Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Proc Natl Acad Sci U S A ; 120(7): e2208509120, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36745791

ABSTRACT

Antigenic peptides derived from introns are presented on major histocompatibility (MHC) class I molecules, but how these peptides are produced is poorly understood. Here, we show that an MHC class I epitope (SL8) sequence inserted in the second intron of the ß-globin gene in a C57BL/6 mouse (HBB) generates immune tolerance. Introduction of SL8-specific CD8+ T cells derived from OT-1 transgenic mice resulted in a threefold increase in OT-1 T cell proliferation in HBB animals, as compared to wild-type animals. The growth of MCA sarcoma cells expressing the intron-derived SL8 epitope was suppressed in wild-type animals compared to HBB mice. The ß-globin pre-mRNA was detected in the light polysomal fraction, and introducing stop codons identified a non-AUG initiation site between +228 and +255 nts upstream of the SL8. Isolation of ribosome footprints confirmed translation initiation within this 27 nt sequence. Furthermore, treatment with splicing inhibitor shifts the translation of the pre-mRNA to monosomal fractions and results in an increase of intron-derived peptide substrate as shown by polysome profiling and cell imaging. These results show that non-AUG-initiated translation of pre-mRNAs generates peptides for MHC class I immune tolerance and helps explain why alternative tissue-specific splicing is tolerated by the immune system.


Subject(s)
Histocompatibility Antigens Class I , RNA Precursors , Animals , Mice , Histocompatibility Antigens Class I/genetics , RNA Precursors/genetics , CD8-Positive T-Lymphocytes , Protein Biosynthesis , Antigen Presentation , Mice, Inbred C57BL , Peptides/metabolism , Immune Tolerance/genetics , Epitopes , Histocompatibility Antigens Class II/genetics
2.
Circ Res ; 133(4): 298-312, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37435729

ABSTRACT

BACKGROUND: Giant cell arteritis (GCA) causes severe inflammation of the aorta and its branches and is characterized by intense effector T-cell infiltration. The roles that immune checkpoints play in the pathogenesis of GCA are still unclear. Our aim was to study the immune checkpoint interplay in GCA. METHODS: First, we used VigiBase, the World Health Organization international pharmacovigilance database, to evaluate the relationship between GCA occurrence and immune checkpoint inhibitors treatments. We then further dissected the role of immune checkpoint inhibitors in the pathogenesis of GCA, using immunohistochemistry, immunofluorescence, transcriptomics, and flow cytometry on peripheral blood mononuclear cells and aortic tissues of GCA patients and appropriated controls. RESULTS: Using VigiBase, we identified GCA as a significant immune-related adverse event associated with anti-CTLA-4 (cytotoxic T-lymphocyte-associated protein-4) but not anti-PD-1 (anti-programmed death-1) nor anti-PD-L1 (anti-programmed death-ligand 1) treatment. We further dissected a critical role for the CTLA-4 pathway in GCA by identification of the dysregulation of CTLA-4-derived gene pathways and proteins in CD4+ (cluster of differentiation 4) T cells (and specifically regulatory T cells) present in blood and aorta of GCA patients versus controls. While regulatory T cells were less abundant and activated/suppressive in blood and aorta of GCA versus controls, they still specifically upregulated CTLA-4. Activated and proliferating CTLA-4+ Ki-67+ regulatory T cells from GCA were more sensitive to anti-CTLA-4 (ipilimumab)-mediated in vitro depletion versus controls. CONCLUSIONS: We highlighted the instrumental role of CTLA-4 immune checkpoint in GCA, which provides a strong rationale for targeting this pathway.


Subject(s)
CTLA-4 Antigen , Giant Cell Arteritis , Humans , Aorta , Immune Checkpoint Inhibitors , Leukocytes, Mononuclear , T-Lymphocytes, Regulatory , CTLA-4 Antigen/metabolism
3.
Int J Mol Sci ; 23(23)2022 Nov 29.
Article in English | MEDLINE | ID: mdl-36499300

ABSTRACT

We previously reported that a novel peptide vaccine platform, based on synthetic melanin nanoaggregates, triggers strong cytotoxic immune responses and significantly suppresses tumor growth in mice. However, the mechanisms underlying such an efficacy remained poorly described. Herein, we investigated the role of dendritic cells (DCs) in presenting the antigen embedded in the vaccine formulation, as well as the potential stimulatory effect of melanin upon these cells, in vitro by coculture experiments and ELISA/flow cytometry analysis. The vaccine efficiency was evaluated in FLT3-L-/- mice constitutively deficient in DC1, DC2, and pDCs, in Zbtb46DTR chimera mice deficient in DC1 and DC2, and in LangerinDTR mice deficient in dermal DC1 and Langerhans cells. We concluded that DCs, and especially migratory conventional type 1 dendritic cells, seem crucial for mounting the immune response after melanin-based vaccination. We also assessed the protective effect of L-DOPA melanin on peptides from enzymatic digestion, as well as the biodistribution of melanin-peptide nanoaggregates, after subcutaneous injection using [18F]MEL050 PET imaging in mice. L-DOPA melanin proved to act as an efficient carrier for peptides by fully protecting them from enzymatic degradation. L-DOPA melanin did not display any direct stimulatory effects on dendritic cells in vitro. Using PET imaging, we detected melanin-peptide nanoaggregates up to three weeks after subcutaneous injections within the secondary lymphoid tissues, which could explain the sustained immune response observed (up to 4 months) with this vaccine technology.


Subject(s)
Cancer Vaccines , Neoplasms , Mice , Animals , Melanins/metabolism , Tissue Distribution , Dendritic Cells , Peptides/pharmacology , Mice, Inbred C57BL , Neoplasms/metabolism
4.
Nat Immunol ; 9(6): 676-83, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18469816

ABSTRACT

Dendritic cell (DC) development begins in the bone marrow but is not completed until after immature progenitors reach their sites of residence in lymphoid organs. The hematopoietic growth factors regulating these processes are poorly understood. Here we examined the effects of signaling by the receptor tyrosine kinase Flt3 on macrophage DC progenitors in the bone marrow and on peripheral DCs. We found that the macrophage DC progenitor compartment was responsive to superphysiological amounts of Flt3 ligand but was not dependent on Flt3 for its homeostatic maintenance in vivo. In contrast, Flt3 was essential to the regulation of homeostatic DC development in the spleen, where it was needed to maintain normal numbers of DCs by controlling their division in the periphery.


Subject(s)
Bone Marrow Cells/cytology , Dendritic Cells/immunology , Lymphoid Tissue/cytology , Membrane Proteins/physiology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Clonal Anergy/immunology , Dendritic Cells/cytology , Lymphoid Tissue/immunology , Membrane Proteins/metabolism , Mice
5.
J Immunol ; 191(5): 2273-81, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23913969

ABSTRACT

Regulatory T cells (Tregs) play crucial roles in both fetal and tumor development. We recently showed that immunosurveillance by pre-existing CD44(high)CD62L(low) activated/memory Tregs (amTregs) specific for self-Ags protects emergent tumor cells in mice. This Treg response of a memory type is more rapid than and dominates the antitumor response of tumor-specific effector T cells. In this study, we report striking similarities between the early Treg responses to embryo and tumor implantation. Tregs are rapidly recruited to uterus-draining lymph nodes and activated in the first days after embryo implantation in both syngeneic and allogeneic matings; express the markers of the amTreg subset; and are at least in part self-Ag specific, as seen in tumor emergence. Unlike in the tumor emergence setting, however, for which preimmunization against tumor Ags is sufficient for complete tumor eradication even in the presence of Tregs, Treg depletion is additionally required for high frequencies of fetus loss after preimmunization against paternal tissue Ags. Thus, amTregs play a major role in protecting embryos in both naive and preimmune settings. This role and the ensuing therapeutic potential are further highlighted by showing that Treg stimulation, directly by low-dose IL-2 or indirectly by Fms-related tyrosine kinase 3 ligand, led to normal pregnancy rates in a spontaneous abortion-prone model.


Subject(s)
Embryo Implantation/immunology , Embryo, Mammalian/immunology , Immune Tolerance/immunology , Immunologic Memory/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Female , Flow Cytometry , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
6.
Cell Rep Med ; 4(12): 101256, 2023 12 19.
Article in English | MEDLINE | ID: mdl-38118422

ABSTRACT

FLT3-L-dependent classical dendritic cells (cDCs) recruit anti-tumor and tumor-protecting lymphocytes. We evaluate cancer growth in mice with low, normal, or high levels of cDCs. Paradoxically, both low or high numbers of cDCs improve survival in mice with melanoma. In low cDC context, tumors are restrained by the adaptive immune system through influx of effector T cells and depletion of Tregs and NK cells. High cDC numbers favor the innate anti-tumor response, with massive recruitment of activated NK cells, despite high Treg infiltration. Anti CTLA-4 but not anti PD-1 therapy synergizes with FLT3-L therapy in the cDCHi but not in the cDCLo context. A combination of cDC boost and Treg depletion dramatically improves survival of tumor-bearing mice. Transcriptomic data confirm the paradoxical effect of cDC levels on survival in several human tumor types. cDCHi-TregLo state in such patients predicts best survival. Modulating cDC numbers via FLT3 signaling may have therapeutic potential in human cancer.


Subject(s)
Neoplasms , T-Lymphocytes, Regulatory , Humans , Mice , Animals , Killer Cells, Natural , Dendritic Cells , Homeostasis
7.
Front Immunol ; 14: 1252979, 2023.
Article in English | MEDLINE | ID: mdl-37876927

ABSTRACT

Background: Crohn's disease (CD) is a complex and poorly understood myeloid-mediated disorder. Genetic variants with loss of function in the NOD2 gene confer an increased susceptibility to ileal CD. While Nod2 in myeloid cells may confer protection against T-cell mediated ileopathy, it remains unclear whether it may promote resolution of the inflamed colon. In this study, we evaluated the function of Nod2 in myeloid cells in a model of acute colitis and colitis-associated colon cancer (CAC). Methods: To ablate Nod2 specifically within the myeloid compartment, we generated LysMCre/+;Nod2fl/fl mice. The role of NOD2 was studied in a setting of Dextran Sodium Sulfate (DSS)-induced colitis and in azoxymethane (AOM)/DSS model. Clinical parameters were quantified by colonoscopy, histological, flow cytometry, and qRT-PCR analysis. Results: Upon DSS colitis model, LysMCre/+;Nod2fl/fl mice lost less weight than control littermates and had less severe damage to the colonic epithelium. In the AOM/DSS model, endoscopic monitoring of tumor progression revealed a lowered number of adenomas within the colon of LysMCre/+;Nod2fl/fl mice, associated with less expression of Tgfb. Mechanistically, lysozyme M was required for the improved disease severity in mice with a defect of NOD2 in myeloid cells. Conclusion: Our results indicate that loss of Nod2 signaling in myeloid cells aids in the tissue repair of the inflamed large intestine through lysozyme secretion by myeloid cells. These results may pave the way to design new therapeutics to limit the inflammatory and tumorigenic functions of NOD2.


Subject(s)
Colitis , Crohn Disease , Macrophages , Nod2 Signaling Adaptor Protein , Animals , Mice , Azoxymethane , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Macrophages/metabolism , Muramidase/genetics , Nod2 Signaling Adaptor Protein/genetics
8.
J Exp Med ; 198(5): 737-46, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12939344

ABSTRACT

Despite a growing interest in CD4+ CD25+ regulatory T cells (Treg) that play a major role in self-tolerance and immunoregulation, fundamental parameters of the biology and homeostasis of these cells are poorly known. Here, we show that this population is composed of two Treg subsets that have distinct phenotypes and homeostasis in normal unmanipulated mice. In the steady state, some Treg remain quiescent and have a long lifespan, in the order of months, whereas the other Treg are dividing extensively and express multiple activation markers. After adoptive transfer, tissue-specific Treg rapidly divide and expand preferentially in lymph nodes draining their target self-antigens. These results reveal the existence of a cycling Treg subset composed of autoreactive Treg that are continuously activated by tissue self-antigens.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Receptors, Antigen, T-Cell/immunology , Receptors, Interleukin-2/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/cytology , Cell Survival , Crosses, Genetic , Female , Immune Tolerance , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Models, Immunological , Receptors, Antigen, T-Cell/genetics , Time Factors
9.
Nat Commun ; 8(1): 1640, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29158474

ABSTRACT

Dendritic cells (DC) have the unique ability to present exogenous antigens via the major histocompatibility complex class I pathway to stimulate naive CD8+ T cells. In DCs with a non-functional mutation in Unc93b1 (3d mutation), endosomal acidification, phagosomal maturation, antigen degradation, antigen export to the cytosol and the function of the store-operated-Ca2+-entry regulator STIM1 are impaired. These defects result in compromised antigen cross-presentation and anti-tumor responses in 3d-mutated mice. Here, we show that UNC93B1 interacts with the calcium sensor STIM1 in the endoplasmic reticulum, a critical step for STIM1 oligomerization and activation. Expression of a constitutively active STIM1 mutant, which no longer binds UNC93B1, restores antigen degradation and cross-presentation in 3d-mutated DCs. Furthermore, ablation of STIM1 in mouse and human cells leads to a decrease in cross-presentation. Our data indicate that the UNC93B1 and STIM1 cooperation is important for calcium flux and antigen cross-presentation in DCs.


Subject(s)
Antigen Presentation , Calcium/metabolism , Dendritic Cells/immunology , Membrane Transport Proteins/metabolism , Stromal Interaction Molecule 1/metabolism , Animals , Antigens/immunology , Antigens/metabolism , Cells, Cultured , Cross-Priming , Dendritic Cells/metabolism , Endoplasmic Reticulum/metabolism , Female , Humans , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/immunology , Mice , Mice, Inbred C57BL , Protein Binding , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/immunology
11.
Immunol Lett ; 102(1): 106-9, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16199095

ABSTRACT

Recent evidences indicate that naturally occurring CD4+CD25+ regulatory/suppressor T cells (T(reg)) regulate not only autoimmunity, but also alloreactivity. In mice, they notably control tolerance to allogeneic transplants and graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Here, we studied the role of T(reg) in maternal tolerance to fetuses, i.e. natural semi-allogeneic grafts. We show that semi-allogeneic pregnancies in mice induce an expansion of T(reg), but not of activated CD4+ and CD8+ T cells, in para-aortic lymph nodes draining fetal antigens. The treatment of female mice with an anti-CD25 antibody before mating results in depletion of T(reg) and expansion of activated CD4+ and CD8+ T cells solely in the draining lymph nodes, ultimately leading to fetus rejection. These observations were not made in the context of syngeneic pregnancies. Thus, T(reg) play a major role in maternal-fetal tolerance.


Subject(s)
Embryo Loss/immunology , Embryo Loss/prevention & control , Receptors, Interleukin-2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Female , Lymph Nodes/immunology , Male , Mice , Pregnancy
13.
Cold Spring Harb Protoc ; 2016(2): pdb.prot078345, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26832681

ABSTRACT

The mammary gland is an important model system in metastasis research. Mammary epithelial stem cells are of particular interest because of their capacity for regeneration and their role in cancer initiation. This protocol describes how to enrich for mammary basal and luminal epithelial cells using fluorescence-activated cell sorting (FACS).


Subject(s)
Epithelial Cells/physiology , Flow Cytometry/methods , Mammary Neoplasms, Animal/pathology , Neoplasm Metastasis/pathology , Neoplasm Metastasis/physiopathology , Animals , Mice
14.
Cold Spring Harb Protoc ; 2016(2): pdb.top069948, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26832689

ABSTRACT

Metastasis is often modeled by xenotransplantation of cell lines in immunodeficient mice. A wealth of information about tumor cell behavior in the new environment is obtained from these efforts. Yet by design, this approach is "tumor-centric," as it focuses on cell-autonomous determinants of human tumor dissemination in mouse tissues, in effect using the animal body as a sophisticated "Petri dish" providing nutrients and support for tumor growth. Transgenic or gene knockout mouse models of cancer allow the study of tumor spread as a systemic disease and offer a complimentary approach for studying the natural history of cancer. This introduction is aimed at describing the overall methodological approach to studying metastasis in genetically modified mice, with a particular focus on using animals with regulated expression of potent human oncogenes in the breast.


Subject(s)
Mice, Transgenic , Neoplasm Metastasis/pathology , Animals , Mice
15.
Methods Mol Biol ; 1267: 381-94, 2015.
Article in English | MEDLINE | ID: mdl-25636480

ABSTRACT

The studies of oncogene dependence are aimed to understand an unfortunate and puzzling aspect of targeted anticancer treatments-their progression to drug resistance. Drug resistance develops from a pool of cells that survive the original treatment, called minimal residual disease. Mouse models based on tetracycline-dependent expression of transgenic oncogenes are used to imitate targeted oncogene blockade and to reproduce minimal residual disease in humans. Here we describe a novel method for generating oncogene-dependent mammary tumors using somatic transfer of transactivator-containing retroviruses into transgenic mice with tetracycline-dependent oncogenes and a method for measuring continuous mitotic activity in epithelial cells in real time.


Subject(s)
Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Oncogenes , Animals , Cell Proliferation , Cell Transformation, Neoplastic , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Flow Cytometry , Fluoresceins/metabolism , Injections , Mammary Glands, Animal/pathology , Mice , Neoplasm, Residual , Staining and Labeling , Succinimides/metabolism , Time Factors , Transduction, Genetic
16.
Front Immunol ; 4: 292, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24133490

ABSTRACT

The influence of CD4(+)CD25(+)Foxp3(+) regulatory T-cells (Tregs) on cancer progression has been demonstrated in a large number of preclinical models and confirmed in several types of malignancies. Neoplastic processes trigger an increase of Treg numbers in draining lymph nodes, spleen, blood, and tumors, leading to the suppression of anti-tumor responses. Treg-depletion before or early in tumor development may lead to complete tumor eradication and extends survival of mice and humans. However this strategy is ineffective in established tumors, highlighting the critical role of the early Treg-tumor encounters. In this review, after discussing old and new concepts of immunological tumor tolerance, we focus on the nature (thymus-derived vs. peripherally derived) and status (naïve or activated/memory) of the regulatory T-cells at tumor emergence. The recent discoveries in this field suggest that the activation status of Tregs and effector T-cells (Teffs) at the first encounter with the tumor are essential to shape the fate and speed of the immune response across a variety of tumor models. The relative timing of activation/recruitment of anti-tumor cells vs. tolerogenic cells at tumor emergence appears to be crucial in the identification of tumor cells as friend or foe, which has broad implications for the design of cancer immunotherapies.

17.
J Exp Med ; 209(6): 1153-65, 2012 Jun 04.
Article in English | MEDLINE | ID: mdl-22615130

ABSTRACT

Classical dendritic cells (cDCs), monocytes, and plasmacytoid DCs (pDCs) arise from a common bone marrow precursor (macrophage and DC progenitors [MDPs]) and express many of the same surface markers, including CD11c. We describe a previously uncharacterized zinc finger transcription factor, zDC (Zbtb46, Btbd4), which is specifically expressed by cDCs and committed cDC precursors but not by monocytes, pDCs, or other immune cell populations. We inserted diphtheria toxin (DT) receptor (DTR) cDNA into the 3' UTR of the zDC locus to serve as an indicator of zDC expression and as a means to specifically deplete cDCs. Mice bearing this knockin express DTR in cDCs but not other immune cell populations, and DT injection into zDC-DTR bone marrow chimeras results in cDC depletion. In contrast to previously characterized CD11c-DTR mice, non-cDCs, including pDCs, monocytes, macrophages, and NK cells, were spared after DT injection in zDC-DTR mice. We compared immune responses to Toxoplasma gondii and MO4 melanoma in DT-treated zDC- and CD11c-DTR mice and found that immunity was only partially impaired in zDC-DTR mice. Our results indicate that CD11c-expressing non-cDCs make significant contributions to initiating immunity to parasites and tumors.


Subject(s)
Cell Lineage/physiology , Dendritic Cells/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , 3' Untranslated Regions , Animals , CD11c Antigen/genetics , CD11c Antigen/immunology , CD11c Antigen/metabolism , Dendritic Cells/metabolism , Diphtheria Toxin/pharmacology , Gene Expression Regulation , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/drug effects , Killer Cells, Natural/physiology , Macrophages/drug effects , Macrophages/physiology , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/physiology , Toxoplasma/pathogenicity , Toxoplasmosis/immunology , Transcription Factors/immunology , Zinc Fingers
18.
J Exp Med ; 209(5): 1011-28, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22547652

ABSTRACT

Delta-like ligand 4 (Dll4)-Notch signaling is essential for T cell development and alternative thymic lineage decisions. How Dll4-Notch signaling affects pro-T cell fate and thymic dendritic cell (tDC) development is unknown. We found that Dll4 pharmacological blockade induces accumulation of tDCs and CD4(+)CD25(+)FoxP3(+) regulatory T cells (T(reg) cells) in the thymic cortex. Both genetic inactivation models and anti-Dll4 antibody (Ab) treatment promote de novo natural T(reg) cell expansion by a DC-dependent mechanism that requires major histocompatibility complex II expression on DCs. Anti-Dll4 treatment converts CD4(-)CD8(-)c-kit(+)CD44(+)CD25(-) (DN1) T cell progenitors to immature DCs that induce ex vivo differentiation of naive CD4(+) T cells into T(reg) cells. Induction of these tolerogenic DN1-derived tDCs and the ensuing expansion of T(reg) cells are Fms-like tyrosine kinase 3 (Flt3) independent, occur in the context of transcriptional up-regulation of PU.1, Irf-4, Irf-8, and CSF-1, genes critical for DC differentiation, and are abrogated in thymectomized mice. Anti-Dll4 treatment fully prevents type 1 diabetes (T1D) via a T(reg) cell-mediated mechanism and inhibits CD8(+) T cell pancreatic islet infiltration. Furthermore, a single injection of anti-Dll4 Ab reverses established T1D. Disease remission and recurrence are correlated with increased T(reg) cell numbers in the pancreas-draining lymph nodes. These results identify Dll4-Notch as a novel Flt3-alternative pathway important for regulating tDC-mediated T(reg) cell homeostasis and autoimmunity.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/physiology , Diabetes Mellitus, Type 1/prevention & control , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/metabolism , Animals , Antibodies/pharmacology , Blotting, Western , Calcium-Binding Proteins , DNA Primers/genetics , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Genes, MHC Class II/immunology , Intracellular Signaling Peptides and Proteins/immunology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/metabolism , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Pancreas/pathology , Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/cytology , Thymus Gland/immunology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
19.
Autoimmunity ; 44(6): 471-82, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21370936

ABSTRACT

Induction of immune tolerance as therapeutic approach for autoimmune diseases constitutes a current research focal point. In this sense, we aimed to evaluate an altered peptide ligand (APL) for induction of peripheral tolerance in patients with rheumatoid arthritis (RA). A novel T-cell epitope from human heat-shock protein 60 (Hsp60), an autoantigen involved in the pathogenesis of RA, was identified by bioinformatics tools and an APL was design starting from this epitope. We investigated the ability of this APL for inducing regulatory T cells (Treg cells) in mice and evaluated the therapeutic effect of this peptide in an adjuvant-induced arthritis (AA) rat model. Clinical score, TNFα levels and histopathology were monitored, as well as the capacity of this APL for inducing Treg cells. Finally, the potentialities of the APL for inducing Treg cells were evaluated in ex vivo assays using mononuclear cells isolated from peripheral blood (PBMC). The APL induced an increase of the proportions of Treg cells in the draining lymph nodes of the injected site in mice. The APL efficiently inhibited the course of AA, with significant reduction of the clinical and histopathology score. This effect was associated with an increase of the proportions of Treg cells and a decrease of TNFα levels in spleen. Finally, stimulation of PBMCs from RA patients by the APL increases the proportions of the CD4(+)CD25(high)FoxP3(+) Treg cells. These results indicate a therapeutic potentiality of APL and support further investigation of this candidate drug for treatment of RA.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Chaperonin 60/immunology , Epitopes, T-Lymphocyte/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , Aged , Animals , Chaperonin 60/therapeutic use , Female , Humans , Immune Tolerance , Ligands , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mice , Middle Aged , Peptides/immunology , Rats , Rats, Inbred Lew , T-Lymphocytes, Regulatory/metabolism
20.
J Clin Invest ; 119(9): 2648-62, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19652360

ABSTRACT

Early responses of Tregs and effector T cells (Teffs) to their first encounter with tumor cells have been poorly characterized. Here we have shown, in both implanted and in situ-induced mouse tumor models, that the appearance of tumor cells is immediately sensed by CD44hi memory Tregs that are specific for self antigens. The rapid response of these Tregs preceded and prevented activation of naive antitumor Teffs. The relative speed of the Treg versus the Teff response within the first 2-4 days determined the outcome of the antitumor immune response: tolerance or rejection. If antitumor memory Teffs were present at the time of tumor emergence, both Tregs and Teffs were recruited and activated with memory kinetics; however, the Tregs were unable to control the Teffs, which eradicated the tumor cells. This balance between effector and regulatory responses did not depend on the number of Tregs and Teffs, but rather on their memory status. Thus, in the natural setting, dominant tolerogenic immunosurveillance by self-specific memory Tregs protects tumors, just as it protects normal tissues. More generally, our results reveal that the timing of Treg and Teff engagement, determined by their memory status, is an important mode of regulation of immune responses.


Subject(s)
Hyaluronan Receptors/metabolism , Neoplasms, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antigens, Polyomavirus Transforming/genetics , Base Sequence , Cell Line, Tumor , DNA, Complementary/genetics , Female , Immunologic Memory , Mammary Neoplasms, Experimental/immunology , Melanoma, Experimental/immunology , Mesothelioma/immunology , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Models, Immunological , Neoplasms, Experimental/etiology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Oncogenes , Self Tolerance , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/classification , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL