Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Immunol ; 207(2): 421-435, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34233909

ABSTRACT

Intracellular ion fluxes emerge as critical actors of immunoregulation but still remain poorly explored. In this study, we investigated the role of the redundant cation channels TMEM176A and TMEM176B (TMEM176A/B) in retinoic acid-related orphan receptor γt+ cells and conventional dendritic cells (DCs) using germline and conditional double knockout mice. Although Tmem176a/b appeared surprisingly dispensable for the protective function of Th17 and group 3 innate lymphoid cells in the intestinal mucosa, we found that they were required in conventional DCs for optimal Ag processing and presentation to CD4+ T cells. Using a real-time imaging method, we show that TMEM176A/B accumulate in dynamic post-Golgi vesicles preferentially linked to the late endolysosomal system and strongly colocalize with HLA-DM. Taken together, our results suggest that TMEM176A/B ion channels play a direct role in the MHC class II compartment of DCs for the fine regulation of Ag presentation and naive CD4+ T cell priming.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Membrane Proteins/immunology , Animals , Endosomes/immunology , Female , Genes, MHC Class II/immunology , Golgi Apparatus/immunology , Immunity, Innate/immunology , Intestinal Mucosa/immunology , Ion Channels/immunology , Lymphocytes/immunology , Lysosomes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Th17 Cells/immunology , Tretinoin/immunology
2.
Cancer Sci ; 112(5): 1723-1734, 2021 May.
Article in English | MEDLINE | ID: mdl-33609296

ABSTRACT

T cells could be engineered to overcome the aberrant metabolic milieu of solid tumors and tip the balance in favor of a long-lasting clinical response. Here, we explored the therapeutic potential of stably overexpressing cystathionine-gamma-lyase (CTH, CSE, or cystathionase), a pivotal enzyme of the transsulfuration pathway, in antitumor CD8+ T cells with the initial aim to boost intrinsic cysteine metabolism. Using a mouse model of adoptive cell transfer (ACT), we found that CTH-expressing T cells showed a superior control of tumor growth compared to control T cells. However, contrary to our hypothesis, this effect was not associated with increased T cell expansion in vivo or proliferation rescue in the absence of cysteine/cystine in vitro. Rather than impacting methionine or cysteine, ACT with CTH overexpression unexpectedly reduced glycine, serine, and proline concentration within the tumor interstitial fluid. Interestingly, in vitro tumor cell growth was mostly impacted by the combination of serine/proline or serine/glycine deprivation. These results suggest that metabolic gene engineering of T cells could be further investigated to locally modulate amino acid availability within the tumor environment while avoiding systemic toxicity.


Subject(s)
Adoptive Transfer/methods , CD8-Positive T-Lymphocytes/metabolism , Cystathionine gamma-Lyase/metabolism , Cysteine/biosynthesis , Animals , Cell Engineering , Cell Line, Tumor , Cell Proliferation , Extracellular Fluid/metabolism , Female , Glycine/metabolism , Methionine/metabolism , Mice , Mice, Inbred C57BL , Models, Animal , Ovarian Neoplasms/therapy , Proline/metabolism , Serine/metabolism , Tumor Microenvironment/immunology
3.
J Immunol ; 198(9): 3671-3678, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28356382

ABSTRACT

Psoriasis is a chronic inflammatory disease resulting from dysregulated immune activation associated with a large local secretion of cytokines. Among them, IL-22 largely contributes to epithelial remodeling and inflammation through inhibiting the terminal differentiation of keratinocytes and inducing antimicrobial peptides and selected chemokines. The activity of IL-22 is regulated by IL-22 binding protein (IL-22BP); however, the expression and role of IL-22BP in psoriatic skin has remained unknown so far. Here we showed that nonaffected skin of psoriasis patients displayed lower expression of IL-22BP than skin of healthy controls. Furthermore, the strong IL-22 increase in lesional psoriatic skin was accompanied by a moderate induction of IL-22BP. To investigate the role of IL-22BP in controlling IL-22 during skin inflammation, we used imiquimod-induced skin disease in rodents and showed that rats with genetic IL-22BP deficiency (Il22ra2-/-) displayed exacerbated disease that associated with enhanced expression of IL-22-inducible antimicrobial peptides. We further recapitulated these findings in mice injected with an anti-IL-22BP neutralizing Ab. Hypothesizing that the IL-22/IL-22BP expression ratio reflects the level of bioactive IL-22 in psoriasis skin, we found positive correlations with the expression of IL-22-inducible molecules (IL-20, IL-24, IL-36γ, CXCL1, and BD2) in keratinocytes. Finally, we observed that serum IL-22/IL-22BP protein ratio strongly correlated with psoriasis severity. In conclusion, we propose that although IL-22BP can control deleterious actions of IL-22 in the skin, its limited production prevents a sufficient neutralization of IL-22 and contributes to the development and maintenance of epidermal alterations in psoriasis.


