linked Proteins , Hematopoietic Stem Cells , Interleukin-15 , Monocytes , Receptors, IgG , Humans , Anemia, Aplastic/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Interleukin-15/pharmacology , Interleukin-15/immunology , Receptors, IgG/metabolism , Receptors, IgG/immunology , Monocytes/immunology , Monocytes/drug effects , Female , Male , Adult , Hematopoietic Stem Cells/immunology , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/immunology , Middle Aged , Fas Ligand Protein/metabolism , Fas Ligand Protein/immunology , Young Adult , Adolescent , Interferon-gamma/immunology , Interferon-gamma/metabolism , Receptors, Interleukin-15/metabolism , Receptors, Interleukin-15/immunology , Apoptosis/drug effects , Cell Differentiation/immunology
2.
J Autoimmun ; 145: 103217, 2024 May.
Article En | MEDLINE | ID: mdl-38581915

The autoimmunity-promoting cytokine, Interleukin-15 (IL-15), is often claimed to be a key pathogenic cytokine in alopecia areata (AA). Yet, rhIL-15 promotes human hair follicle (HF) growth ex vivo. We have asked whether the expression of IL-15 and its receptor (IL-15R) isoforms is altered in human AA and how IL-15 impacts on human HF immune privilege (HF-IP) in the presence/absence of interferon-γ (IFNγ), the well-documented key AA-pathogenic cytokine, as well as on hair regrowth after experimental AA induction in vivo. Quantitative immunohistomorphometry showed the number of perifollicular IL-15+ T cells in AA skin biopsies to be significantly increased compared to healthy control skin, while IL-15, IL-15Rα, and IL-15Rγ protein expression within the hair bulb were significantly down-regulated in AA HFs. In organ-cultured human scalp HFs, rhIL-15 significantly reduced hair bulb expression of MICA, the key "danger" signal in AA pathogenesis, and increased production of the HF-IP guardian, α-MSH. Crucially, ex vivo, rhIL-15 prevented IFNγ-induced HF-IP collapse, restored a collapsed HF-IP by IL-15Rα-dependent signaling (as documented by IL-15Rα-silencing), and protected AA-preventive immunoinhibitory iNKT10 cells from IFNγ-induced apoptosis. rhIL-15 even promoted hair regrowth after experimental AA induction in human scalp skin xenotransplants on SCID/beige mice in vivo. Our data introduce IL-15 as a novel, functionally important HF-IP guardian whose signaling is constitutively defective in scalp HFs of AA patients. Our data suggest that selective stimulation of intrafollicular IL-15Rα signaling could become a novel therapeutic approach in AA management, while blocking it pharmacologically may hinder both HF-IP restoration and hair re-growth and may thus make HFs more vulnerable to AA relapse.


Alopecia Areata , Hair Follicle , Immune Privilege , Interferon-gamma , Interleukin-15 , Interleukin-15/metabolism , Interleukin-15/immunology , Hair Follicle/immunology , Hair Follicle/metabolism , Humans , Animals , Alopecia Areata/immunology , Alopecia Areata/metabolism , Mice , Interferon-gamma/metabolism , Female , Receptors, Interleukin-15/metabolism , Receptors, Interleukin-15/immunology , Male , Adult , Middle Aged , Interleukin-15 Receptor alpha Subunit/metabolism , Interleukin-15 Receptor alpha Subunit/immunology , Skin/immunology , Skin/metabolism , Skin/pathology , Disease Models, Animal
3.
Biomed Pharmacother ; 168: 115740, 2023 Dec.
Article En | MEDLINE | ID: mdl-37865999

The efficacy of CAR-T cell therapy has been hindered by several factors that are intrinsic to the tumor microenvironment. Many strategies are being employed to overcome these barriers and improve immunotherapies efficacy. Interleukin (IL)- 4 is a cytokine released by tumor cells inside the tumor microenvironment and it can oppose T cell effector functions via engagement with the IL-4 receptor on the surface of T cells. To overcome IL-4-mediated immunosuppressive signals, we designed a novel inverted cytokine receptor (ICR). Our novel CAR construct (4/15NKG2D-CAR), consisted of an NKG2D-based chimeric antigen receptor, co-expressing IL-4R as an extracellular domain and IL-15R as a transmembrane and intracellular domain. In this way, IL-4R inhibitory signals were converted into IL-15R activation signals downstream. This strategy increased the efficacy of NKG2D-CAR-T cells in the pancreatic tumor microenvironment in vitro and in vivo. 4/15NKG2D-CAR-T cells exhibited increased activation, degranulation, cytokine release, and cytotoxic ability of NKG2D-CAR-T cells against IL-4+ pancreatic cell lines. Furthermore, 4/15NKG2D-CAR-T cells exhibited more expansion, less exhaustion, and an increased percentage of less differentiated T cell phenotypes in vitro when compared with NKG2D-CAR-T cells. That is why IL-4R/IL-15R-modified CAR-T cells eradicated more tumors in vivo and outperformed NKG2D-CAR-T cells. Thus, we report here a novel NKG2D-CAR-T cells that could overcome IL-4-mediated immunosuppression in solid tumors.


