Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Reprod Biol Endocrinol ; 19(1): 114, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34289871

ABSTRACT

BACKGROUND: The immune mechanism was shown to be involved in the development of adenomyosis. The aim of the current study was to evaluate the expression of the immune checkpoints B7-H2, B7-H3, B7-H4 and PD-L2 in adenomyosis and to explore the effect of mifepristone on the expression of these immune checkpoints. METHODS: The expression of B7-H2, B7-H3, B7-H4 and PD-L2 in normal endometria and adenomyosis patient samples treated with or without mifepristone was determined by immunohistochemistry analysis. RESULTS: In adenomyosis patient samples, the expression of B7-H2, B7-H3 and B7-H4 was increased in the eutopic and ectopic endometria compared with normal endometria, both in the proliferative and secretory phases. Moreover, the expression of B7-H2 and B7-H3 was higher in adenomyotic lesions than in the corresponding eutopic endometria, both in the proliferative and secretory phases. The expression of PD-L2 was higher in adenomyotic lesions than in normal endometria in both the proliferative and secretory phases. In the secretory phase but not the proliferative phase, the expression of B7-H4 and PD-L2 in adenomyotic lesions was significantly higher than that in the corresponding eutopic endometria. In normal endometria and eutopic endometria, the expression of B7-H4 was elevated in the proliferative phase compared with that in the secretory phase, while in the ectopic endometria, B7-H4 expression was decreased in the proliferative phase compared with the secretory phase. In addition, the expression of B7-H2, B7-H3, B7-H4 and PD-L2 was significantly decreased in adenomyosis tissues after treatment with mifepristone. CONCLUSIONS: The expression of the immune checkpoint proteins B7-H2, B7-H3, B7-H4 and PD-L2 is upregulated in adenomyosis tissues and is downregulated with mifepristone treatment. The data suggest that B7 immunomodulatory molecules are involved in the pathophysiology of adenomyosis.


Subject(s)
Adenomyosis/metabolism , B7 Antigens/biosynthesis , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Mifepristone/therapeutic use , Programmed Cell Death 1 Ligand 2 Protein/biosynthesis , V-Set Domain-Containing T-Cell Activation Inhibitor 1/biosynthesis , Adenomyosis/drug therapy , Adenomyosis/genetics , Adult , B7 Antigens/antagonists & inhibitors , B7 Antigens/genetics , Endometrium/drug effects , Endometrium/metabolism , Female , Gene Expression , Hormone Antagonists/pharmacology , Hormone Antagonists/therapeutic use , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Ligand/genetics , Middle Aged , Mifepristone/pharmacology , Programmed Cell Death 1 Ligand 2 Protein/antagonists & inhibitors , Programmed Cell Death 1 Ligand 2 Protein/genetics , V-Set Domain-Containing T-Cell Activation Inhibitor 1/antagonists & inhibitors , V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics
2.
Allergy ; 74(4): 650-662, 2019 04.
Article in English | MEDLINE | ID: mdl-30220084

ABSTRACT

BACKGROUND: Allergic asthma is characterized by chronic inflammation and remodelling of the airways, associated with dysregulated type 2 immune responses and allergen-specific IgE. T follicular helper cells (TFH ) are crucial in T-dependent B-cell responses and have been implicated in allergic airway disease (AAD). TFH , unlike other CD4+ T cells, are uniquely reliant on continuous ICOS signalling to maintain their phenotype after T-cell priming; therefore, disrupting this signal can impair TFH responses. However, the contribution of TFH to disease during chronic aero-allergen exposure and the therapeutic potential of targeting these cells have not been evaluated. METHODS: To establish AAD, female BALB/c mice were repeatedly exposed to house dust mite or Alternaria alternata three times a week for up to 5 weeks. To examine the impact of TFH on AAD, mice were allergen exposed for 5 weeks and co-administered anti-ICOS Ligand-targeted antibodies, three times a week for the last 2 weeks. RESULTS: TFH were first observed in the lung-draining lymph nodes and with further exposure were also found locally within the lungs. TFH accumulated with sustained allergen exposure, alongside germinal centre (GC) B cells. Blockade of ICOS signalling after AAD establishment successfully depleted TFH but did not affect the differentiation of other CD4+ T-cell subsets. This reduced GC responses, allergen-specific IgE, inflammation, pulmonary IL-13 and airway hyper-responsiveness. CONCLUSIONS: TFH are crucial in the regulation of AAD and the ICOS/ICOS-L pathway could represent a novel therapeutic target in allergic asthma.