Subject(s)
Inflammation/immunology , Interleukins/metabolism , Keratinocytes/metabolism , Psoriasis/immunology , Receptors, Interleukin/metabolism , Skin/immunology , Adult , Aged , Aminoquinolines , Animals , Antibodies, Blocking/administration & dosage , Cells, Cultured , Female , Gene Knockout Techniques , Humans , Imiquimod , Keratinocytes/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Psoriasis/chemically induced , Rats , Rats, Sprague-Dawley , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Signal Transduction , Young Adult , Interleukin-22
4.
J Immunol ; 196(9): 3716-28, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27016604

ABSTRACT

Langerhans cells (LCs) are epithelial APCs that sense danger signals and in turn trigger specific immune responses. In steady-state, they participate in the maintenance of peripheral tolerance to self-antigens whereas under inflammation LCs efficiently trigger immune responses in secondary lymphoid organs. It has been demonstrated in mice that LC-deprived epithelia are rapidly replenished by short half-life langerin-expressing monocyte-derived LCs (MDLCs). These surrogate LCs are thought to be progressively replaced by langerin(high) LCs arising from self-renewing epithelial precursors of hematopoietic origin. How LCs arise from blood monocytes is not fully understood. Hence, we sought to characterize key factors that induce differentiation of langerin(high)-expressing monocyte-derived Langerhans-like cells. We identified GM-CSF and TGF-ß1 as key cytokines to generate langerin(high)-expressing cells but only in serum-free conditions. These cells were shown to express the LC-specific TROP-2 and Axl surface markers and contained Birbeck granules. Surprisingly, E-cadherin was not spontaneously expressed by these cells but required a direct contact with keratinocytes to be stably induced. MDLCs induced stronger allogeneic T cell proliferations but released low amounts of inflammatory cytokines upon TLR stimulation compared with donor-paired monocyte-derived dendritic cells. Immature langerin(high) MDLCs were responsive to MIP-3ß/CCL20 and CTAC/CCL27 chemokine stimulations. Finally, we demonstrated that those cells behaved as bona fide LCs when inserted in a three-dimensional rebuilt epithelium by becoming activated upon TLR or UV light stimulations. Collectively, these results prompt us to propose these langerin(high) MDLCs as a relevant model to address LC biology-related questions.


Subject(s)
Blood Cells/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Keratinocytes/physiology , Langerhans Cells/immunology , Monocytes/physiology , T-Lymphocytes/immunology , Transforming Growth Factor beta1/metabolism , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cells, Cultured , Humans , Isoantigens/immunology , Lipopolysaccharide Receptors/metabolism , Lymphocyte Activation , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, IgG/metabolism , Self Tolerance , Ultraviolet Rays , Axl Receptor Tyrosine Kinase
6.
J Immunol ; 186(6): 3317-26, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21300823

ABSTRACT

Studying the activity of homogeneous regulatory T cell (Treg) populations will advance our understanding of their mechanisms of action and their role in human disease. Although isolating human Tregs exhibiting low expression of CD127 markedly increases purity, the resulting Treg populations are still heterogeneous. To examine the complexity of the Tregs defined by the CD127 phenotype in comparison with the previously described CD4(+)CD25(hi) subpopulations, we subdivided the CD25(hi) population of memory Tregs into subsets based on expression of CD127 and HLA-DR. These subsets exhibited differences in suppressive capacity, ability to secrete IL-10 and IL-17, Foxp3 gene methylation, cellular senescence, and frequency in neonatal and adult blood. The mature, short telomere, effector CD127(lo)HLA-DR(+) cells most strongly suppressed effector T cells within 48 h, whereas the less mature CD127(lo)HLA-DR(-) cells required 96 h to reach full suppressive capacity. In contrast, whereas the CD127(+)HLA-DR(-) cells also suppressed proliferation of effector cells, they could alternate between suppression or secretion of IL-17 depending upon the stimulation signals. When isolated from patients with multiple sclerosis, both the nonmature and the effector subsets of memory CD127(lo) Tregs exhibited kinetically distinct defects in suppression that were evident with CD2 costimulation. These data demonstrate that natural and not induced Tregs are less suppressive in patients with multiple sclerosis.


Subject(s)
CD2 Antigens/physiology , CD4 Antigens/biosynthesis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Adult , Cell Differentiation/immunology , Cells, Cultured , Coculture Techniques , Fetal Blood/cytology , Fetal Blood/immunology , Fetal Blood/metabolism , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/pharmacokinetics , Humans , Infant, Newborn , Interleukin-7 Receptor alpha Subunit/genetics , Interleukin-7 Receptor alpha Subunit/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Middle Aged , Multiple Sclerosis/metabolism , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/metabolism , Young Adult
7.
J Immunol ; 184(2): 685-93, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20018615

ABSTRACT

Functional studies of human primary immune cells have been hampered by the lack of tools to silence gene functions. In this study, we report the application of a lentiviral RNA interference library in primary human T cells. Using a subgenomic short hair RNA library targeting approximately 1000 signaling genes, we identified novel genes that control the levels of IL-10 produced. IL-10 is a potent anti-inflammatory cytokine secreted by several cell types, including T regulatory type 1 cells, a subset of T regulatory cells that exert their suppressive activity through IL-10 secretion. FLT3, a known hematopoeitic growth factor, was found to be a negative regulator of IL-10 levels in activated T cells. This was based on several observations. First, FLT3 and its ligand (FL) were both induced by T cell activation. Second, silencing of FLT3 led to increased IL-10 levels, whereas addition of FL suppressed IL-10 secretion and increased FLT3 surface levels. Third, engagement of CD46, a known inducer of T regulatory type 1 cells, upregulated surface FLT3, and secreted FL, which then inhibited IL-10 production by T cells. Hence, FL and FLT3 form a novel regulatory feedback loop that limits IL-10 production in T cells. Our results identified FLT3 as a new regulator of T cell function and offer a strategy to genetically dissect specific pathways in T cells.


Subject(s)
Interleukin-10/metabolism , RNA, Small Interfering/pharmacology , T-Lymphocytes/immunology , fms-Like Tyrosine Kinase 3/immunology , Cells, Cultured , Feedback, Physiological , Gene Expression Regulation , Gene Library , Humans , Immunologic Factors , Interleukin-10/genetics , Lentivirus/genetics , Ligands , Lymphocyte Activation , Membrane Cofactor Protein/metabolism , RNA Interference/immunology
8.
J Immunol ; 185(1): 46-54, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20498357

ABSTRACT

The secretion of IL-9, initially recognized as a Th2 cytokine, was recently attributed to a novel CD4 T cell subset termed Th9 in the murine system. However, IL-9 can also be secreted by mouse Th17 cells and may mediate aspects of the proinflammatory activities of Th17 cells. Here we report that IL-9 is secreted by human naive CD4 T cells in response to differentiation by Th9 (TGF-beta and IL-4) or Th17 polarizing conditions. Yet, these differentiated naive cells did not coexpress IL-17 and IL-9, unless they were repeatedly stimulated under Th17 differentiation-inducing conditions. In contrast to the naive cells, memory CD4 T cells were induced to secrete IL-9 by simply providing TGF-beta during stimulation, as neither IL-4 nor proinflammatory cytokines were required. Furthermore, the addition of TGF-beta to the Th17-inducing cytokines (IL-1beta, IL-6, IL-21, IL-23) that induce memory cells to secrete IL-17, resulted in the marked coexpression of IL-9 in IL-17 producing memory cells. The proinflammatory cytokine mediating TGF-beta-dependent coexpression of IL-9 and IL-17 was identified to be IL-1beta. Moreover, circulating monocytes were potent costimulators of IL-9 production by Th17 cells via their capacity to secrete IL-1beta. Finally, to determine whether IL-9/IL-17 coproducing CD4 cells were altered in an inflammatory condition, we examined patients with autoimmune diabetes and demonstrated that these subjects exhibit a higher frequency of memory CD4 cells with the capacity to transition into IL-9(+)IL-17(+) cells. These data demonstrate the presence of IL-17(+)IL-9(+) CD4 cells induced by IL-1beta that may play a role in human autoimmune disease.