Cytokines , Receptors, Chimeric Antigen , Cell Line, Tumor , Cytokines/metabolism , Immunotherapy, Adoptive , Interleukin-15/metabolism , Interleukin-4/metabolism , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Receptors, Interleukin-15/metabolism , T-Lymphocytes , Tumor Microenvironment , Humans , HEK293 Cells
4.
Hepatology ; 77(5): 1486-1498, 2023 05 01.
Article En | MEDLINE | ID: mdl-36106384

BACKGROUND AND AIMS: Long-lasting immunological memory is the ultimate goal of vaccination. Homeostatic maintenance of memory CD8 + cytotoxic T cells (MemCD8TCs) is thought to be mediated by IL-15/IL-15R heterodimer (15HD)-expressing myeloid cells. Nonmyeloid hepatic stellate cells (HSCs) also express 15HD, but their role in maintaining MemCD8TC homeostasis is unknown. APPROACH AND RESULTS: We engineered a genetically engineered mouse in which IL-15R complementary DNA (cDNA) had been inserted in-frame with lecithin-retinol acyltransferase gene and bred onto an IL-15R-KO (15R-KO) genetic background (L15R) that expressed IL-15R in HSCs at normal levels, but not in other liver cells. Outside of the liver of L15R mice, IL-15R expression was found in a number of organs, but not in dendritic cells and macrophages. The low IL-15R expression in the bone marrow (BM) of L15R mice was eliminated by the reconstitution of lethally-irradiated L15R mice with 15R-KO BM to generate L15RC mice. Because MemCD8TC maintenance is mediated by 15HD, not empty IL-15R, 15HD content in L15R mice was determined and found for liver, lung, kidney, and heart. L15R and L15RC mice developed and maintained long-lasting, systemic antigen-specific MemCD8TCs that were efficacious against tumor growth and Listeria monocytogenes infection in an antigen-specific manner. Among the four organs with 15HD content, liver-associated MemCD8TCs were different from those found in the lung, kidney, and heart in two ways: (1) they were quantitatively the most numerous, and (2) they appeared uniquely in the form of clusters in a specialized structure, sinusoidal niches of the liver. CONCLUSIONS: The liver, the largest organ of the body, is endowed with the capability of effectuating long-lasting functional cytotoxic T cell memory.


CD8-Positive T-Lymphocytes , Hepatic Stellate Cells , Mice , Animals , Receptors, Interleukin-15/metabolism , Immunologic Memory , Liver , Mice, Inbred C57BL
5.
Cancer Cell ; 40(7): 720-737.e5, 2022 07 11.
Article En | MEDLINE | ID: mdl-35660135

Aerobic exercise is associated with decreased cancer incidence and cancer-associated mortality. However, little is known about the effects of exercise on pancreatic ductal adenocarcinoma (PDA), a disease for which current therapeutic options are limited. Herein, we show that aerobic exercise reduces PDA tumor growth, by modulating systemic and intra-tumoral immunity. Mechanistically, exercise promotes immune mobilization and accumulation of tumor-infiltrating IL15Rα+ CD8 T cells, which are responsible for the tumor-protective effects. In clinical samples, an exercise-dependent increase of intra-tumoral CD8 T cells is also observed. Underscoring the translational potential of the interleukin (IL)-15/IL-15Rα axis, IL-15 super-agonist (NIZ985) treatment attenuates tumor growth, prolongs survival, and enhances sensitivity to chemotherapy. Finally, exercise or NIZ985 both sensitize pancreatic tumors to αPD-1, with improved anti-tumor and survival benefits. Collectively, our findings highlight the therapeutic potential of an exercise-oncology axis and identify IL-15 activation as a promising treatment strategy for this deadly disease.


Antineoplastic Agents , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Receptors, Interleukin-15/metabolism , Antineoplastic Agents/pharmacology , CD8-Positive T-Lymphocytes , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Humans , Immunotherapy , Interleukin-15/metabolism , Interleukin-15/pharmacology , Interleukin-15/therapeutic use , Pancreatic Neoplasms/drug therapy , Tumor Microenvironment , Pancreatic Neoplasms
6.
J Immunol ; 208(2): 338-346, 2022 01 15.
Article En | MEDLINE | ID: mdl-34893528

