Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Autoimmun ; 77: 89-95, 2017 02.
Article in English | MEDLINE | ID: mdl-27863820

ABSTRACT

B cells are involved in immune thrombocytopenia (ITP) pathophysiology by producing antiplatelet auto-antibodies. However more than a half of ITP patients do not respond to B cell depletion induced by rituximab (RTX). The persistence of splenic T follicular helper cells (TFH) that we demonstrated to be expanded during ITP and to support B cell differentiation and antiplatelet antibody-production may participate to RTX inefficiency. Whereas it is well established that the survival of TFH depends on B cells in animal models, nothing is known in humans yet. To determine the effect of B cell depletion on human TFH, we quantified B cells and TFH in the spleen and in the blood from ITP patients treated or not with RTX. We showed that B cell depletion led to a dramatic decrease in splenic TFH and in CXCL13 and IL-21, two cytokines predominantly produced by TFH. The absolute count of circulating TFH and serum CXCL13 also decreased after RTX treatment, whatever the therapeutic response. Therefore, we showed that the maintenance of TFH required B cells and that TFH are not involved in the inefficiency of RTX in ITP.


Subject(s)
B-Lymphocytes/immunology , Lymphocyte Count , Lymphocyte Depletion , Purpura, Thrombocytopenic, Idiopathic/immunology , Spleen/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adult , Aged , B-Lymphocytes/metabolism , Biomarkers , Combined Modality Therapy , Cytokines/metabolism , Female , Humans , Immunologic Factors/therapeutic use , Lymphocyte Activation , Male , Middle Aged , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/diagnosis , Purpura, Thrombocytopenic, Idiopathic/therapy , Rituximab/therapeutic use , Spleen/metabolism , Spleen/pathology , T-Lymphocytes, Helper-Inducer/metabolism , Young Adult
2.
J Autoimmun ; 72: 73-83, 2016 08.
Article in English | MEDLINE | ID: mdl-27236507

ABSTRACT

CD8(+) T cells participate in the pathogenesis of some vasculitides. However, little is known about their role in Giant Cell Arteritis (GCA). This study was conducted to investigate CD8(+) T cell involvement in the pathogenesis of GCA. Analyses were performed at diagnosis and after 3 months of glucocorticoid treatment in 34 GCA patients and 26 age-matched healthy volunteers. Percentages of CD8(+) T-cell subsets, spectratype analysis of the TCR Vß families of CD8(+) T cells, levels of cytokines and chemokines and immunohistochemistry of temporal artery biopsies (TAB) were assessed. Among total CD8(+) T cells, percentages of circulating cytotoxic CD8 T lymphocytes (CTL, CD3(+)CD8(+)perforin(+)granzymeB(+)), Tc17 (CD3(+)CD8(+)IL-17(+)), CD63(+)CD8(+) T cells and levels of soluble granzymes A and B were higher in patients than in controls, whereas the percentage of Tc1 cells (CD3(+)CD8(+)IFN-γ(+)) was similar. Moreover, CD8(+) T cells displayed a restricted TCR repertoire in GCA patients. Percentages of circulating CTL, Tc17 and soluble levels of granzymes A and B decreased after treatment. CXCR3 expression on CD8(+) T cells and its serum ligands (CXCL9, -10, -11) were higher in patients. Analyses of TAB revealed high expression of CXCL9 and -10 associated with infiltration by CXCR3(+)CD8(+) T cells expressing granzyme B and TiA1. The intensity of the CD8 T-cell infiltrate in TAB was predictive of the severity of the disease. This study demonstrates the implication and the prognostic value of CD8(+) T-cells in GCA and suggests that CD8(+) T-cells are recruited within the vascular wall through an interaction between CXCR3 and its ligands.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Giant Cell Arteritis/immunology , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Chemokine CXCL11/immunology , Chemokine CXCL11/metabolism , Chemokine CXCL9/immunology , Chemokine CXCL9/metabolism , Cytokines/metabolism , Female , Giant Cell Arteritis/drug therapy , Giant Cell Arteritis/metabolism , Glucocorticoids/therapeutic use , Granzymes/immunology , Granzymes/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Prednisone/therapeutic use , Prognosis , Prospective Studies , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism
3.
Blood ; 124(18): 2858-66, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25232056