Subject(s)
Asthma/drug therapy , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Protein/antagonists & inhibitors , T-Lymphocytes, Helper-Inducer/immunology , Animals , Asthma/pathology , B-Lymphocytes/immunology , Female , Germinal Center/immunology , Germinal Center/pathology , Inducible T-Cell Co-Stimulator Ligand/metabolism , Inducible T-Cell Co-Stimulator Protein/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Pyroglyphidae/immunology
3.
J Immunol ; 195(3): 944-52, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26109645

ABSTRACT

Regulatory T cells (Tregs), a subset of CD4(+) T cells, dramatically accumulate with age in humans and mice and contribute to age-related immune suppression. Recently, we showed that a majority of accumulating Tregs in aged mice expressed low levels of CD25, and their accrual is associated with declining levels of IL-2 in aged mice. In this study, we further investigated the origin of CD25(lo) Tregs in aged mice. First, aged Tregs had high expression of neuropilin-1 and Helios, and had a broad Vß repertoire. Next, we analyzed the gene expression profile of Tregs, naive T cells, and memory T cells in aged mice. We found that the gene expression profile of aged CD25(lo) Tregs were more related to young CD25(lo) Tregs than to either naive or memory T cells. Further, the gene expression profile of aged Tregs was consistent with recently described "effector" Tregs (eTregs). Additional analysis revealed that nearly all Tregs in aged mice were of an effector phenotype (CD44(hi)CD62L(lo)) and could be further characterized by high levels of ICOS and CD69. ICOS contributed to Treg maintenance in aged mice, because in vivo Ab blockade of ICOSL led to a loss of eTregs, and this loss was rescued in Bim-deficient mice. Further, serum levels of IL-6 increased with age and contributed to elevated expression of ICOS on aged Tregs. Finally, Treg accrual was significantly blunted in aged IL-6-deficient mice. Together, our data show a role for IL-6 in promoting eTreg accrual with age likely through maintenance of ICOS expression.


Subject(s)
Aging/immunology , Inducible T-Cell Co-Stimulator Protein/immunology , Interleukin-6/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Apoptosis Regulatory Proteins/genetics , Base Sequence , Bcl-2-Like Protein 11 , Cell Death , Cell Survival , DNA-Binding Proteins/biosynthesis , Gene Expression Profiling , Hyaluronan Receptors/biosynthesis , Immunologic Memory/genetics , Immunologic Memory/immunology , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-6/blood , Interleukin-6/genetics , L-Selectin/biosynthesis , Lectins, C-Type/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropilin-1/biosynthesis , Proto-Oncogene Proteins/genetics , Sequence Analysis, DNA , Transcription Factors/biosynthesis
4.
Am J Pathol ; 182(6): 2204-13, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23578385

ABSTRACT

In a successful pregnancy, the semiallogeneic fetus is not rejected by the maternal immune system, which implies tolerance mechanisms protecting fetal tissues from maternal immune attack. Here we report that the ICOS-B7h costimulatory pathway plays a critical role in maintaining the equilibrium at the fetomaternal interface. Blockade of this pathway increased fetal resorption and decreased fetal survival in an allogeneic pregnancy model (CBA female × B6 male). Locally in the placenta, levels of regulatory markers such as IDO and TGF-ß1 were reduced after anti-B7h monoclonal antibody treatment, whereas levels of effector cytokines (eg, IFN-γ) were significantly increased. In secondary lymphoid organs, enhanced IFN-γ and granzyme B production (predominantly by CD8(+) T cells) was observed in the anti-B7h-treated group. The deleterious effect of B7h blockade in pregnancy was maintained only in CD4 knockout mice, not in CD8 knockout mice, which suggests a role for CD8(+) T cells in immune regulation by the ICOS-B7h pathway. In accord, regulatory CD8(+) T cells (in particular, CD8(+)CD103(+) cells) were significantly decreased after anti-B7h monoclonal antibody treatment, and adoptive transfer of this subset abrogated the deleterious effect of B7h blockade in fetomaternal tolerance. Taken together, these data support the hypothesis that B7h blockade abrogates tolerance at the fetomaternal interface by enhancing CD8(+) effector response and reducing local immunomodulation mediated by CD8(+) regulatory T cells.


Subject(s)
Immune Tolerance/immunology , Inducible T-Cell Co-Stimulator Ligand/immunology , Maternal-Fetal Exchange/immunology , Placenta/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Cytokines/biosynthesis , Embryo Loss/immunology , Female , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Litter Size/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Pregnancy , Spleen/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation
5.
Clin Immunol ; 148(3): 369-75, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23680362

ABSTRACT

Co-stimulatory molecules help to regulate interactions between T cells and antigen-presenting cells and may play an important role in the pathogenesis of lupus. Both work in murine models and some early studies in human lupus support further examination of these molecules as therapeutic targets. Complexities of lupus clinical trial variables may have hampered progress in this area but recent developments in the field may make interventional trials more feasible in the near future. To date biologics which provide direct blockade of interactions between CD40 and CD154, B7RP-1 and ICOS, and CD80 or CD86 with CD28 have been assessed in multicenter clinical trials. These data will be reviewed and critiqued.


Subject(s)
B7-1 Antigen/antagonists & inhibitors , B7-2 Antigen/antagonists & inhibitors , CD40 Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Protein/antagonists & inhibitors , Lupus Erythematosus, Systemic/drug therapy , Animals , Clinical Trials as Topic , Humans
6.
Clin Pharmacol Ther ; 114(2): 371-380, 2023 08.
Article in English | MEDLINE | ID: mdl-37150935

ABSTRACT

Rozibafusp alfa (AMG 570) is a first-in-class bispecific IgG2-peptide fusion designed to inhibit inducible T-cell costimulator ligand (ICOSL) and B-cell activating factor (BAFF). The pharmacokinetics (PK) and pharmacodynamics (PD) of rozibafusp alfa were investigated in two randomized, placebo-controlled clinical studies: a phase Ia single ascending-dose study (7-700 mg subcutaneously (s.c.)) in healthy subjects and a phase Ib multiple ascending-dose study (70-420 mg s.c. every 2 weeks (q2w)) in patients with rheumatoid arthritis. Rozibafusp alfa exhibited nonlinear PK and dose-related and reversible dual-target engagement. Maximal reduction of naïve B cells from baseline (> 40%), reflective of BAFF inhibition, was achieved with rozibafusp alfa exposure (area under the concentration-time curve from time 0 to time infinity (AUCinf ) and AUC within a dosing interval from day 0 to day 14 (AUCtau )) above 51 and 57 days•µg/mL for the single-dose (≥ 70 mg) and multiple-dose studies (≥ 70 mg q2w), respectively. ICOSL receptor occupancy on circulating B cells, a surrogate PD end point for ICOSL inhibition, was directly related to drug concentration. PK/PD analysis showed > 90% RO at rozibafusp alfa ≥ 22.2 µg/mL (≥ 420-mg single dose or ≥ 210 mg q2w multiple dose), with saturation occurring at higher drug concentrations. These results informed the design and dose selection of a phase IIb study assessing the safety and efficacy of rozibafusp alfa in patients with active systemic lupus erythematosus.


Subject(s)
Arthritis, Rheumatoid , Lupus Erythematosus, Systemic , Humans , Area Under Curve , Arthritis, Rheumatoid/drug therapy , B-Cell Activating Factor/antagonists & inhibitors , Dose-Response Relationship, Drug , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Lupus Erythematosus, Systemic/drug therapy
7.
J Clin Immunol ; 32(6): 1253-61, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22706735

ABSTRACT

BACKGROUND: The role of costimulatory molecules expressed on lymphocytes and thyrocytes in hyperthyroidism has attracted increasing attention and research has shown a close correlation between variant expression of these molecules on lymphocytes and thyrocytes and the development of GD. MATERIALS AND METHODS: [corrected] Thyroid tissues were collected from GD patients during surgery and from Hashimoto disease (HT) and non-toxic goiter (NTG) patients as controls. ICOSL expression on infiltrated B cells and TFC was detected by flow cytometry (FCM), reverse transcription polymerase chain reaction (RT-PCR) and immunohistochemistry (IHC). Variation in ICOSL expression on TFC in primary cultures was analyzed in the absence or presence of cytokines using FCM assays. The role of ICOS-ICOSL signaling in proliferation, thyroid hormone production and thyroglobulin (Tg) release was investigated in primary TFC cultures using ICOS gene transfected L929 cells (ICOS-L929 cells) and the blocking ICOSL antibody (11 C4) in MTT assays and radioimmunoassays. RESULTS AND DISCUSSION: ICOSL expression on infiltrated B cells and TFC was detected in GD patient tissue. However, ICOSL expression was only detected on infiltrated B cells in control HT and NTG patient tissue. ICOSL expression on TFC was induced in vitro by the proinflammatory cytokines IFN-γ, IL-6 and TNF-α. Compared with mock transfected L929 (mock-L929) control cells, ICOS-L929 cells promoted significant proliferation of primary cultured TFC, with increased thyroid hormone and Tg production (all P < 0.01). TFC proliferation and production of thyroid hormones and Tg were inhibited significantly in the presence of ICOSL blocking antibody (11 C4) (all P < 0.05). Our observations suggest that ICOS-ICOSL signal plays a direct role in proliferation and differentiation of TFC and may exert important effects in the initiation, maintenance and exaggeration of autoimmune responses in local tissue.


Subject(s)
Goiter, Endemic/genetics , Graves Disease/genetics , Hashimoto Disease/genetics , Inducible T-Cell Co-Stimulator Ligand/genetics , Thyroid Gland/metabolism , Adult , Animals , Antibodies/pharmacology , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Gene Expression/drug effects , Goiter, Endemic/immunology , Goiter, Endemic/metabolism , Goiter, Endemic/pathology , Graves Disease/immunology , Graves Disease/metabolism , Graves Disease/pathology , Hashimoto Disease/immunology , Hashimoto Disease/metabolism , Hashimoto Disease/pathology , Humans , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Ligand/metabolism , Interferon-gamma/pharmacology , Interleukin-6/pharmacology , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/pathology , Male , Mice , Middle Aged , Primary Cell Culture , Signal Transduction/drug effects , Thyroid Gland/immunology , Thyroid Gland/pathology , Transfection , Tumor Necrosis Factor-alpha/pharmacology
8.
Int Immunopharmacol ; 99: 107979, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34293711

ABSTRACT

The strong genetic association between autoimmune regulator (AIRE) and autoimmune diseases indicates its critical role in immune tolerance. AIRE deficiency is thought to promote the development of follicular helper T (TFH) cells, which are considered to be essential in B cell proliferation. Excessive TFH cell generation is a key step towards the development of autoimmune diseases, including type 1 diabetes. However, the potential mechanism by which AIRE contributes to the generation and function of the TFH cell population has remained elusive. We show that AIRE reduced TFH cell generation by inhibiting the expression of inducible costimulatory ligand (ICOSL), interleukin (IL)-6 and IL-27 in dendritic cells (DCs). To understand the precise impact of AIRE-overexpressing bone marrow-derived DCs (AIRE-BMDCs) on type 1 diabetes progression and the associated molecular mechanisms, we transferred AIRE-BMDCs to recipient NOD mice and found that transplantation of AIRE-BMDCs can prevent or delay the onset of diabetes, attenuate diabetes after the establishment of overt hyperglycaemia, and lead to the inhibition of autoreactive pathological TFH cells and germinal centre (GC) B cells. To further determine the potential mechanism underlying this TFH cell depletion, BMDCs were cotransferred with recombinant mouse ICOSL (ICOSLG protein). We demonstrated that NOD mice were more susceptible to diabetes when they received AIRE-BMDCs and ICOSLG than when they received only mock-vehicle BMDCs (GFP-BMDCs). In addition, we did not observe the reversal of diabetes in any mice subjected to this cotransfer system. A single cycle of ICOSLG treatment temporarily promoted TFH cell proliferation and GC development. Our results reveal a mechanistic role of AIRE-BMDCs in the initiation of TFH cell differentiation, and the AIRE-mediated decrease in ICOSL expression in BMDCs plays a critical role. The effect of decreased ICOSL expression in type 1 diabetes will guide the design and evaluation of parallel studies in patients.


Subject(s)
Autoimmune Diseases/prevention & control , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Dendritic Cells/metabolism , Diabetes Mellitus, Type 1/prevention & control , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , T-Lymphocytes, Helper-Inducer/drug effects , Transcription Factors/biosynthesis , Animals , Dendritic Cells/transplantation , Female , Germinal Center , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Interleukin-6/antagonists & inhibitors , Interleukins/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Transcription Factors/genetics , AIRE Protein
9.
Cell Rep ; 36(11): 109705, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525377

ABSTRACT

Mouse embryonic stem cells (mESCs) can be maintained in vitro in defined N2B27 medium supplemented with two chemical inhibitors for GSK3 and MEK (2i) and the cytokine leukemia inhibitory factor (LIF), which act synergistically to promote self-renewal and pluripotency. Here, we find that genetic deletion of the four genes encoding the TCF/LEF transcription factors confers mESCs with the ability to self-renew in N2B27 medium alone. TCF/LEF quadruple knockout (qKO) mESCs display dysregulation of several genes, including Aire, Dnmt3l, and IcosL, located adjacent to each other within a topologically associated domain (TAD). Aire, Dnmt3l, and IcosL appear to be regulated by TCF/LEF in a ß-catenin independent manner. Moreover, downregulation of Aire and Dnmt3l in wild-type mESCs mimics the loss of TCF/LEF and increases mESC survival in the absence of 2iL. Hence, this study identifies TCF/LEF effectors that mediate exit from the pluripotent state.


Subject(s)
Cell Self Renewal , Hepatocyte Nuclear Factor 1-alpha/genetics , Lymphoid Enhancer-Binding Factor 1/genetics , Transcription Factor 7-Like 1 Protein/genetics , Transcription Factor 7-Like 2 Protein/genetics , Animals , Benzamides/pharmacology , Cell Self Renewal/drug effects , Culture Media/chemistry , Culture Media/pharmacology , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Down-Regulation/drug effects , Gene Editing , Hepatocyte Nuclear Factor 1-alpha/deficiency , Hepatocyte Nuclear Factor 1-alpha/metabolism , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Ligand/genetics , Inducible T-Cell Co-Stimulator Ligand/metabolism , Lymphoid Enhancer-Binding Factor 1/deficiency , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factor 7-Like 1 Protein/deficiency , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factor 7-Like 2 Protein/deficiency , Transcription Factor 7-Like 2 Protein/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin/deficiency , beta Catenin/genetics , AIRE Protein
10.
Front Immunol ; 9: 2227, 2018.
Article in English | MEDLINE | ID: mdl-30319662

ABSTRACT

CD4+CD25+Foxp3+ regulatory T cells (Tregs) accumulate in bone marrow microenvironment in acute myeloid leukemia (AML). However, little is known about how the tumor environment including tumor cells themselves affects this process. Here we demonstrated that AML cells expressed inducible T-cell costimulator ligand (ICOSL) that can provide costimulation through ICOS for the conversion and expansion of Tregs sustaining high Foxp3 and CD25 expression as well as a suppressive function. TNF-a stimulation up-regulated the expression of ICOSL. Furthermore, both the conversion and expansion of CD4+CD25+Foxp3+ T cells and CD4+ICOS+Foxp3+ T cells were induced by co-culture with AML cells overexpressed ICOSL. CD4+CD25+ICOS+ T cells possessed stronger ability to secrete IL-10 than CD4+CD25+ICOS- T cells. The mechanism by which IL-10 promoted the proliferation of AML cells was dependent on the activation of the Akt, Erk1/2, p38, and Stat3 signaling pathways. Blockade of ICOS signaling using anti-ICOSL antibody impaired the generation of Tregs and retarded the progression of an AML mice model injected with C1498 cells. The expression of ICOSL of patient AML cells and ICOS+ Tregs were found to be predictors for overall survival and disease-free survival in patients with AML, with ICOS+ Treg cell subset being a stronger predictor than total Tregs. These results suggest that ICOSL expression by AML cells may directly drive Treg expansion as a mechanism of immune evasion and ICOS+ Treg cell frequency is a better prognostic predictor in patients with AML.


Subject(s)
Gene Expression Regulation, Neoplastic/immunology , Inducible T-Cell Co-Stimulator Ligand/metabolism , Leukemia, Myeloid, Acute/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Escape/immunology , Adolescent , Adult , Animals , Biopsy , Bone Marrow/pathology , Cell Line, Tumor/transplantation , Cell Proliferation , Disease Models, Animal , Disease-Free Survival , Female , Humans , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Ligand/immunology , Interleukin-10/immunology , Interleukin-10/metabolism , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Mice, Inbred C57BL , Middle Aged , Primary Cell Culture , Prognosis , T-Lymphocytes, Regulatory/metabolism , Tumor Microenvironment/immunology , Young Adult
11.
Cancer Res ; 76(16): 4648-60, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27246829

ABSTRACT

The prognosis of follicular lymphoma (FL) patients is suspected to be influenced by tumor-infiltrating regulatory T cells (Treg). The mechanism of Treg enrichment in FL and their impact on malignant FL B cells remains to be elucidated. We analyzed 46 fresh lymph node biopsy samples, including FL (n = 20), diffuse large B-cell lymphoma (n = 10), classical Hodgkin lymphoma (n = 9), and reactive lymphadenitis (n = 7). Using multicolor flow cytometry and cell sorting, we observed an accumulation of CD25(high)CD127(low/neg) Tregs in FL tissues. These Tregs comprised activated ICOS(+) Tregs that were able to suppress not only conventional T cells, but also FL B cells. These FL B cells were able to express ICOSL in vitro and to generate CD25(high)FoxP3(high) Tregs expressing ICOS. Treg generation was associated with ICOS/ICOSL engagement and was abrogated by antagonist anti-ICOS and anti-ICOSL antibodies. Interactions between Tregs and FL B cells resulted in ICOSL downregulation on FL B cells. Our results highlight a key role for Tregs in FL pathogenesis and suggest that targeting the ICOS/ICOSL pathway may be a promising immunotherapy for FL treatment. Cancer Res; 76(16); 4648-60. ©2016 AACR.


Subject(s)
Inducible T-Cell Co-Stimulator Ligand/immunology , Inducible T-Cell Co-Stimulator Protein/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, B-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Cell Separation , Flow Cytometry , Humans , Immunohistochemistry , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Protein/antagonists & inhibitors , Mice , Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocytes, Regulatory/drug effects
12.
J Immunol Methods ; 424: 14-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25956037

ABSTRACT

Activation of the inducible costimulator (ICOS) signaling pathway in T cells is difficult to assess with bioassays, because most T cell lines do not constitutively express ICOS. Additionally, engagement of ICOS by its natural ligand B7 related protein 1 (B7RP1) is insufficient to elicit ICOS signaling, but requires simultaneous costimulation of the T cell receptor (TCR) to be effective. Here we describe a genetically engineered human T cell line that expresses a chimeric receptor (ICOS-CD3) consisting of full-length human ICOS fused at its C-terminal end to the cytoplasmic domain of human CD3 zeta. When engaged by B7RP1, ICOS-CD3 initiated signaling independently of TCR costimulation and induced substantially more IL-2 secretion in Jurkat T cells compared to wildtype ICOS. We demonstrate that this signaling-enhanced chimeric receptor can be used in simple and sensitive bioassays to detect bioactive B7RP1, anti-B7RP1 drugs, and the presence of corresponding neutralizing anti-drug antibodies.


Subject(s)
Inducible T-Cell Co-Stimulator Protein/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Biological Assay/methods , CD3 Complex/chemistry , CD3 Complex/genetics , CD3 Complex/metabolism , Cell Line , Cell Membrane/metabolism , Drug Evaluation, Preclinical/methods , Gene Expression , Humans , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Ligand/metabolism , Inducible T-Cell Co-Stimulator Protein/chemistry , Inducible T-Cell Co-Stimulator Protein/genetics , Interleukin-2/biosynthesis , Protein Binding , Protein Interaction Domains and Motifs/genetics , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/genetics , Signal Transduction/drug effects , T-Lymphocytes/immunology
13.
Monoclon Antib Immunodiagn Immunother ; 32(2): 125-31, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23607348

ABSTRACT

ICOSL, a newly identified member of the B7 superfamily, plays a major role in immune responses. In this study, a functional anti-human ICOSL monoclonal antibody (MAb) 3B3 was obtained and characterized by means of flow cytometry, Western blot, and competition assay. This MAb could specifically recognize a distinct epitope of the ICOSL molecule. As a functional antibody, MAb 3B3 could inhibit the proliferation of T lymphocytes stimulated by ICOSL-L929 transfectants. Furthermore, it could enhance IgG production of PWM-driven B cells. The results indicate that the ICOS-ICOSL signal is critically involved in specific humoral immunity.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , B-Lymphocytes/immunology , Immunoglobulin G/biosynthesis , Inducible T-Cell Co-Stimulator Ligand/immunology , Animals , B-Lymphocytes/metabolism , Binding, Competitive , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Epitope Mapping , Humans , Hybridomas , Immunity, Humoral , Immunoglobulin G/immunology , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Ligand/metabolism , Mice , Mice, Inbred BALB C , Pokeweed Mitogens/immunology , Pokeweed Mitogens/pharmacology , Protein Binding , T-Lymphocytes/drug effects , T-Lymphocytes/physiology
14.
Bone Marrow Transplant ; 48(1): 124-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22732700

ABSTRACT

As a member of the B7 family, inducible co-stimulator ligand (ICOSLG) expressed on tumor cell has been reported to have an important role in tumor immunity. In this study, we sought to determine whether the expression of ICOSLG in mouse hematological malignancy cells influences GVL reaction after mouse allogeneic BMT. In our study, we analyzed the expression of ICOSLG in six mice hematological malignancy cell lines for the first time, and found that FBL3, A20 and P388 cells expressed high levels of ICOSLG. Then, we chose A20 cells as targets to construct a GVL model and study the effects on the GVL reaction by silencing the ICOSLG gene. The survival was analyzed. We found that in GVL model, mortality of interference groups was significantly delayed compared with the control group (P=0.0005). Our results indicate that knockdown of ICOSLG of mouse leukemic cells may significantly enhance GVL effect after allogeneic BMT.


Subject(s)
Bone Marrow Transplantation/adverse effects , Graft vs Leukemia Effect , Inducible T-Cell Co-Stimulator Ligand/metabolism , Leukemia/therapy , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Gene Silencing , Gene Transfer Techniques , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Ligand/genetics , Leukemia/immunology , Leukemia/metabolism , Leukemia/pathology , Leukemic Infiltration/immunology , Leukemic Infiltration/pathology , Leukemic Infiltration/prevention & control , Liver/immunology , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Spleen/immunology , Spleen/pathology , Survival Analysis , Transplantation, Homologous
15.
Cancer Res ; 72(23): 6130-41, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23026134

ABSTRACT

Human breast tumors are infiltrated by memory CD4(+) T cells along with increased numbers of regulatory T cells (Treg) and plasmacytoid dendritic cells (pDC) that facilitate immune escape and correlate with poor prognosis. Here, we report that inducible costimulatory molecule (ICOS), a T cell costimulatory molecule of the CTLA4/PD1/CD28 family, is expressed mostly by tumor-associated Treg in primary breast tumors. A large proportion of these ICOS(+) Treg were Ki67(+) and this evident proliferative expansion was found to rely on interactions with tumor-associated pDC. Indeed, tumor-associated Treg highly expanded in presence of pDC but failed to proliferate under CD3/CD28 signal. In vitro experiments revealed that the addition of a neutralizing anti-ICOS antibody blocked pDC-induced Treg expansion and interleukin-10 secretion by memory CD4(+) T cells, establishing a pivotal role for ICOS in this process. Supporting these findings, the presence of ICOS(+) cells in clinical specimens of breast cancer correlated with a poor prognosis. Together, our results highlight an important relationship between Treg and pDC in breast tumors, and show that ICOS/ICOS-L interaction is a central event in immunosuppression of tumor-associated memory CD4(+) T cells. These findings strongly rationalize antibody-mediated ICOS blockade as a powerful clinical strategy to correct immune escape and promote therapeutic responses in breast cancer.


Subject(s)
Breast Neoplasms/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Disease Progression , Female , Humans , Immunohistochemistry , Inducible T-Cell Co-Stimulator Ligand/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Ligand/immunology , Inducible T-Cell Co-Stimulator Protein/antagonists & inhibitors , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Inducible T-Cell Co-Stimulator Protein/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Lymphocyte Activation , Retrospective Studies , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
SELECTION OF CITATIONS
SEARCH DETAIL