IL-15 exhibits pleiotropic effects on NK and CD8+ T cells and contributes to host protection or immunopathology during infection. Although both type I IFNs and IFN-γ upregulate IL-15 expression, their effects on IL-15 upregulation and underlying mechanisms have not been compared comprehensively. In addition, little is known about trans-presentation of IL-15 by epithelial cells to lymphocytes. In this study, we analyzed the expression of IL-15 and IL-15Rα in the human hepatocyte-derived Huh-7 cell line after stimulation with IFN-α, IFN-ß, or IFN-γ using RT-PCR, flow cytometry, and confocal microscopy. We also performed knockdown experiments to investigate the signaling pathway involved in IL-15 upregulation. IFN-γ more potently upregulated IL-15 expression in Huh-7 cells than IFN-α and IFN-ß. Knockdown experiments revealed that IFN-γ- and IFN-ß-induced IL-15 expression relied on IFN regulatory factor 1 (IRF1), which is upregulated by STAT1 and IFN-stimulated gene factor 3, respectively. Inhibitor of κB kinase α/ß was also involved in IFN-γ-induced upregulation of IL-15. Furthermore, human NK cells were activated by coculture with IFN-γ-treated Huh-7 cells, which was abrogated by knocking down IL-15Rα in IFN-γ-treated Huh-7 cells, indicating that IFN-γ-induced IL-15 on Huh-7 cells activates NK cells via trans-presentation. In summary, our data demonstrate that IFN-γ potently elicits IL-15 trans-presentation by epithelial cells via IRF1. These data also suggest that the IFN-γ-IRF1-IL-15 axis may be a regulatory target for the treatment of diseases with IL-15 dysregulation.


CD8-Positive T-Lymphocytes/immunology , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/immunology , Interleukin-15/metabolism , Killer Cells, Natural/immunology , A549 Cells , Cell Line, Tumor , Epithelial Cells/metabolism , HeLa Cells , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-alpha/immunology , Interferon-beta/immunology , Lymphocyte Activation/immunology , Receptors, Interleukin-15/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction/physiology , Transcriptional Activation/genetics , Up-Regulation/genetics
7.
Front Immunol ; 12: 793918, 2021.
Article En | MEDLINE | ID: mdl-34956227

Interleukin-15, produced by hematopoietic and parenchymal cells, maintains immune cell homeostasis and facilitates activation of lymphoid and myeloid cell subsets. IL-15 interacts with the ligand-binding receptor chain IL-15Rα during biosynthesis, and the IL-15:IL-15Rα complex is trans-presented to responder cells that express the IL-2/15Rßγc complex to initiate signaling. IL-15-deficient and IL-15Rα-deficient mice display similar alterations in immune cell subsets. Thus, the trimeric IL-15Rαßγc complex is considered the functional IL-15 receptor. However, studies on the pathogenic role of IL-15 in inflammatory and autoimmune diseases indicate that IL-15 can signal independently of IL-15Rα via the IL-15Rßγc dimer. Here, we compared the ability of mice lacking IL-15 (no signaling) or IL-15Rα (partial/distinct signaling) to control Listeria monocytogenes infection. We show that IL-15-deficient mice succumb to infection whereas IL-15Rα-deficient mice clear the pathogen as efficiently as wildtype mice. IL-15-deficient macrophages did not show any defect in bacterial uptake or iNOS expression in vitro. In vivo, IL-15 deficiency impaired the accumulation of inflammatory monocytes in infected spleens without affecting chemokine and pro-inflammatory cytokine production. The inability of IL-15-deficient mice to clear L. monocytogenes results from impaired early IFNγ production, which was not affected in IL-15Rα-deficient mice. Administration of IFNγ partially enabled IL-15-deficient mice to control the infection. Bone marrow chimeras revealed that IL-15 needed for early bacterial control can originate from both hematopoietic and non-hematopoietic cells. Overall, our findings indicate that IL-15-dependent IL-15Rα-independent signaling via the IL-15Rßγc dimeric complex is necessary and sufficient for the induction of IFNγ from sources other than NK/NKT cells to control bacterial pathogens.


Interferon-gamma/metabolism , Interleukin-15/metabolism , Listeria monocytogenes/physiology , Listeriosis/immunology , Macrophages/immunology , Receptors, Interleukin-15/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Interferon-gamma/genetics , Interleukin-15/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phagocytosis , Receptors, Interleukin-15/genetics , Signal Transduction , Transplantation Chimera
8.
Mol Ther ; 29(10): 2949-2962, 2021 10 06.
Article En | MEDLINE | ID: mdl-34091051

Advances in immunostimulatory and anti-immunosuppressive therapeutics have revolutionized cancer treatment. However, novel immunotherapeutics with these dual functions are not frequently reported. Here we describe the creation of a heterodimeric bifunctional fusion molecule, HCW9218, constructed using our soluble tissue factor (TF)-based scaffold technology. This complex comprises extracellular domains of the human transforming growth factor-ß (TGF-ß) receptor II and a human interleukin-15 (IL-15)/IL-15 receptor α complex. HCW9218 can be readily expressed in CHO cells and purified using antibody-based affinity chromatography in a large-scale manufacturing setting. HCW9218 potently activates mouse natural killer (NK) cells and CD8+ T cells in vitro and in vivo to enhance cell proliferation, metabolism, and antitumor cytotoxic activities. Similarly, human immune cells become activated with increased cytotoxicity following incubation with HCW9218. This fusion complex also exhibits TGF-ß neutralizing activity in vitro and sequesters plasma TGF-ß in vivo. In a syngeneic B16F10 melanoma model, HCW9218 displayed strong antitumor activity mediated by NK cells and CD8+ T cells and increased their infiltration into tumors. Repeat-dose subcutaneous administration of HCW9218 was well tolerated by mice, with a half-life sufficient to provide long-lasting biological activity. Thus, HCW9218 may serve as a novel therapeutic to simultaneously provide immunostimulation and lessen immunosuppression associated with tumors.


CD8-Positive T-Lymphocytes/metabolism , Interleukin-15/genetics , Killer Cells, Natural/metabolism , Melanoma, Experimental/drug therapy , Receptor, Transforming Growth Factor-beta Type II/chemistry , Receptors, Interleukin-15/genetics , Recombinant Fusion Proteins/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Injections, Subcutaneous , Interleukin-15/metabolism , Melanoma, Experimental/immunology , Mice , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptors, Interleukin-15/metabolism , Recombinant Fusion Proteins/pharmacology , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
9.
J Immunother Cancer ; 9(5)2021 05.
Article En | MEDLINE | ID: mdl-34001523

BACKGROUND: NKTR-255 is a novel polyethylene glycol-conjugate of recombinant human interleukin-15 (rhIL-15), which was designed to retain all known receptor binding interactions of the IL-15 molecule. We explored the biologic and pharmacologic differences between endogenous IL-15 receptor α (IL-15Rα)-dependent (NKTR-255 and rhIL-15) and IL-15Rα-independent (precomplexed rhIL-15/IL-15Rα) cytokines. METHODS: In vitro pharmacological properties of rhIL-15, NKTR-255 and precomplex cytokines (rhIL-15/IL-15Rα and rhIL-15 N72D/IL-15Rα Fc) were investigated in receptor binding, signaling and cell function. In vivo pharmacokinetic (PK) and pharmacodynamic profile of the cytokines were evaluated in normal mice. Finally, immunomodulatory effect and antitumor activity were assessed in a Daudi lymphoma model. RESULTS: NKTR-255 and rhIL-15 exhibited similar in vitro properties in receptor affinity, signaling and leukocyte degranulation, which collectively differed from precomplexed cytokines. Notably, NKTR-255 and rhIL-15 stimulated greater granzyme B secretion in human peripheral blood mononuclear cells versus precomplexed cytokines. In vivo, NKTR-255 exhibited a PK profile with reduced clearance and a longer half-life relative to rhIL-15 and demonstrated prolonged IL-15R engagement in lymphocytes compared with only transient engagement observed for rhIL-15 and precomplexed rhIL-15 N72D/IL-15Rα Fc. As a consequent, NKTR-255 provided a durable and sustained proliferation and activation of natural killer (NK) and CD8+ T cells. Importantly, NKTR-255 is more effective than the precomplexed cytokine at inducing functionally competent, cytotoxic NK cells in the tumor microenvironment and the properties of NKTR-255 translated into superior antitumor activity in a B-cell lymphoma model versus the precomplexed cytokine. CONCLUSIONS: Our results show that the novel immunotherapeutic, NKTR-255, retains the full spectrum of IL-15 biology, but with improved PK properties, over rhIL-15. These findings support the ongoing phase 1 first-in-human trial (NCT04136756) of NKTR-255 in participants with relapsed or refractory hematologic malignancies, potentially advancing rhIL-15-based immunotherapies for the treatment of cancer.


Antineoplastic Agents/therapeutic use , Burkitt Lymphoma/drug therapy , Interleukin-15/therapeutic use , Lymphocytes/drug effects , Polyethylene Glycols/therapeutic use , Receptors, Interleukin-15/agonists , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Burkitt Lymphoma/pathology , Cell Degranulation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/metabolism , Female , HEK293 Cells , Humans , Interleukin-15/pharmacokinetics , Interleukin-15/pharmacology , Lymphocyte Activation/drug effects , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/pharmacology , Receptors, Interleukin-15/genetics , Receptors, Interleukin-15/metabolism , Signal Transduction , Tumor Microenvironment
10.
Dev Comp Immunol ; 121: 104103, 2021 08.
Article En | MEDLINE | ID: mdl-33857470

IL-35 plays a key role in regulatory T (Treg) and regulatory B (Breg) cell functions in mammals. CD25 has been demonstrated as one of the markers of Treg cells, and CD19+CD25hiCD71hi cells have been verified as a type of Breg cells in humans. These results indicate that there is a close relationship between IL-35 and CD25+ cells. In mammals, CD25 (alias IL-2Rα) has been identified as having high affinity and specificity for IL-2 binding, and is closely linked and structurally related to IL-15Rα, which having high affinity for IL-15 binding. In teleost, IL-15Rα can bind to both IL-2 and IL-15, with higher affinity to IL-15 than IL-2, and has been termed a CD25-like molecule in some research studies. To date, no studies of IL-35 and IL-15Rα have been documented in fish. In this work, five isoforms of IL-15Rα were cloned from grass carp, and a monoclonal antibody to the protein was developed. The results of flow cytometry and quantitative real-time PCR analyses demonstrated that grass carp IL-35 subunit genes EBI3a and IL-12p35 were mainly expressed in IL-15Rα+ cells, while the expression levels of IL-10 and TGF-ß in IL-15Rα+ and IL-15Rα- cells were insignificant. Recombinant grass carp IL-35 (rgcIL-35) could increase the proportion of IL-15Rα+ cells in leukocytes, and a certain proportion of IL-15Rα+ cells also appeared in myeloid cell subset II after stimulation with rgcIL-35. Meanwhile, the migration, phagocytic ability, and bactericidal ability of grass carp neutrophils were significantly decreased after stimulation with certain concentrations of rgcIL-35. Moreover, neutrophil apoptosis could be significantly inhibited by rgcIL-35.