ABSTRACT

Antiplatelet-antibody-producing B cells play a key role in immune thrombocytopenia (ITP) pathogenesis; however, little is known about T-cell dysregulations that support B-cell differentiation. During the past decade, T follicular helper cells (TFHs) have been characterized as the main T-cell subset within secondary lymphoid organs that promotes B-cell differentiation leading to antibody class-switch recombination and secretion. Herein, we characterized TFHs within the spleen of 8 controls and 13 ITP patients. We show that human splenic TFHs are the main producers of interleukin (IL)-21, express CD40 ligand (CD154), and are located within the germinal center of secondary follicles. Compared with controls, splenic TFH frequency is higher in ITP patients and correlates with germinal center and plasma cell percentages that are also increased. In vitro, IL-21 stimulation combined with an anti-CD40 agonist antibody led to the differentiation of splenic B cells into plasma cells and to the secretion of antiplatelet antibodies in ITP patients. Overall, these results point out the involvement of TFH in ITP pathophysiology and the potential interest of IL-21 and CD40 as therapeutic targets in ITP.


Subject(s)
Antibody Formation/immunology , B-Lymphocytes/pathology , Blood Platelets/immunology , Cell Differentiation/immunology , Purpura, Thrombocytopenic, Idiopathic/immunology , Spleen/pathology , T-Lymphocytes, Helper-Inducer/cytology , Adult , Aged , Antibody Formation/drug effects , Antigens, CD/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Blood Platelets/drug effects , CD40 Ligand/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Female , Germinal Center/pathology , Humans , Immunoglobulin G/biosynthesis , Interleukins/pharmacology , Lymphocyte Count , Male , Middle Aged , Phenotype , Plasma Cells/drug effects , Plasma Cells/metabolism , Plasma Cells/pathology , Purpura, Thrombocytopenic, Idiopathic/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology
4.
J Allergy Clin Immunol ; 135(6): 1614-24.e4, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25630940

ABSTRACT

BACKGROUND: Adoptive transfer of immunosuppressive cells has emerged as a promising strategy for the treatment of immune-mediated disorders. However, only a limited number of such cells can be isolated from in vivo specimens. Therefore efficient ex vivo differentiation and expansion procedures are critically needed to produce a clinically relevant amount of these suppressive cells. OBJECTIVE: We sought to develop a novel, clinically relevant, and feasible approach to generate ex vivo a subpopulation of human suppressor cells of monocytic origin, referred to as human monocyte-derived suppressive cells (HuMoSCs), which can be used as an efficient therapeutic tool to treat inflammatory disorders. METHODS: HuMoSCs were generated from human monocytes cultured for 7 days with GM-CSF and IL-6. The immune-regulatory properties of HuMoSCs were investigated in vitro and in vivo. The therapeutic efficacy of HuMoSCs was evaluated by using a graft-versus-host disease (GvHD) model of humanized mice (NOD/SCID/IL-2Rγc(-/-) [NSG] mice). RESULTS: CD33+ HuMoSCs are highly potent at inhibiting the proliferation and activation of autologous and allogeneic effector T lymphocytes in vitro and in vivo. The suppressive activity of these cells depends on signal transducer and activator of transcription 3 activation. Of therapeutic relevance, HuMoSCs induce long-lasting memory forkhead box protein 3-positive CD8+ regulatory T lymphocytes and significantly reduce GvHD induced with human PBMCs in NSG mice. CONCLUSION: Ex vivo-generated HuMoSCs inhibit effector T lymphocytes, promote the expansion of immunosuppressive forkhead box protein 3-positive CD8+ regulatory T cells, and can be used as an efficient therapeutic tool to prevent GvHD.


Subject(s)
Graft vs Host Disease/prevention & control , Monocytes/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/drug effects , Cell Proliferation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunosuppression Therapy , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Interleukin-6/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Monocytes/cytology , Monocytes/drug effects , Monocytes/transplantation , Primary Cell Culture , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transplantation, Heterologous
5.
Eur J Immunol ; 44(8): 2489-2499, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24777831

ABSTRACT

Primarily defined by their antigen-presenting property, dendritic cells (DCs) are being implemented as cancer vaccines in immunotherapeutic interventions. DCs can also function as direct tumor cell killers. How DC cytotoxic activity can be efficiently harnessed and the mechanisms controlling this nonconventional property are not fully understood. We report here that the tumoricidal potential of mouse DCs generated from myeloid precursors with GM-CSF and IL-15 (IL-15 DCs) can be triggered with the Toll-like receptor (TLR) 4 ligand lipopolysaccharide to a similar extent compared with that of their counterparts, conventionally generated with IL-4 (IL-4 DCs). The mechanism of tumor cell killing depends on the induction of iNOS expression by DCs. In contrast, interferon (IFN)-γ induces the cytotoxic activity of IL-4 but not IL-15 DCs. Although the IFN-γ-STAT-1 signaling pathway is overall functional in IL-15 DCs, IFN-γ fails to induce iNOS expression in these cells. iNOS expression is negatively controlled in IFN-γ-stimulated IL-15 DCs by the cooperation between the E3 SUMO ligase PIAS1 and STAT-3, and can be partially restored with PIAS1 siRNA and STAT-3 inhibitors.


Subject(s)
Dendritic Cells/metabolism , Interferon-gamma/metabolism , Interleukin-15/metabolism , Protein Inhibitors of Activated STAT/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-4/metabolism , Ligands , Lipopolysaccharides/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction/physiology , Toll-Like Receptor 4/metabolism
6.
Blood ; 122(14): 2477-86, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23963041

ABSTRACT

The pathogenic role of B cells in immune thrombocytopenia (ITP) has justified the therapeutic use of anti-CD20 antibodies such as rituximab (RTX). However, 60% of ITP patients do not respond to RTX. To decipher the mechanisms implicated in the failure of RTX, and because the spleen plays a well-recognized role in ITP pathogenesis, 12 spleens from ITP patients who had been nonresponders to RTX therapy were compared with 11 spleens from RTX-untreated ITP patients and 9 controls. We here demonstrate that in RTX-nonresponder ITP patients, preferential Th1 and Tc1 T lymphocyte polarizations occur, associated with an increase in splenic effector memory CD8(+) T-cell frequency. Moreover, in the RTX- nonresponder patient group, the CD8(+) T-cell repertoire displays a restricted pattern. In the blood, the phenotype of CD8(+) T cells before and after RTX treatment is not modified in responders or nonresponders. Altogether, these results demonstrate for the first time an activation of splenic CD8(+) T cells in ITP patients who did not respond to RTX and suggest their involvement in platelet destruction in these patients.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Drug Resistance/immunology , Lymphocyte Activation/immunology , Purpura, Thrombocytopenic, Idiopathic/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Female , Humans , Immunohistochemistry , Immunologic Factors/therapeutic use , Male , Middle Aged , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Real-Time Polymerase Chain Reaction , Rituximab , Spleen/immunology , Young Adult
7.
Blood ; 117(5): 1555-64, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-21123824

ABSTRACT

Therapeutic strategies combining the induction of effective antitumor immunity with the inhibition of the mechanisms of tumor-induced immunosuppression represent a key objective in cancer immunotherapy. Herein we demonstrate that effector/memory CD4(+) T helper-1 (Th-1) lymphocytes, in addition to polarizing type-1 antitumor immune responses, impair tumor-induced CD4(+)CD25(+)FoxP3(+) regulatory T lymphocyte (Treg) immunosuppressive function in vitro and in vivo. Th-1 cells also inhibit the generation of FoxP3(+) Tregs from naive CD4(+)CD25(-)FoxP3(-) T cells by an interferon-γ-dependent mechanism. In addition, in an aggressive mouse leukemia model (12B1), Th-1 lymphocytes act synergistically with a chaperone-rich cell lysate (CRCL) vaccine, leading to improved survival and long-lasting protection against leukemia. The combination of CRCL as a source of tumor-specific antigens and Th-1 lymphocytes as an adjuvant has the potential to stimulate efficient specific antitumor immunity while restraining Treg-induced suppression.


Subject(s)
Cancer Vaccines/administration & dosage , Cell Extracts/administration & dosage , Forkhead Transcription Factors/metabolism , Immunologic Memory/immunology , Leukemia, Experimental/therapy , Molecular Chaperones/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Blotting, Western , Cancer Vaccines/immunology , Cell Extracts/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interferon-gamma/metabolism , Leukemia, Experimental/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/metabolism , Tumor Cells, Cultured
8.
Blood ; 118(16): 4394-400, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-21876120

ABSTRACT

Immune thrombocytopenia (ITP) is an autoimmune disease with a complex pathogenesis. As in many B cell-related autoimmune diseases, rituximab (RTX) has been shown to increase platelet counts in some ITP patients. From an immunologic standpoint, the mode of action of RTX and the reasons underlying its limited efficacy have yet to be elucidated. Because splenectomy is a cornerstone treatment of ITP, the immune effect of RTX on this major secondary lymphoid organ was investigated in 18 spleens removed from ITP patients who were treated or not with RTX. Spleens from ITP individuals had follicular hyperplasia consistent with secondary follicles. RTX therapy resulted in complete B-cell depletion in the blood and a significant reduction in splenic B cells, but these patients did not achieve remission. Moreover, whereas the percentage of circulating regulatory T cells (Tregs) was similar to that in controls, splenic Tregs were reduced in ITP patients. Interestingly, the ratio of proinflammatory Th1 cells to suppressive Tregs was increased in the spleens of patients who failed RTX therapy. These results indicate that although B cells are involved in ITP pathogenesis, RTX-induced total B-cell depletion is not correlated with its therapeutic effects, which suggests additional immune-mediated mechanisms of action of this drug.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Immunologic Factors/immunology , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Spleen/drug effects , Adult , Aged , Antibodies, Monoclonal, Murine-Derived/therapeutic use , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Female , Humans , Immunologic Factors/therapeutic use , Male , Middle Aged , Purpura, Thrombocytopenic, Idiopathic/immunology , Rituximab , Spleen/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/drug effects , Th1 Cells/immunology
9.
Arthritis Rheum ; 64(8): 2499-503, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22488116

ABSTRACT

OBJECTIVE: From an immunologic standpoint, the mechanisms by which treatment with tocilizumab (TCZ), a humanized anti-interleukin-6 (anti-IL-6) receptor antibody, results in improvement in rheumatoid arthritis (RA) patients are still not fully understood. In vitro studies and studies in mouse models have demonstrated the critical role of IL-6 in Th17 cell differentiation. Th17 lymphocytes have been shown to be strongly involved in RA pathogenesis, and the purpose of this study was to investigate the effect of IL-6 blockade on the balance between Th17 cells and Treg cells in patients with active RA. METHODS: Patients with active RA for whom TCZ had been prescribed by a rheumatologist were enrolled in this study. Phenotypic analyses of T cell populations were performed, and the Disease Activity Score in 28 joints (DAS28) was assessed. Serum cytokine levels and other parameters of inflammation were measured before the first infusion and after the third infusion of TCZ (8 mg/kg). RESULTS: Compared to controls, levels of Th17 cells (CD4+IL-17+) were increased and Treg cells (CD4+CD25(high) FoxP3+) were decreased in the peripheral blood of patients with active RA. The suppressive function of circulating Treg cells was not impaired in patients with active RA. TCZ treatment induced a significant decrease in the DAS28 associated with a significant decrease in the percentage of Th17 cells (from a median of 0.9% to 0.45%; P = 0.009) and an increase in the percentage of Treg cells (from a median of 3.05% to 3.94%; P = 0.0039) in all patients. CONCLUSION: This study demonstrates for the first time that inhibition of IL-6 function by TCZ corrects the imbalance between Th17 cells and Treg cells in patients with RA.


Subject(s)
Arthritis, Rheumatoid/pathology , Interleukin-6/antagonists & inhibitors , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology , Adult , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/drug therapy , Case-Control Studies , Cell Count , Female , Humans , Interleukin-6/blood , Male , Middle Aged , Phenotype , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/drug effects , Severity of Illness Index
10.
Arthritis Rheum ; 64(11): 3788-98, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22833233

ABSTRACT

OBJECTIVE: Giant cell arteritis (GCA) is the most frequently occurring vasculitis in elderly individuals, and its pathogenesis is not fully understood. The objective of this study was to decipher the role of the major CD4+ T cell subsets in GCA and its rheumatologic form, polymyalgia rheumatica (PMR). METHODS: A prospective study of the phenotype and the function of major CD4+ T cell subsets (Th1, Th17, and Treg cells) was performed in 34 untreated patients with GCA or PMR, in comparison with 31 healthy control subjects and with the 27 treated patients who remained after the 7 others withdrew. RESULTS: Compared with control subjects, patients with GCA and patients with PMR had a decreased frequency of Treg cells and Th1 cells, whereas the percentage of Th17 cells was significantly increased. Furthermore, an analysis of temporal artery biopsy specimens obtained from patients affected by GCA for whom biopsy results were positive demonstrated massive infiltration by Th17 and Th1 lymphocytes without any Treg cells. After glucocorticoid treatment, the percentages of circulating Th1 and Th17 cells decreased, whereas no change in the Treg cell frequency was observed. The frequency of CD161+CD4+ T cells, which are considered to be Th17 cell precursors, was similar in patients and control subjects. However, these cells highly infiltrated GCA temporal artery biopsy specimens, and their ability to produce interleukin-17 in vitro was significantly enhanced in patients with GCA and patients with PMR and was correlated with a decrease in the phosphorylated form of STAT-1. CONCLUSION: This study is the first to demonstrate that the frequency of Treg cells is decreased in patients with GCA and patients with PMR, and that CD161+CD4+ T lymphocytes, differentiated into Th1 cells and Th17 cells, are involved in the pathogenesis of GCA and PMR.


Subject(s)
Giant Cell Arteritis/immunology , NK Cell Lectin-Like Receptor Subfamily B/immunology , Polymyalgia Rheumatica/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Adult , Aged , Cell Differentiation/immunology , Cells, Cultured , Female , Flow Cytometry , Giant Cell Arteritis/pathology , Humans , Male , Middle Aged , Polymyalgia Rheumatica/pathology , Prospective Studies , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/cytology , Th17 Cells/cytology
11.
J Immunol ; 187(5): 2775-82, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21804019

ABSTRACT

Known for years as professional APCs, dendritic cells (DCs) are also endowed with tumoricidal activity. This dual role of DC as killers and messengers may have important implications for tumor immunotherapy. However, the tumoricidal activity of DCs has mainly been investigated in animal models. Cancer cells inhibit antitumor immune responses using numerous mechanisms, including the induction of immunosuppressive/ tolerogenic DCs that have lost their ability to present Ags in an immunogenic manner. In this study, we evaluated the possibility of generating tumor killer DCs from patients with advanced-stage cancers. We demonstrate that human monocyte-derived DCs are endowed with significant cytotoxic activity against tumor cells following activation with LPS. The mechanism of DC-mediated tumor cell killing primarily involves peroxynitrites. This observed cytotoxic activity is restricted to immature DCs. Additionally, after killing, these cytotoxic DCs are able to activate tumor Ag-specific T cells. These observations may open important new perspectives for the use of autologous cytotoxic DCs in cancer immunotherapy strategies.


Subject(s)
Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Immunotherapy/methods , Neoplasms/immunology , Dendritic Cells/metabolism , Flow Cytometry , Humans , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/immunology , Neoplasms/therapy , Peroxynitrous Acid/immunology , Peroxynitrous Acid/metabolism , T-Lymphocytes/immunology
12.
J Immunol ; 187(12): 6310-7, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22075702