Subject(s)
Interleukin-17/biosynthesis , Interleukin-9/biosynthesis , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Transforming Growth Factor beta1/physiology , Adult , Cell Polarity/immunology , Cells, Cultured , Coculture Techniques , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Gene Expression Regulation/immunology , Humans , Immunohistochemistry , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Interleukin-9/genetics , Interleukin-9/metabolism , Middle Aged , Resting Phase, Cell Cycle/immunology , Young Adult
9.
Curr Opin Organ Transplant ; 17(1): 42-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22227722

ABSTRACT

PURPOSE OF REVIEW: We discuss the use of tolerogenic dendritic cells (TolDCs) as a therapeutic tool in solid organ transplantation, with particular emphasis on recent experimental and preclinical data supporting the clinical translation of TolDC therapy. RECENT FINDINGS: TolDC have been successfully used in rodents to promote long-term allograft survival. Although most studies have focused on donor dendritic cells or donor antigen-pulsed dendritic cells, our group investigated a strategy based on the administration of autologous dendritic cells (not pulsed with donor antigens). We discuss the therapeutic efficacy, mechanisms, and potential risks and advantages of each strategy. We also highlight recent findings on the generation of clinical grade human TolDC from blood monocytes. Finally, we discuss preliminary experience with dendritic cells in humans and critical issues regarding the implementation of TolDC therapy to clinical organ transplantation. SUMMARY: TolDC hold therapeutic promise for the treatment of transplanted patients. Cell therapy with unpulsed, autologous dendritic cells appears as a well tolerated, clinically relevant approach that might help in improving long-term allograft survival and limit the harmful effects of immunosuppressive treatments.


Subject(s)
Dendritic Cells/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival/immunology , Organ Transplantation , Transplantation Tolerance/immunology , Transplantation, Homologous/immunology , Animals , Antigens, CD/immunology , Dendritic Cells/transplantation , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Immunotherapy, Adoptive/methods , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation, Autologous
10.
Front Immunol ; 13: 1034570, 2022.
Article in English | MEDLINE | ID: mdl-36311796

ABSTRACT

Crohn's disease (CD), a form of inflammatory bowel disease (IBD), is characterized by impaired epithelial barrier functions and dysregulated mucosal immune responses. IL-22 binding protein (IL-22BP) is a soluble inhibitor regulating IL-22 bioactivity, a cytokine proposed to play protective roles during CD. We and others have shown that IL-22BP is produced in IBD inflamed tissues, hence suggesting a role in CD. In this work, we extended the characterization of IL-22BP production and distribution in CD tissues by applying enzyme-linked immunosorbent assays to supernatants obtained from the culture of endoscopic biopsies of patients, and reverse transcription-quantitative polymerase chain reaction on sorted immune cell subsets. We reveal that IL-22BP levels are higher in inflamed ileums than colons. We observe that in a cell-intrinsic fashion, populations of mononuclear phagocytes and eosinophils express IL-22BP at the highest levels in comparison to other sources of T cells. We suggest the enrichment of intestinal eosinophils could explain higher IL-22BP levels in the ileum. In inflamed colon, we reveal the presence of increased IL-22/IL22BP ratios compared to controls, and a strong correlation between IL-22BP and CCL24. We identify monocyte-derived dendritic cells (moDC) as a cellular subtype co-expressing both cytokines and validate our finding using in vitro culture systems. We also show that retinoic acid induces the secretion of both IL-22BP and CCL24 by moDC. Finally, we report on higher IL-22BP levels in active smokers. In conclusion, our work provides new information relevant to therapeutic strategies modulating IL-22 bioactivity in CD, especially in the context of disease location.


Subject(s)
Crohn Disease , Inflammatory Bowel Diseases , Humans , Carrier Proteins/metabolism , Colon , Cytokines/metabolism , Intestines/pathology
11.
Blood ; 113(18): 4240-9, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19171879

ABSTRACT

Although implicated in antagonistic functions, both regulatory T cells (Tregs) and Th17 effector cells play an important role in controlling autoimmune pathogenesis. Paradoxically, recent studies indicate that Tregs have the capacity to produce interleukin-17 (IL-17), although the ability of these cells to retain their suppressive function remains unknown. Here we report that human Tregs within the CD4(+)CD45RA(-)CD25(high)CCR6(+)HLA-DR(-)FoxP3(+) population produce IL-17 when activated in the presence of the proinflammatory cytokines IL-1beta and IL-6, whereas IL-17 secretion was inhibited by TGFbeta. To assess the ability of a single Treg to secrete IL-17 and to suppress in vitro immune function, we isolated clones from this population. We found that IL-17(+)/FoxP3(+) Treg clones retain suppressive function and exhibit the plasticity to secrete IL-17 or suppress depending on the nature of the stimulus provided. IL-17 production by these Treg clones was accompanied by sustained FoxP3 expression and concomitant, but reversible, loss of suppressive activity. Our data demonstrate that at the single cell level a subset of in vitro suppressive FoxP3(+) cells can be driven to secrete IL-17 under inflammatory conditions. These findings suggest a new mechanism by which inflammation can drive Tregs to secrete IL-17, thereby dampening suppression and promoting an inflammatory milieu.


Subject(s)
Immune Tolerance/physiology , Interleukin-17/biosynthesis , T-Lymphocytes, Regulatory/immunology , Antigens, CD/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Forkhead Transcription Factors/metabolism , HLA-DR Antigens/metabolism , Humans , Interleukin-17/antagonists & inhibitors , Interleukin-1beta/pharmacology , Interleukin-6/pharmacology , Lymphocyte Activation/drug effects , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transforming Growth Factor beta/pharmacology
12.
Front Immunol ; 11: 255, 2020.
Article in English | MEDLINE | ID: mdl-32140157

ABSTRACT

Innate lymphoid cells (ILCs) are tissue-resident lymphocytes that lack antigen-specific receptors and exhibit innate effector functions such as cytokine production that play an important role in immediate responses to pathogens especially at mucosal sites. Mouse and human ILC subsets have been extensively characterized in various tissues and in blood. In this study, we present the first characterization of ILCs and ILC subsets in rat gut and secondary lymphoid organs using flow cytometry and single cell RNA sequencing. Our results show that phenotype and function of rat ILC subsets are conserved as compared to human and mouse ILCs. However, and in contrast to human and mouse, our study unexpectedly revealed that ILC2 and not ILC3 was the dominant ILC subset in the rat intestinal lamina propria. ILC2 predominance in the gut was independent of rat strain, sex or housing facility. In contrast, ILC3 was the predominant ILC subset in mesenteric lymph nodes and Peyer patches. In conclusion, our study demonstrates that in spite of highly conserved phenotype and function between mice, rat and humans, the distribution of ILC subsets in the intestinal mucosa is dependent on the species likely in response to both genetic and environmental factors.


Subject(s)
Intestinal Mucosa/immunology , Lymphocyte Subsets/immunology , Lymphocytes/immunology , Animals , Cell Count , Cells, Cultured , Cytokines/metabolism , Flow Cytometry , Humans , Immunity, Innate , Male , Mice , Rats , Rats, Sprague-Dawley , Th2 Cells/immunology
13.
Cell Metab ; 30(6): 1075-1090.e8, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31801055

ABSTRACT

Cell therapy is a promising strategy for treating patients suffering from autoimmune or inflammatory diseases or receiving a transplant. Based on our preclinical studies, we have generated human autologous tolerogenic dendritic cells (ATDCs), which are being tested in a first-in-man clinical trial in kidney transplant recipients. Here, we report that ATDCs represent a unique subset of monocyte-derived cells based on phenotypic, transcriptomic, and metabolic analyses. ATDCs are characterized by their suppression of T cell proliferation and their expansion of Tregs through secreted factors. ATDCs produce high levels of lactate that shape T cell responses toward tolerance. Indeed, T cells take up ATDC-secreted lactate, leading to a decrease of their glycolysis. In vivo, ATDCs promote elevated levels of circulating lactate and delay graft-versus-host disease by reducing T cell proliferative capacity. The suppression of T cell immunity through lactate production by ATDCs is a novel mechanism that distinguishes ATDCs from other cell-based immunotherapies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immune Tolerance , Immunosuppression Therapy , Lactic Acid/biosynthesis , Animals , Autoimmune Diseases/therapy , CD4-Positive T-Lymphocytes/cytology , Cells, Cultured , Dendritic Cells/metabolism , Female , Humans , Lymphocyte Activation , Male , Mice , Mice, Inbred NOD , Mice, SCID , Monocytes/immunology
14.
Transplantation ; 85(9): 1351-6, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18475195