Carps/immunology , Fish Proteins/metabolism , Interleukin-12 Subunit p35/metabolism , Neutrophils/immunology , Receptors, Interleukin-15/metabolism , Animals , Apoptosis/immunology , Carps/genetics , Cells, Cultured , Fish Proteins/genetics , Fish Proteins/isolation & purification , Head Kidney/cytology , Head Kidney/immunology , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p35/isolation & purification , Neutrophils/metabolism , Phagocytosis , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
11.
Biochem Biophys Res Commun ; 556: 53-58, 2021 06 04.
Article En | MEDLINE | ID: mdl-33839414

Interleukin 15 receptor (IL-15R) is a transmembrane signalling protein consisting of 3 subsets: α, ß (IL-15Rß), and γ (γc). IL-2 and IL-15 share the signalling domains IL-15Rß and γc, although they bind to intrinsic α-subsets and non-signalling domains. Additionally, IL-2 and IL-15 play different roles; therefore, there have been many observations of the dynamic behaviours of IL-15R, which are linked to physiological functions. For more practical discrimination between IL-2 and IL-15, a study was designed and carried out in which α-subsets were removed and a cytoplasmic inhibitor was applied to create a simplified environment in which secondary signalling molecules were reduced. We also applied a new measurement method, diffracted X-ray blinking (DXB), to achieve higher accuracy (<0.01 Å). The dynamics of IL-2 binding (confined motion, max range = 0.71 Å) and IL-15 binding (normal motion) in live natural killer cells were different. We also confirmed. that DXB was a suitable method to quantitatively evaluate the transmembrane protein dynamics of inner/outer live cell membranes by labeling the extracellular domain since the measurements were dependent on the cytosolic environment.


Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Receptors, Interleukin-15/analysis , Receptors, Interleukin-15/metabolism , X-Ray Diffraction/methods , Cell Survival , Diffusion , Humans , Hydroxybenzoates , Interleukin-15/metabolism , Interleukin-2/metabolism , Molecular Dynamics Simulation , Nitrofurans , Protein Domains , Substrate Specificity
13.
Front Immunol ; 12: 623280, 2021.
Article En | MEDLINE | ID: mdl-33732245

Expression of tissue-restricted antigens (TRAs) in thymic epithelial cells (TECs) ensures negative selection of highly self-reactive T cells to establish central tolerance. Whether some of these TRAs could exert their canonical biological functions to shape thymic environment to regulate T cell development is unclear. Analyses of publicly available databases have revealed expression of transcripts at various levels of many cytokines and cytokine receptors such as IL-15, IL-15Rα, IL-13, and IL-23a in both human and mouse TECs. Ablation of either IL-15 or IL-15Rα in TECs selectively impairs type 1 innate like T cell, such as iNKT1 and γδT1 cell, development in the thymus, indicating that TECs not only serve as an important source of IL-15 but also trans-present IL-15 to ensure type 1 innate like T cell development. Because type 1 innate like T cells are proinflammatory, our data suggest the possibility that TEC may intrinsically control thymic inflammatory innate like T cells to influence thymic environment.


Epithelial Cells/metabolism , Immunity, Innate , Interleukin-15/metabolism , Receptors, Interleukin-15/metabolism , T-Lymphocyte Subsets/metabolism , Thymocytes/metabolism , Thymus Gland/metabolism , Animals , Cell Communication , Cellular Microenvironment , Databases, Genetic , Epithelial Cells/immunology , Humans , Interleukin-15/genetics , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Mice , Mice, Knockout , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Receptors, Interleukin-15/genetics , Signal Transduction , T-Lymphocyte Subsets/immunology , Thymocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology
14.
Front Immunol ; 11: 1385, 2020.
Article En | MEDLINE | ID: mdl-32793194

Objective: Interleukin-15 (IL-15) is a pro-inflammatory cytokine that is increased in joint fluids of early-stage osteoarthritis (OA) patients, and has been associated with expression of proteases that can damage cartilage, and the development of neuropathic pain-like symptoms (NP) after nerve injury. The objective of this study was to further explore the role of IL-15 in the pathogenesis of OA cartilage degeneration and test genetic variation in the IL-15 receptor α gene (IL15RA) for an association with OA with radiographic severity and symptoms. Methods: Cartilage samples from donors (n = 10) were analyzed for expression of the IL15 receptor α-chain using immunohistochemistry, and for responses to IL-15 in vitro using explant cultures. Data from two independent Nottinghamshire-based studies (n = 795 and n = 613) were used to test genetic variants in the IL15RA gene (rs2228059 and rs7097780) for an association with radiographic severity, symptomatic vs. asymptomatic OA and NP. Results: IL-15Rα was expressed in chondrocytes from cartilage obtained from normal and degenerative knees. IL-15 significantly increased the release of matrix metalloproteinase-1 and -3 (MMP-1 and -3), but did not affect loss of proteoglycan from the articular matrix. Genetic variants in the IL15RA gene are associated with risk of symptomatic vs. asymptomatic OA (rs7097780 OR = 1.48 95% 1.10-1.98 p < 0.01) and with the risk of NP post-total joint replacement (rs2228059 OR = 0.76 95% 0.63-0.92 p < 0.01) but not with radiographic severity. Conclusions: In two different cohorts of patients, we show an association between genetic variation at the IL15 receptor and pain. Although ex vivo cartilage explants could respond to IL-15 with increased protease production, we found no effect of IL-15 on cartilage matrix loss and no association between IL15RA variants and radiographic severity. Together, these results suggest that IL-15 signaling may be a target for pain, but may not impact structural progression, in OA.


Interleukin-15/metabolism , Osteoarthritis , Peptide Hydrolases/metabolism , Receptors, Interleukin-15/genetics , Adolescent , Adult , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Female , Humans , Male , Middle Aged , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Pain/etiology , Polymorphism, Single Nucleotide , Receptors, Interleukin-15/metabolism , Young Adult
15.
Cell Oncol (Dordr) ; 43(6): 1085-1097, 2020 Dec.
Article En | MEDLINE | ID: mdl-32767257

PURPOSE: Epstein-Barr virus associated gastric cancer (EBVaGC) often exhibits a favorable prognosis that correlates with highly methylated viral and host genes and significant immune cell infiltration compared to EBV-negative gastric cancers (GCs). Previously, it has been reported that expression of the IL-15 receptor α (IL-15Rα) is down-regulated in EBVaGC via promoter hypermethylation. In the present study, we offer a novel explanation for this puzzle by associating IL-15Rα expression with infiltration of lymphocytes in GC lesions. METHODS: We investigated the expression of IL-15Rα by RT-PCR, Western-blotting and immunohistochemistry in GC cell lines and primary tissues, respectively. IL-15Rα promoter methylation was analyzed using genomic methylation sequencing. The growth behavior of GC cells was analyzed using MTT, flow cytometry, colony formation, transwell invasion and scratch wound healing assays. Demethylation of IL-15Rα was carried out using 5-Aza-CdR, and rIL-15 was added to evaluate growth promoting effects of the IL-15/IL-15Rα complex. Human peripheral blood mononuclear cells (PBMCs) were co-cultured with GC cells with/without the addition of rIL-15, after which the phosphorylation of STAT5 in PBMCs was evaluated using flow cytometry to estimate the activation of these immune cells through IL-15 binding to IL-2Rß/γ receptors by in trans presentation. RESULTS: We found that EBV-positive GC cells (AE) expressed IL-15Rα at a significantly lower level than EBV-negative GC cells (AGS) due to promoter hypermethylation. In the absence of immune cells, IL-15Rα on the cancer cell surface induced a malignant phenotype, including augmented cell growth, migration and invasion, and decreased apoptosis. 5-Aza-CdR reverted AE cells to a more malignant phenotype similar to AGS cells, which may be attributed to activation of the STAT1, STAT3 and ERK1/2 pathways. However, when PBMCs were added to the GC cell cultures, these immune cells were activated as detected by increased pSTAT5 levels. Also, more GC cells underwent apoptosis. These effects were enhanced by the addition of rIL-15 and, subsequently, confirmed in EBVaGC patient samples exhibiting increased expression of T cell surface markers and activation of immune co-stimulating pathways. CONCLUSIONS: Our findings suggest a mechanistic explanation for the clinical association of EBVaGC with a lower IL-15Rα expression, a better prognosis and an increased lymphocyte infiltration. We propose that in highly infiltrated GCs the IL-15/IL-15Rα complex on the GC cell surface may present IL-15 in trans to IL-2Rß/γ-expressing immune cells to activate these cells in the tumor microenvironment.


Disease Progression , Epigenesis, Genetic , Herpesvirus 4, Human/physiology , Receptors, Interleukin-15/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Azacitidine/pharmacology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Methylation/drug effects , DNA Methylation/genetics , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Herpesvirus 4, Human/drug effects , Humans , Models, Biological , Neoplasm Invasiveness , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Stomach Neoplasms/pathology , Survival Analysis , Up-Regulation/drug effects , Up-Regulation/genetics
16.
Cell Rep ; 31(9): 107720, 2020 06 02.
Article En | MEDLINE | ID: mdl-32492428

Natural killer (NK) cells are cytotoxic innate lymphoid cells (ILCs) that mediate antiviral and antitumor responses and require the transcriptional regulator Eomesodermin (Eomes) for early development. However, the role of Eomes and its molecular program in mature NK cell biology is unclear. To address this, we develop a tamoxifen-inducible, type-1-ILC-specific (Ncr1-targeted) cre mouse and combine this with Eomes-floxed mice. Eomes deletion after normal NK cell ontogeny results in a rapid loss of NK cells (but not ILC1s), with a particularly profound effect on penultimately mature stage III NK cells. Mechanisms responsible for stage III reduction include increased apoptosis and impaired maturation from stage II precursors. Induced Eomes deletion also decreases NK cell cytotoxicity and abrogates in vivo rejection of major histocompatibility complex (MHC)-class-I-deficient cells. However, other NK cell functional responses, and stage IV NK cells, are largely preserved. These data indicate that mature NK cells have distinct Eomes-dependent and -independent stages.


Killer Cells, Natural/immunology , T-Box Domain Proteins/metabolism , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Apoptosis , Cell Cycle Checkpoints , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , Receptors, Interleukin-15/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Spleen/cytology , Spleen/immunology , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics
17.
Respir Res ; 21(1): 33, 2020 Jan 29.
Article En | MEDLINE | ID: mdl-31996218

BACKGROUND: Interleukin 15 (IL-15) is a growth and modulating factor for B, T lymphocytes and natural killer cells (NK). Its action on innate and adaptive immunity is modulated by its alpha chain receptor (IL-15Rα). The IL-15/sIL-15Rα complex (IL-15Cx) increases the bioavailability and activity of the cytokine in vivo. IL-15Cx has been used in diseases to dampen IL-15 inflammation by the use of soluble IL-15Ralpha specificity. Here, we aim to evaluate the interest of IL-15Cx in a mouse model of asthma. METHODS: Using a mouse model of asthma consisting in percutaneous sensitization and intranasal challenge with total house dust mite extract, we evaluated the effect of IL-15Cx injected intraperitoneally four times after a first nasal challenge. Respiratory function was assessed by the technique of forced oscillations (Flexivent®). The effect on bronchial remodeling was evaluated by lung histology. The inflammatory status was analyzed by flow cytometry. RESULTS: We observed that the IL-15Cx modulates lung and systemic inflammation by increasing NK cells, CD8+ memory T cells and regulatory cells. However, IL-15Cx displays no effect on bronchial hyperreactivity, bronchial remodeling nor cellular bronchial infiltrate, but limits the secretion of bronchial mucus and modulates only inflammatory response in a HDM-allergic asthma murine model. CONCLUSIONS: IL-15Cx has a limited effect on immune response in asthma and has no effect on lung function in mice. Thus, it limits its therapeutic potential but might suggest a combinatory potential with other therapeutics.


Adaptive Immunity/immunology , Asthma/immunology , Immunity, Cellular/immunology , Interleukin-15/immunology , Receptors, Interleukin-15/immunology , Adaptive Immunity/drug effects , Allergens/immunology , Allergens/toxicity , Animals , Asthma/chemically induced , Asthma/metabolism , Female , Humans , Immunity, Cellular/drug effects , Interleukin-15/metabolism , Mice , Mice, Inbred BALB C , Receptors, Interleukin-15/metabolism
18.
Immunology ; 159(2): 221-230, 2020 02.
Article En | MEDLINE | ID: mdl-31663113

Oxidized low-density lipoprotein (LDL) contributes to cardiovascular disease in part by mediating activation and maturation of monocytes and macrophages. Furthermore, co-localization studies using histochemical approaches have implicated a potential role for oxidized LDL as a mediator of interleukin-15 (IL-15) expression in myeloid cells of atherosclerotic plaque. The latter activity could be an important pro-inflammatory mechanism that mediates myeloid cell/T-cell crosstalk. Here, we examined the responses of primary human monocytes to highly oxidized LDL molecules. Oxidized LDL readily induced secretion of chemokines MCP-1 (CCL2) and GRO-α (CXCL1) but unlike lipopolysaccharide (LPS), has limited capacity to induce a variety of other cytokines including tumor necrosis factor-α, IL-6, IL-1ß and interferon-γ-induced protein-10 and also displayed a poor capacity to induce p-Akt or P-S6 signaling. Failure of oxidized LDL to induce IL-1ß secretion was associated with limited induction of caspase-1 activation. Furthermore, despite finding evidence that oxidized LDL could enhance the expression of IL-15 and IL-15 receptor expression in monocytes, we found no evidence that it could confer IL-15 transpresentation capability to these cells. This observation contrasted with induction of IL-15 transpresentation in lipopolysaccharide-stimulated monocytes. Overall, our data suggest that highly oxidized LDL is a selective inducer of monocyte activation. Sterile inflammatory mediators, particularly those implicated in Toll-like receptor 4 signaling, may play a role in vascular pathology but the activities of these agents are not uniform.


Interleukin-15/metabolism , Interleukin-1beta/metabolism , Lipoproteins, LDL/pharmacology , Monocytes/drug effects , Caspase 1/metabolism , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-15/immunology , Interleukin-1beta/immunology , Lipopolysaccharides/pharmacology , Monocytes/immunology , Monocytes/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin-15/immunology , Receptors, Interleukin-15/metabolism , Ribosomal Protein S6 Kinases/metabolism , Secretory Pathway , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
19.
Immunol Lett ; 217: 65-71, 2020 01.
Article En | MEDLINE | ID: mdl-31738956

Highly conserved 2D receptor clusters (membrane rafts) of immunological signaling molecules with MHCI and MHCII antigens as their cores have been observed in the past on the surface of T- and B-cell lines of lymphoid origin, as well as on cells from patients with colon tumor and Crohn's disease. Conservativity is related to the ever presence of MHCI molecules. Although they are suspected to play a role in maintaining these clusters and facilitating transmembrane signaling, their exact role has been left largely enigmatic. Here we are suggesting stochastic resonance (SR), or "noise-assisted signal detection", as a general organizing principle for transmembrane signaling events evoked by processes like immune recognition and cytokine binding taking place in these clusters. In the conceptual framework of SR, in immune recognition as a prototype of transmembrane signaling, the sea of self-peptide-MHC complexes around a nonself-peptide presenting MHC is conceived as a source of quickly fluctuating unspecific signal ("athermal noise") serving the extra energy for amplifying the weak sub-threshold specific signal of the nonself-peptide presenting MHC. This same noise is also utilized for a readjustment of the threshold - and also the sensitivity and specificity - of detection by a closed loop feedback control of the TcR-CD8 (CD4) proximity on the detecting T-cell. The weak sub threshold specific signal of nonself-peptide presenting MHC is amplified by the superposing unspecific signals of the neighboring self peptide-MHC complexes towards the T-cell receptor as the detector. Because in a successful detection event both self- and nonself-peptides are detected simultaneously, the principle of coincidence (or lock-in) detection is also realized. The ever presence of MHC islands gets a natural explanation as a source of extra power - in a form of "athermal noise" - needed for coincidence detection and frequency encoding the evoked downstream signals. The effect is quite general, because the actual type of molecules surrounding a chief signaling molecule - like nonself-peptide holding MHC, interleukin-2 and -15 cytokine receptors (IL-2R/15R) - as the fluctuating interaction energy sources is immaterial. The model applies also for other types of signaling, such as those evoked by cytokine binding. The phenomenon of SR can also be interpreted as sampling of a low frequency, specific signal with a high frequency unspecific signal, the "noise". Recipes for identifying other forms of SR in membrane clusters with biophysical tools are recommended.


Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class I/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Adaptive Immunity , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Fluorescence Polarization , Fluorescence Resonance Energy Transfer , Humans , Immune Tolerance , Patch-Clamp Techniques , Receptors, Interleukin-15/metabolism , Receptors, Interleukin-2/metabolism , Stochastic Processes
20.
J Immunother Cancer ; 7(1): 355, 2019 12 19.
Article En | MEDLINE | ID: mdl-31856922

Cytokines of the common γ-chain receptor family such as IL-15 are vital with respect to activating immune cells, sustaining healthy immune functions, and augmenting the anti-tumor activity of effector cells, making them ideal candidates for cancer immunotherapy. IL-15, either in its soluble form (IL-15sol) or complexed with IL-15Rα (IL-15Rc), has been shown to exhibit potent anti-tumor activities in various experimental cancer studies. Here we describe the impact of intraperitoneal IL-15 in a cancer cell-delivered IL-15 immunotherapy approach using the 70Z/3-L leukemia mouse model. Whereas both forms of IL-15 led to significantly improved survival rates compared to the parent cell line, there were striking differences in the extent of the improved survival: mice receiving cancer cells secreting IL-15sol showed significantly longer survival and protective long-term immunity compared to those producing IL-15Rc. Interestingly, injection of leukemia cells secreting IL-15sol lead to heightened expansion of CD4+ and CD8+ T-cell populations in the peritoneum compared to IL-15Rc. Cell-secreted IL-15Rc resulted in an influx and/or expansion of NK1.1+ cells in the peritoneum which was much less pronounced in the IL-15sol model. Furthermore, IL-15Rc but not IL-15sol lead to T-cell exhaustion and disease progression. To our knowledge, this is the first study detailing a significantly different biological effect of cell-delivered IL-15sol versus IL-15Rc in a mouse cancer immunotherapy study.


Immunomodulation , Immunotherapy , Interleukin-15/metabolism , Leukemia/etiology , Leukemia/metabolism , Receptors, Interleukin-15/metabolism , Animals , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Interleukin-15/blood , Interleukin-15/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia/pathology , Leukemia/therapy , Melanoma, Experimental , Mice , Protein Binding , Receptors, Interleukin-15/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transduction, Genetic , Treatment Outcome , Xenograft Model Antitumor Assays
...