ABSTRACT

Dendritic cells (DCs) encompass a heterogeneous population of cells capable of orchestrating innate and adaptive immune responses. The ability of DCs to act as professional APCs has been the foundation for the development and use of these cells as vaccines in cancer immunotherapy. DCs are also endowed with the nonconventional property of directly killing tumor cells. The current study investigates the regulation of murine DC cytotoxic function by T lymphocytes. We provide evidence that CD4(+) Th-1, but not Th-2, Th-17 cells, or regulatory T cells, are capable of inducing DC cytotoxic function. IFN-γ was identified as the major factor responsible for Th-1-induced DC tumoricidal activity. Tumor cell killing mediated by Th-1-activated killer DCs was dependent on inducible NO synthase expression and NO production. Importantly, Th-1-activated killer DCs were capable of presenting the acquired Ags from the killed tumor cells to T lymphocytes in vitro or in vivo. These observations offer new possibilities for the application of killer DCs in cancer immunotherapy.


Subject(s)
Cytotoxicity, Immunologic , Dendritic Cells/immunology , Interferon-gamma/physiology , Mammary Neoplasms, Experimental/immunology , Melanoma, Experimental/immunology , Receptors, Interferon/physiology , Th1 Cells/immunology , Th1 Cells/metabolism , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic/genetics , Dendritic Cells/metabolism , Female , Interferon-gamma/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Interferon gamma Receptor
13.
J Immunol ; 184(4): 1876-84, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20089706

ABSTRACT

Dendritic cells (DCs), essential for the initiation and regulation of adaptive immune responses, have been used as anticancer vaccines. DCs may also directly trigger tumor cell death. In the current study, we have investigated the tumoricidal and immunostimulatory activities of mouse bone marrow-derived DCs. Our results indicate that these cells acquire killing capabilities toward tumor cells only when activated with LPS or Pam3Cys-SK4. Using different transgenic mouse models including inducible NO synthase or GP91 knockout mice, we have further established that LPS- or Pam3Cys-SK4-activated DC killing activity involves peroxynitrites. Importantly, after killing of cancer cells, DCs are capable of engulfing dead tumor cell fragments and of presenting tumor Ags to specific T lymphocytes. Thus, upon specific stimulation, mouse bone marrow-derived DCs can directly kill tumor cells through a novel peroxynitrite-dependent mechanism and participate at virtually all levels of antitumor immune responses, which reinforces their interest in immunotherapy.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Peroxynitrous Acid/toxicity , Animals , Cell Death/immunology , Cell Line, Tumor , Coculture Techniques , HeLa Cells , Humans , Melanoma, Experimental , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms, Experimental/metabolism
14.
Radiol Case Rep ; 17(10): 3607-3610, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35923339

ABSTRACT

Radiofrequency ablation (RFA) has widespread popularity due to its immune-modulation effects in many cancers. Optimal settings to apply RFA in pancreatic cancer, in which the advanced stage of the tumor at the diagnosis makes various therapeutic approaches fail, are still demanding. We report the case of a patient with unresectable pancreatic cancer in which 3 repetitive RFA has been applied over a period of 3 months. Results revealed an improvement in the patient's clinical condition associated with the reduced incidence of CD4+CD45RO+ T lymphocytes and declined TGF-ß level in serum. The good quality of life and disease-free survival were maintained for the next months. Booster application of RFA procedure might be a promising option to improve the quality of life in pancreatic cancer patients.

15.
Front Immunol ; 13: 827712, 2022.
Article in English | MEDLINE | ID: mdl-35345675

ABSTRACT

Recently developed cell-based therapies have shown potential for graft-versus-host disease (GvHD) mitigation. Our team previously developed a protocol to generate human monocyte-derived suppressor Cells (HuMoSC), a subpopulation of CD33+ suppressor cells of monocytic origin. CD33+HuMoSC successfully reduced xenoGvHD severity in NOD/SCID/IL-2Rγc-/- (NSG) mice. While CD33+ HuMoSC culture supernatant inhibits T cell activation and proliferation, the recovery of CD33+ HuMoSC immunosuppressive cells and the subsequent production of their supernatant is limited. An attractive solution would be to use both the CD33+ and the large number of CD14+ cells derived from our protocol. Here, we assessed the immunoregulatory properties of the CD14+HuMoSC supernatant and demonstrated that it inhibited both CD4 and CD8 T cell proliferation and decreased CD8 cytotoxicity. In vivo, injection of CD14+HuMoSC supernatant reduced xenoGvHD in NSG mice. Furthermore, CD14+HuMoSC supernatant maintained its immunoregulatory properties in an inflammatory environment. Proteomic and multiplex analyses revealed the presence of immunosuppressive proteins such as GPNMB, galectin-3 and IL-1R(A) Finally, CD14+HuMoSC supernatant can be produced using good manufacturing practices and be used as complement to current immunosuppressive drugs. CD14+HuMoSC supernatant is thus a promising therapy for preventing GvHD. .


Subject(s)
Graft vs Host Disease , Monocytes , Animals , CD8-Positive T-Lymphocytes , Graft vs Host Disease/metabolism , Graft vs Host Disease/prevention & control , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Monocytes/metabolism , Proteomics
16.
Clin Dev Immunol ; 2011: 430394, 2011.
Article in English | MEDLINE | ID: mdl-22110524

ABSTRACT

Tumor cells commonly escape from elimination by innate and adaptive immune responses using multiple strategies among which is the active suppression of effector immune cells. Regulatory T lymphocytes (Treg) and tolerogenic dendritic cells play essential roles in the establishment and persistence of cancer-induced immunosuppression. Differentiating dendritic cells (DCs) exposed to tumor-derived factors may be arrested at an immature stage becoming inept at initiating immune responses and may induce effector T-cell anergy or deletion. These tolerogenic DCs, which accumulate in patients with different types of cancers, are also involved in the generation of Treg. In turn, Treg that expand during tumor progression contribute to the immune tolerance of cancer by impeding DCs' ability to orchestrate immune responses and by directly inhibiting antitumoral T lymphocytes. Herein we review these bidirectional communications between DCs and Treg as they relate to the promotion of cancer-induced tolerance.


Subject(s)
Dendritic Cells/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Escape , Animals , Antigens, Neoplasm/immunology , Cell Communication , Cell Differentiation , Cells, Cultured , Clonal Anergy , Dendritic Cells/pathology , Disease Progression , Humans , Immune Tolerance , Immunosuppression Therapy , Neoplasms , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
18.
Oncoimmunology ; 10(1): 1880046, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33659098

ABSTRACT

Background: Immunosuppressive cell-based therapy is a recent strategy for controlling Graft-versus-Host Disease (GvHD). Such cells ought to maintain their suppressive function in inflammatory conditions and in the presence of immunosuppressive agents currently used in allogeneic hematopoietic cell transplantation (allo-HCT). Moreover, these therapies should not diminish the benefits of allo-HCT, the Graft-versus-Leukemia (GvL) effect. We have previously reported on a novel subset of human monocyte-derived suppressor cells (HuMoSC) as a prospective approach for controlling GvHD.Objective.The objective of this study was to explore the therapeutic relevance of the HuMoSC in clinical conditions. Methods: Immune regulatory functions of HuMoSC were assessed in inflammatory conditions and in the presence of immunosuppressants. The therapeutic efficiency of the association of HuMoSC with immunosuppressants was evaluated in an experimental model of GvHD induced by human PBMC in NOD/SCID/IL2-Rγc-/- (NSG) mice.Interestingly, the inhibitory functions of HuMoSC against T lymphocytes and their ability to polarize Treg are preserved, in vitro, in inflammatory environments and are not affected by immunosuppressive agents. In vivo, the association of HuMoSC-based treatment with an immunosuppressive drug showed a synergistic effect for controlling GvHD. Furthermore, HuMoSC control GvHD while preserving GvL effect in a xeno-GvHD conditioned mouse model with cell neoplasm (CAL-1). HuMoSC are generated according to good manufacturing practices (GMP) and we demonstrated that these cells tolerate long-term preservation with unaltered phenotype and function.Conclusion.HuMoSC-based therapy represents a promising approach for controlling GvHD and could be quickly implemented in clinical practice.


Subject(s)
Graft vs Host Disease , Leukemia , Animals , Graft vs Host Disease/prevention & control , Humans , Leukocytes, Mononuclear , Mice , Mice, Inbred NOD , Mice, SCID , Monocytes
19.
Gastroenterology ; 137(3): 965-75, 975.e1-10, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19450596

ABSTRACT

BACKGROUND & AIMS: NHE3 is a target of inhibition by proinflammatory cytokines and pathogenic bacteria, an event contributing to diarrhea in infectious and idiopathic colitis. In mice, NHE3 deficiency leads to mild diarrhea, increased intestinal expression of interferon (IFN)-gamma, and distal colitis, suggesting its role in epithelial barrier homeostasis. Our aim was to investigate the role of NHE3 in maintaining mucosal integrity. METHODS: Control or dextran sulfate sodium (DSS)-treated, 6- to 8-week-old wild-type (WT) and NHE3(-/-) mice were used for the experiments. Small intestines were dissected for further analysis. RESULTS: NHE3(-/-) mice have elevated numbers of CD8alpha(+) T and natural killer cells in the intraepithelial lymphocytes and lamina propria lymphocytes compartments, representing the source of IFN-gamma. NHE3(-/-) mice display alterations in epithelial gene and protein expression patterns that predispose them to a high susceptibility to DSS, with accelerated mortality resulting from intestinal bleeding, hypovolemic shock, and sepsis, even at a very low DSS concentration. Microarray analysis and intestinal hemorrhage indicate that NHE3 deficiency predisposes mice to DSS-induced small intestinal injury, a segment never reported as affected by DSS, and demonstrate major differences in the colonic response to DSS challenge in WT and NHE3(-/-) mice. In NHE3(-/-) mice, broad-spectrum oral antibiotics or anti-asialo GM1 antibodies reduce the expression of IFN-gamma and iNOS to basal levels and delay but do not prevent severe mortality in response to DSS treatment. CONCLUSIONS: These results suggest that NHE3 participates in mucosal responses to epithelial damage, acting as a modifier gene determining the extent of the gut inflammatory responses in the face of intestinal injury.


Subject(s)
Dextran Sulfate/toxicity , Homeostasis , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Sodium-Hydrogen Exchangers/physiology , Animals , Blood Cell Count , Colon/metabolism , Down-Regulation , Endothelin-1/metabolism , G(M1) Ganglioside/metabolism , Gastrointestinal Hemorrhage/chemically induced , Interferon-gamma/metabolism , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestine, Small/pathology , Mice , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Signal Transduction , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics
20.
J Immunol ; 181(10): 6955-63, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18981115

ABSTRACT

Imatinib mesylate (Gleevec, STI571), a selective inhibitor of a restricted number of tyrosine kinases, has been effectively used for the treatment of Philadelphia chromosome-positive leukemias and gastrointestinal stromal tumors. Imatinib may also directly influence immune cells. Suppressive as well as stimulating effects of this drug on CD4(+) and CD8(+) T lymphocytes or dendritic cells have been reported. In the current study, we have investigated the influence of imatinib mesylate on CD4(+)CD25(+)FoxP3(+) regulatory T cells (Treg), a critical population of lymphocytes that contributes to peripheral tolerance. Used at concentrations achieved clinically, imatinib impaired Treg immunosuppressive function and FoxP3 expression but not production of IL-10 and TGF-beta in vitro. Imatinib significantly reduced the activation of the transcription factors STAT3 and STAT5 in Treg. Analysis of Treg TCR-induced signaling cascade indicated that imatinib inhibited phosphorylation of ZAP70 and LAT. Substantiating these observations, imatinib treatment of mice decreased Treg frequency and impaired their immunosuppressive function in vivo. Furthermore, imatinib mesylate significantly enhanced antitumor immune responses to dendritic cell-based immunization against an imatinib-resistant BCR-ABL negative lymphoma. The clinical applications of imatinib mesylate might thus be expanded with its use as a potent immunomodulatory agent targeting Treg in cancer immunotherapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Dendritic Cells/transplantation , Immunotherapy, Active/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Piperazines/administration & dosage , Pyrimidines/administration & dosage , T-Lymphocytes, Regulatory/drug effects , Animals , Benzamides , Blotting, Western , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/drug effects , Imatinib Mesylate , Immunohistochemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/drug effects , STAT5 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL