Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
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.
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
3.
Invest Ophthalmol Vis Sci ; 57(15): 6815-6823, 2016 12 01.
Article in English | MEDLINE | ID: mdl-28002569

ABSTRACT

Purpose: The interaction between the inducible costimulatory molecule (ICOS) and ICOS ligand (ICOSL) has been implicated in the differentiation and functions of T cells. The purpose of the present study was to determine the role of ICOS-ICOSL in the immune privilege of corneal allografts. Methods: Expression of ICOS and ICOSL mRNA from mouse eyes was assessed by RT-PCR. Corneas of C57BL/6 mice were orthotopically transplanted into the eyes of ICOS-/- BALB/c recipients and BALB/c wild-type (WT) recipients treated with anti-ICOSL mAb, and graft survival was assessed. A separate set of WT and ICOS-/- BALB/c mice received an anterior chamber injection of C57BL/6 splenocytes, and induction of allospecific anterior chamber-associated immune deviation (ACAID) was assessed. In vitro, cornea was incubated with T cells from WT and ICOS-/- BALB/c mice, and destruction of corneal endothelial cells (CECs) and the population of Foxp3+ CD25+ CD4+ T cells was assessed. Results: Inducible costimulatory molecule ligand mRNA was constitutively expressed in the cornea, iris-ciliary body, and retina. Allograft survival in ICOS-/- recipients and WT recipients treated with anti-ICOSL mAb was significantly shorter than in control recipients. Anterior chamber-associated immune deviation was induced less efficiently in ICOS-/- mice. Destruction of CECs by alloreactive ICOS-/- T cells was enhanced compared with WT T cells. After coincubation with allogeneic corneal tissue, the proportion of regulatory T cells was significantly greater among WT T cells than in ICOS-/- T cells. Conclusions: The expression of ICOSL in the cornea and the ICOS-mediated induction of Foxp3+ CD4+ regulatory T cells may contribute to successful corneal allograft survival.


Subject(s)
Cornea/metabolism , Corneal Transplantation , Gene Expression Regulation , Graft Survival/immunology , Immunity, Cellular , Inducible T-Cell Co-Stimulator Ligand/genetics , Inducible T-Cell Co-Stimulator Protein/genetics , Animals , Cornea/immunology , Cornea/pathology , Flow Cytometry , Graft Rejection/genetics , Graft Rejection/immunology , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Inducible T-Cell Co-Stimulator Protein/biosynthesis , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , Transplantation, Homologous
4.
Cell Immunol ; 297(1): 1-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26051632

ABSTRACT

Multiple myeloma (MM) produces significant cellular and humoral immune defects. We have previously reported that MM induces CD4(+)CD25(+)FoxP3(+) cells (TRegs), via tumour expression of the immune checkpoint regulator, ICOS-L. We sought to define what impact the immunomodulatory drug lenalidomide, alone or with dexamethasone, has on TReg cell generation. Lenalidomide pre-treatment of MM cell lines reduced TReg generation and the concomitant TReg:TEff (CD4(+)CD25(+)FoxP3(-): effector T cells) ratio, as a consequence of reduced ICOSL transcription. Dexamethasone did not affect surface ICOS-L expression but did induce TReg cell apoptosis without impacting on TEff cell survival. Combined lenalidomide and dexamethasone significantly reduced both TReg induction and the TReg:TEff cell ratio. In vivo, serial analysis of the TReg:TEff ratio in MM patients on lenalidomide-dexamethasone therapy revealed a progressive reduction towards age-matched control values, though not complete correction. Our data demonstrate for the first time immune synergism to explain the observed immune-modulation associated with lenalidomide-dexamethasone therapy.


Subject(s)
Dexamethasone/therapeutic use , Immunologic Factors/therapeutic use , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Multiple Myeloma/drug therapy , T-Lymphocytes, Regulatory/cytology , Thalidomide/analogs & derivatives , Adult , Aged , Aged, 80 and over , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dexamethasone/pharmacology , Down-Regulation , Female , Humans , Immunologic Factors/pharmacology , Lenalidomide , Male , Middle Aged , Multiple Myeloma/pathology , T-Lymphocytes, Regulatory/immunology , Thalidomide/pharmacology , Thalidomide/therapeutic use
5.
Oncol Rep ; 33(6): 3075-84, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25963805

ABSTRACT

Immunotherapy is a promising strategy for the treatment of various types of cancer. An antibody that targets programmed death ligand-1 (PD-L1) pathway has been shown to be active towards various types of cancer, including melanoma and lung cancer. MPDL3280A, an anti-PD-L1 antibody, has shown clear clinical activity in PD-L1-overexpressing bladder cancer with an objective response rate of 40-50%, resulting in a breakthrough therapy designation granted by FDA. These events pronounce the importance of targeting the PD-L1 pathway in the treatment of bladder cancer. In the present study, we investigated the prognostic significance of the expression of three genes in the PD-L1 pathway, including PD-L1, B7.1 and PD-1, in three independent bladder cancer datasets in the Gene Expression Omnibus database. PD-L1, B7.1 and PD-1 were significantly associated with clinicopathological parameters indicative of a more aggressive phenotype of bladder cancer, such as a more advanced stage and a higher tumor grade. In addition, a high level expression of PD-L1 was associated with reduced patient survival. Of note, the combination of PD-L1 and B7.1 expression, but not other combinations of the three genes, were also able to predict patient survival. Our findings support the development of anti-PD-L1, which blocks PD-L1-PD-1 and B7.1-PD-L1 interactions, in treatment of bladder cancer. The observations were consistent in the three independent bladder cancer datasets consisting of a total of 695 human bladder specimens. The datasets were then assessed and it was found that the expression levels of the chemokine CC-motif ligand (CCL), CCL3, CCL8 and CCL18, were correlated with the PD-L1 expression level, while ADAMTS13 was differentially expressed in patients with a different survival status (alive or deceased). Additional investigations are required to elucidate the role of these genes in the PD-L1-mediated immune system suppression and bladder cancer progression. In conclusion, findings of this study suggested that PD-L1 is an important prognostic marker and a therapeutic target for bladder cancer.


Subject(s)
B7-H1 Antigen/biosynthesis , Biomarkers, Tumor/biosynthesis , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Programmed Cell Death 1 Receptor/biosynthesis , Urinary Bladder Neoplasms/genetics , Adult , Aged , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy , Inducible T-Cell Co-Stimulator Ligand/genetics , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Programmed Cell Death 1 Receptor/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
6.
J Immunol ; 193(6): 2753-63, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25108021

ABSTRACT

Immune cells regulate cell surface receptor expression during their maturation, activation, and motility. Although many of these receptors are regulated largely at the level of expression, protease-mediated ectodomain shedding represents an alternative means of refashioning the surface of immune cells. Shedding is largely attributed to a family of a disintegrin and metalloprotease domain (ADAM) metalloproteases, including ADAM17. Although ADAM17 is well known to contribute to the innate immune response, mainly by releasing TNF-α, much less is known about whether/how this metalloprotease regulates adaptive immunity. To determine whether ADAM17 contributes to regulating adaptive immune responses, we took advantage of ADAM17 hypomorphic (ADAM17(ex/ex)) mice, in which ADAM17 expression is reduced by 90-95% compared with wild-type littermates. In this study, we show that that ADAM17 deficiency results in spleen and lymph node enlargement, as well as increased levels of Ag-specific class-switched Ig production following immunization with OVA together with anti-CD40 mAbs and polyinosinic-polycytidylic acid. Moreover, we demonstrate that the costimulatory ligand ICOS ligand (ICOSL) is selectively downregulated on the surface of B cells in an ADAM17-specific manner, although it is not proteolitically processed by recombinant ADAM17 in vitro. Finally, we show that higher cell surface levels of ICOSL in ADAM17(ex/ex) mice may contribute to the development of excessive Ab responses. Therefore, our data suggest a functional link between ADAM17 and ICOSL in controlling adaptive immune responses.


Subject(s)
ADAM Proteins/immunology , B-Lymphocytes/immunology , Immunity, Humoral , Inducible T-Cell Co-Stimulator Ligand/immunology , ADAM Proteins/genetics , ADAM17 Protein , Animals , Antibodies, Monoclonal/immunology , Antibody Formation/immunology , B-Lymphocytes/transplantation , CD40 Antigens/immunology , Cells, Cultured , Female , Immunoglobulin Class Switching , Immunoglobulin Isotypes/biosynthesis , Immunoglobulin Isotypes/immunology , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Lymph Nodes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Poly I-C/immunology , Spleen/pathology
7.
Am J Pathol ; 182(6): 2121-31, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23562272

ABSTRACT

Cellular immunotherapy may provide a strategy to overcome the poor prognosis of metastatic and recurrent rhabdomyosarcoma (RMS) under the current regimen of polychemotherapy. Because little is known about resistance mechanisms of RMS to cytotoxic T cells, we investigated RMS cell lines and biopsy specimens for expression and function of immune costimulatory receptors and anti-apoptotic molecules by RT-PCR, Western blot analysis, IHC, and cytotoxicity assays using siRNA or transfection-modified RMS cell lines, together with engineered RMS-directed cytotoxic T cells specific for the fetal acetylcholine receptor. We found that costimulatory CD80 and CD86 were consistently absent from all RMSs tested, whereas inducible T-cell co-stimulator ligand (ICOS-L; alias B7H2) was expressed by a subset of RMSs and was inducible by tumor necrosis factor α in two of five RMS cell lines. Anti-apoptotic survivin, along with other inhibitor of apoptosis (IAP) family members (cIAP1, cIAP2, and X-linked inhibitor of apoptosis protein), was overexpressed by RMS cell lines and biopsy specimens. Down-regulation of survivin by siRNA or pharmacologically in RMS cells increased their susceptibility toward a T-cell attack, whereas induction of ICOS-L did not. Treatment of RMS-bearing Rag(-/-) mice with fetal acetylcholine receptor-specific chimeric T cells delayed xenograft growth; however, this happened without definitive tumor eradication. Combined blockade of survivin and application of chimeric T cells in vivo suppressed tumor proliferation during survivin inhibition. In conclusion, survivin blockade provides a strategy to sensitize RMS cells for T-cell-based therapy.


Subject(s)
Inhibitor of Apoptosis Proteins/immunology , Receptors, Cholinergic/immunology , Rhabdomyosarcoma/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , B7-1 Antigen/biosynthesis , B7-2 Antigen/biosynthesis , Biopsy , CD28 Antigens/immunology , Child, Preschool , Costimulatory and Inhibitory T-Cell Receptors/metabolism , Cytotoxicity, Immunologic/immunology , Female , Gene Knockdown Techniques , Humans , Immunotherapy, Adoptive/methods , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Inducible T-Cell Co-Stimulator Ligand/immunology , Infant , Inhibitor of Apoptosis Proteins/genetics , Ligands , Male , Mice , Mice, Knockout , Neoplasm Transplantation , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/prevention & control , Signal Transduction/immunology , Survivin , T-Lymphocytes/transplantation , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/immunology
8.
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
9.
Clin Dev Immunol ; 2012: 934161, 2012.
Article in English | MEDLINE | ID: mdl-22969819

ABSTRACT

Abnormalities in monocytes and in peripheral blood dendritic cells (DC) subsets have been reported in systemic lupus erythematosus (SLE). We aim to clarify the tolerogenic or inflammatory role of these cells based on ICOSL or IFN-α and chemokine mRNA expression, respectively, after cell purification. The study included 18 SLE patients with active disease (ASLE), 25 with inactive disease (ISLE), and 30 healthy controls (HG). In purified plasmacytoid DC (pDC) was observed a lower ICOSL mRNA expression in ASLE and an increase in ISLE; similarly, a lower ICOSL mRNA expression in monocytes of ALSE patients was found. However, a higher ICOSL mRNA expression was observed in ASLE compared to HG in myeloid DCs. Interestingly, clinical parameters seem to be related with ICOSL mRNA expression. Regarding the inflammatory activity it was observed in purified monocytes and CD14(-/low) CD16(+) DCs an increase of CCL2, CXCL9, and CXCL10 mRNA expression in ASLE compared to HG. In myeloid DC no differences were observed regarding chemokines, and IFN-α mRNA expression. In pDC, a higher IFN-α mRNA expression was observed in ASLE. Deviations in ICOSL, chemokine, and IFN-α mRNA expression in peripheral blood monocytes and dendritic cells subpopulations in SLE appear to be related to disease activity.


Subject(s)
Dendritic Cells/immunology , Leukocytes, Mononuclear/immunology , Lupus Erythematosus, Systemic/immunology , Adult , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/genetics , Chemokine CXCL9/biosynthesis , Chemokine CXCL9/genetics , Female , Humans , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Inducible T-Cell Co-Stimulator Ligand/genetics , Inflammation/immunology , Interferon-alpha/biosynthesis , Interferon-alpha/genetics , Lupus Erythematosus, Systemic/blood , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Self Tolerance/immunology
SELECTION OF CITATIONS
SEARCH DETAIL