ABSTRACT

Regulatory T cells (Treg) have been identified as playing a pivotal role in the control of tolerance and in the suppression of pathologic immune responses in autoimmune diseases, transplantation, and graft-versus-host disease. Treg expanded ex vivo by dendritic cells could be potential reagents to promote antigen-specific tolerance in vivo. However, in vivo studies have been carried out mostly in rodents and will need validation in primates before clinical application. We characterized macaque dendritic cell derived either from bone marrow with and without prior CD34+ cell selection (BMDC), or from CD14+ peripheral blood mononuclear cells (Mo-DC). We demonstrate that with a semi-mature phenotype, BMDC are superior to Mo-DC in their capacity to expand freshly isolated allogeneic macaque CD4+ CD25+ CD127- Foxp3+ Treg in vitro in the presence of interleukin-2. Moreover, the expanded Treg maintain their phenotype and suppressive activity. These data provide a step toward the use of macaque dendritic cell to expand Treg for future preclinical testing.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Dendritic Cells/immunology , Lymphocyte Activation , Monocytes/cytology , Monocytes/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD34/immunology , Immunophenotyping , Interleukin-2 Receptor alpha Subunit/immunology , Lipopolysaccharide Receptors/immunology , Macaca fascicularis , Models, Animal
16.
Methods Mol Biol ; 1371: 197-203, 2016.
Article in English | MEDLINE | ID: mdl-26530802

ABSTRACT

Inflammatory bowel diseases (IBDs) are complex multifactorial disease thought to result from inappropriate immune responses to the gut microbiota, in genetically susceptible individuals, under the influence of environmental factors. Among the different animal models developed to help in understanding IBDs pathophysiological mechanisms as well as to achieve pharmacological preclinical studies, the dextran sulfate sodium (DSS)-induced colitis model is the most widely used because of its simplicity, cost-effectiveness, and similarity with human IBDs. This section provides with a detailed protocol that we validated in our laboratory to perform DSS-induced acute colitis in the Sprague-Dawley (SPD) rat.


Subject(s)
Colitis/chemically induced , Dextran Sulfate/adverse effects , Disease Models, Animal , Animals , Colitis/pathology , Dextran Sulfate/administration & dosage , Rats , Rats, Sprague-Dawley
17.
Sci Rep ; 6: 23682, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27009467

ABSTRACT

Retinoid-related orphan receptor gamma t (RORγt) is a master transcription factor central to type 17 immunity involving cells such as T helper 17, group 3 innate lymphoid cells or IL-17-producing γδ T cells. Here we show that the intracellular ion channel TMEM176B and its homologue TMEM176A are strongly expressed in these RORγt(+) cells. We demonstrate that TMEM176A and B exhibit a similar cation channel activity and mainly colocalise in close proximity to the trans-Golgi network. Strikingly, in the mouse, the loss of Tmem176b is systematically associated with a strong upregulation of Tmem176a. While Tmem176b single-deficiency has no effect on the course of experimental autoimmune encephalomyelitis, T cell or DSS-induced colitis, it significantly reduces imiquimod-induced psoriasis-like skin inflammation. These findings shed light on a potentially novel specific process linked to post-Golgi trafficking for modulating the function of RORγt(+) cells and indicate that both homologues should be simultaneously targeted to clearly elucidate the role of this intracellular ion flow.


Subject(s)
Membrane Proteins/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Lymphocytes, Helper-Inducer/cytology , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Humans , Membrane Proteins/metabolism , Mice , Psoriasis/chemically induced , Psoriasis/genetics , T-Lymphocytes, Helper-Inducer/metabolism , trans-Golgi Network/genetics , trans-Golgi Network/metabolism
18.
Transplantation ; 79(8): 969-72, 2005 Apr 27.
Article in English | MEDLINE | ID: mdl-15849552

ABSTRACT

We recently showed that injection of recipient-type immature bone marrow-derived dendritic cells (iBMDCs) the day before transplantation induced a significant prolongation of allograft survival. This study aimed at improving the administration protocol to induce allograft tolerance. Various amounts of iBMDCs were administered to syngeneic LEW.1A rats before and after transplantation of an allogeneic LEW.1W heart, with or without additional suboptimal immunosuppression. Allograft survival was not improved by repeated injections of syngeneic iBMDCs or by additional treatment with low-dose rapamycin. Combining injections of iBMDCs and LF 15-0195 showed a striking synergistic effect and induced definitive allograft acceptance in 92% of recipients. Tolerant recipients accepted donor-type, but not third-party type skin grafts, suggesting the development of regulatory mechanisms capable of maintaining donor-specific tolerance. The reported findings may contribute to the development of new therapeutic strategies to induce transplantation tolerance in humans.


Subject(s)
Cell Differentiation , Dendritic Cells/immunology , Dendritic Cells/transplantation , Immunosuppression Therapy , Tissue Donors , Transplantation Tolerance/immunology , Animals , Dendritic Cells/cytology , Male , Rats , Transplantation, Homologous
19.
Transplantation ; 80(10): 1476-84, 2005 Nov 27.
Article in English | MEDLINE | ID: mdl-16340794

ABSTRACT

BACKGROUND: We have previously demonstrated that a short-course treatment with LF15-0195, a 15-deoxyspergualin analogue, induces donor-specific tolerance of cardiac allografts in rats and expansion of splenic CD4CD25 regulatory T cells. METHODS: To further characterize long-term tolerance in this model, we have analyzed the phenotype, regulatory properties and TCR-Vbeta usage of the T cells infiltrating the tolerated allografts. RESULTS: We demonstrate that the tolerated allografts express high levels of FoxP3 transcripts and contain a large number of CD4 T cells, half of which express CD25. Moreover, T cells from these tolerated allografts are very powerful at transferring tolerance to a subsequent allograft recipient, demonstrating the presence of potent regulatory T cells at the site of the graft. Interestingly, the T cells infiltrating the tolerated allografts systematically display restricted Vbeta7 TCR rearrangements. CONCLUSION: These results demonstrate in this model of tolerance, a specific accumulation of T cells with potent regulatory properties and exhibiting restricted Vbeta7-TCR rearrangements at the graft site.


Subject(s)
Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology , Guanidines/pharmacology , Heart Transplantation/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes, Regulatory/physiology , Transplantation Tolerance/immunology , Animals , Heart Transplantation/pathology , Male , Rats , Rats, Inbred Lew , Transplantation Tolerance/drug effects
20.
Immunobiology ; 220(5): 692-700, 2015 May.
Article in English | MEDLINE | ID: mdl-25433635

ABSTRACT

Receptor activating NF-κB ligand (RANKL) is a member of the TNF superfamily that plays a pivotal role in bone homeostasis as being the major osteoclastogenesis factor. RANKL also has pleiotropic effects in the immune system in which it is expressed by activated T and B cells and some innate lymphoid cells. RANKL-RANK interactions mediate lymph node organogenesis and immunoregulatory functions in autoimmune disease and carcinogenesis as well as cross talk between the immune system and bone. In this study, we show that basophils were the strongest RANKL mRNA-expressing cells amongst major leukocyte subsets in human blood. RANKL was preformed as an intracellular protein in resting basophils and was rapidly and strongly expressed on their surface upon stimulation with IL-3, but not other stimuli. This expression was stable for at least 6 days. Activated basophils could also release soluble RANKL in small quantities upon interaction with DCs or monocytes. In the blood, basophils were the sole cells to express membrane RANKL in response to IL-3. This study indicates that basophils should be considered as new players in the pleiotropic and complex RANKL-RANK interaction system and suggests a role for RANKL in the interaction between basophils and immune cells in inflammatory allergic tissues and secondary lymphoid organs.


Subject(s)
Basophils/immunology , Bone Resorption/immunology , Cell Membrane/metabolism , Dendritic Cells/immunology , Intracellular Space/metabolism , Osteoclasts/physiology , RANK Ligand/metabolism , Cell Communication , Cell Differentiation , Cells, Cultured , Humans , Interleukin-3/immunology , Protein Transport/immunology , RANK Ligand/genetics , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL