Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Immunity ; 47(5): 875-889.e10, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29166588

ABSTRACT

Migration of activated regulatory T (Treg) cells to inflamed tissue is crucial for their immune-modulatory function. While metabolic reprogramming during Treg cell differentiation has been extensively studied, the bioenergetics of Treg cell trafficking remains undefined. We have investigated the metabolic demands of migrating Treg cells in vitro and in vivo. We show that glycolysis was instrumental for their migration and was initiated by pro-migratory stimuli via a PI3K-mTORC2-mediated pathway culminating in induction of the enzyme glucokinase (GCK). Subsequently, GCK promoted cytoskeletal rearrangements by associating with actin. Treg cells lacking this pathway were functionally suppressive but failed to migrate to skin allografts and inhibit rejection. Similarly, human carriers of a loss-of-function GCK regulatory protein gene-leading to increased GCK activity-had reduced numbers of circulating Treg cells. These cells displayed enhanced migratory activity but similar suppressive function, while conventional T cells were unaffected. Thus, GCK-dependent glycolysis regulates Treg cell migration.


Subject(s)
Glucokinase/physiology , Glycolysis , T-Lymphocytes, Regulatory/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , CD28 Antigens/physiology , CTLA-4 Antigen/physiology , Cells, Cultured , Humans , Mechanistic Target of Rapamycin Complex 1/physiology , Mechanistic Target of Rapamycin Complex 2/physiology , Mice , Mice, Inbred Strains , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology
2.
Immunity ; 42(6): 1087-99, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26070483

ABSTRACT

Effector-T-cell-mediated immunity depends on the efficient localization of antigen-primed lymphocytes to antigen-rich non-lymphoid tissue, which is facilitated by the expression of a unique set of "homing" receptors acquired by memory T cells. We report that engagement of the hepatocyte growth factor (HGF) receptor c-Met by heart-produced HGF during priming in the lymph nodes instructs T cell cardiotropism, which was associated with a specialized homing "signature" (c-Met(+)CCR4(+)CXCR3(+)). c-Met signals facilitated T cell recruitment to the heart via the chemokine receptor CCR5 by inducing autocrine CCR5 ligand release. c-Met triggering was sufficient to support cardiotropic T cell recirculation, while CCR4 and CXCR3 sustained recruitment during heart inflammation. Transient pharmacological blockade of c-Met during T cell priming led to enhanced survival of heart, but not skin, allografts associated with impaired localization of alloreactive T cells to heart grafts. These findings suggest c-Met as a target for development of organ-selective immunosuppressive therapies.


Subject(s)
Graft Rejection/prevention & control , Heart Transplantation , Heart/physiology , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-met/metabolism , T-Lymphocytes/physiology , Animals , Autocrine Communication , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Graft Rejection/etiology , Graft Rejection/genetics , Humans , Immunologic Memory , Indoles/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Mice , Mice, SCID , Molecular Targeted Therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , RNA, Small Interfering/genetics , Receptors, CCR5/metabolism , Receptors, Chemokine/metabolism , Receptors, Lymphocyte Homing/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfones/pharmacology , T-Lymphocytes/drug effects
3.
Circulation ; 146(25): 1930-1945, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36417924

ABSTRACT

BACKGROUND: Autoimmunity is increasingly recognized as a key contributing factor in heart muscle diseases. The functional features of cardiac autoimmunity in humans remain undefined because of the challenge of studying immune responses in situ. We previously described a subset of c-mesenchymal epithelial transition factor (c-Met)-expressing (c-Met+) memory T lymphocytes that preferentially migrate to cardiac tissue in mice and humans. METHODS: In-depth phenotyping of peripheral blood T cells, including c-Met+ T cells, was undertaken in groups of patients with inflammatory and noninflammatory cardiomyopathies, patients with noncardiac autoimmunity, and healthy controls. Validation studies were carried out using human cardiac tissue and in an experimental model of cardiac inflammation. RESULTS: We show that c-Met+ T cells are selectively increased in the circulation and in the myocardium of patients with inflammatory cardiomyopathies. The phenotype and function of c-Met+ T cells are distinct from those of c-Met-negative (c-Met-) T cells, including preferential proliferation to cardiac myosin and coproduction of multiple cytokines (interleukin-4, interleukin-17, and interleukin-22). Furthermore, circulating c-Met+ T cell subpopulations in different heart muscle diseases identify distinct and overlapping mechanisms of heart inflammation. In experimental autoimmune myocarditis, elevations in autoantigen-specific c-Met+ T cells in peripheral blood mark the loss of immune tolerance to the heart. Disease development can be halted by pharmacologic c-Met inhibition, indicating a causative role for c-Met+ T cells. CONCLUSIONS: Our study demonstrates that the detection of circulating c-Met+ T cells may have use in the diagnosis and monitoring of adaptive cardiac inflammation and definition of new targets for therapeutic intervention when cardiac autoimmunity causes or contributes to progressive cardiac injury.


Subject(s)
Autoimmune Diseases , Cardiomyopathies , Myocarditis , Humans , Mice , Animals , Autoimmunity , Memory T Cells , Myocarditis/etiology , Myocardium , Cardiomyopathies/complications , Cardiac Myosins , Inflammation/complications
5.
Proc Natl Acad Sci U S A ; 112(43): E5815-24, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26392551

ABSTRACT

Constitutive resistance to cell death induced by inflammatory stimuli activating the extrinsic pathway of apoptosis is a key feature of vascular endothelial cells (ECs). Although this property is central to the maintenance of the endothelial barrier during inflammation, the molecular mechanisms of EC protection from cell-extrinsic, proapoptotic stimuli have not been investigated. We show that the Ig-family member CD31, which is expressed by endothelial but not epithelial cells, is necessary to prevent EC death induced by TNF-α and cytotoxic T lymphocytes in vitro. Combined quantitative RT-PCR array and biochemical analysis show that, upon the engagement of the TNF receptor with TNF-α on ECs, CD31 becomes activated and, in turn, counteracts the proapoptotic transcriptional program induced by TNF-α via activation of the Erk/Akt pathway. Specifically, Akt activation by CD31 signals prevents the localization of the forkhead transcription factor FoxO3 to the nucleus, thus inhibiting transcription of the proapoptotic genes CD95/Fas and caspase 7 and de-repressing the expression of the antiapoptotic gene cFlar. Both CD31 intracellular immunoreceptor tyrosine-based inhibition motifs are required for its prosurvival function. In vivo, CD31 gene transfer is sufficient to recapitulate the cytoprotective mechanisms in CD31(-) pancreatic ß cells, which become resistant to immune-mediated rejection when grafted in fully allogeneic recipients.


Subject(s)
Endothelium, Vascular/immunology , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Animals , Mice , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/genetics , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor-alpha/physiology
6.
Proc Natl Acad Sci U S A ; 111(50): 18078-83, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25425665

ABSTRACT

HVCN1 (Hydrogen voltage-gated channel 1) is the only mammalian voltage-gated proton channel. In human B lymphocytes, HVCN1 associates with the B-cell receptor (BCR) and is required for optimal BCR signaling and redox control. HVCN1 is expressed in malignant B cells that rely on BCR signaling, such as chronic lymphocytic leukemia (CLL) cells. However, little is known about its regulation in these cells. We found that HVCN1 was expressed in B cells as two protein isoforms. The shorter isoform (HVCN1S) was enriched in B cells from a cohort of 76 CLL patients. When overexpressed in a B-cell lymphoma line, HVCN1S responded more profoundly to protein kinase C-dependent phosphorylation. This more potent enhanced gating response was mediated by increased phosphorylation of the same residue responsible for enhanced gating in HVCN1L, Thr(29). Furthermore, the association of HVCN1S with the BCR was weaker, which resulted in its diminished internalization upon BCR stimulation. Finally, HVCN1S conferred a proliferative and migratory advantage as well as enhanced BCR-dependent signaling. Overall, our data show for the first time, to our knowledge, the existence of a shorter isoform of HVCN1 with enhanced gating that is specifically enriched in malignant B cells. The properties of HVCN1S suggest that it may contribute to the pathogenesis of BCR-dependent B-cell malignancies.


Subject(s)
B-Lymphocytes/metabolism , Hematologic Neoplasms/immunology , Ion Channels/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Mice , Patch-Clamp Techniques , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism
7.
Proc Natl Acad Sci U S A ; 109(45): E3111-8, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23077253

ABSTRACT

The bias of αß T cells for MHC ligands has been proposed to be intrinsic to the T-cell receptor (TCR). Equally, the CD4 and CD8 coreceptors contribute to ligand restriction by colocalizing Lck with the TCR when MHC ligands are engaged. To determine the importance of intrinsic ligand bias, the germ-line TCR complementarity determining regions were extensively diversified in vivo. We show that engagement with MHC ligands during thymocyte selection and peripheral T-cell activation imposes remarkably little constraint over TCR structure. Such versatility is more consistent with an opportunist, rather than a predetermined, mode of interface formation. This hypothesis was experimentally confirmed by expressing a hybrid TCR containing TCR-γ chain germ-line complementarity determining regions, which engaged efficiently with MHC ligands.


Subject(s)
Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Amino Acid Sequence , Animals , Base Sequence , Cell Lineage/immunology , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Germ Cells/immunology , Ligands , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation/genetics , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Recombination, Genetic/genetics , Selection, Genetic , Thymus Gland/immunology
8.
Immunology ; 138(2): 157-64, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23121382

ABSTRACT

Previously we have shown that indoleamine 2,3-dioxygenase (IDO) and the tryptophan metabolite, 3-hydroxykynurenine (3HK) can prolong corneal allograft survival. IDO modulates the immune response by depletion of the essential amino acid tryptophan by breakdown to kynurenines, which themselves act directly on T lymphocytes. The tryptophan metabolite analogue N-(3,4-dimethoxycinnamonyl) anthranilic acid (DAA, 'Tranilast') shares the anthranilic acid core with 3HK. Systemic administration of DAA to mice receiving a fully MHC-mismatched allograft of cornea or skin resulted in significant delay in rejection (median survival of controls 12 days, 13 days for cornea and skin grafts, respectively, and of treated mice 24 days (P < 0.0001) and 17 days (P < 0.03), respectively). We provide evidence that DAA-induced suppression of the allogeneic response, in contrast to that induced by tryptophan metabolites, was a result of cell cycle arrest rather than T-cell death. Cell cycle arrest was mediated by up-regulation of the cell cycle-specific inhibitors p21 and p15, and associated with a significant reduction in interleukin-2 production, allowing us to characterize a novel mechanism for DAA-induced T-cell anergy. Currently licensed as an anti-allergy drug, the oral bioavailability and safe therapeutic profile of DAA make it a candidate for the prevention of rejection of transplanted cornea and other tissues.


Subject(s)
Anti-Allergic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Corneal Transplantation , Graft Survival/drug effects , Skin Transplantation , T-Lymphocytes/immunology , ortho-Aminobenzoates/pharmacology , Animals , Cell Cycle Checkpoints/immunology , Cell Death/drug effects , Cell Death/immunology , Clonal Anergy/drug effects , Clonal Anergy/immunology , Graft Rejection/drug therapy , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interleukin-2/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes/pathology , Transplantation, Homologous , Tryptophan/immunology
9.
Br J Haematol ; 162(6): 808-18, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23855835

ABSTRACT

Pre-transplant conditioning regimens play a major role in triggering graft-versus-host disease (GVHD). This study investigated the effect of irradiation on donor T cell trafficking to lymphoid and non-lymphoid tissues by comparing the migration of carboxy-fluorescein diacetate succinimidyl ester-labelled, naïve donor T lymphocytes in vivo in irradiated and non-irradiated syngeneic mice recipients. Recruitment of adoptively transferred naïve T cells to secondary lymphoid organs was increased in irradiated mice and naïve T cells also aberrantly localized to non-lymphoid tissues. Irradiation also induced aberrant effector memory T cell migration into lymph nodes and their localization to homing-privileged non-lymphoid sites, such as the gut. The presence of a minor histocompatibility mismatch further enhanced the aberrant accumulation of T cells in both lymphoid and non-lymphoid tissue, whilst their migratory pattern was not modified as compared to fully matched irradiated recipients. These effects correlated with decreased permeability of, and the secretion of chemotactic factors by the endothelium. Our findings are consistent with the possibility that excessive, dysregulated extravasation of T cells induced by irradiation promotes the development of GVHD.


Subject(s)
Graft vs Host Disease/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Transplantation Conditioning/methods , Whole-Body Irradiation/methods , Animals , Chemokines/immunology , Chemotaxis/immunology , Chemotaxis/radiation effects , Female , Immunity, Cellular/immunology , Immunity, Cellular/radiation effects , Immunotherapy, Adoptive/methods , Lymphoid Tissue/immunology , Male , Mice , Mice, Inbred C57BL
10.
Cancer Immunol Immunother ; 62(1): 101-12, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22806093

ABSTRACT

Normal tissue and tumour grafts expressing the same alloantigens often elicit distinct immune responses whereby only normal tissue is rejected. To investigate the mechanisms that underlie these distinct outcomes, we compared the responses of adoptively transferred HY-specific conventional (CD8 and CD4) or regulatory T (Treg) cells in mice bearing HY-expressing tumour, syngeneic male skin graft or both. For local T cell priming, T cell re-circulation, graft localization and retention, skin grafts were more efficient than tumours. Skin grafts were also capable of differentiating CD4 T cells into functional Th1 cells. Donor T cell responses were inversely correlated with tumour progression. When skin graft and tumour transplants were performed sequentially, contemporary graft and tumour burden enhanced CD8 but reduced CD4 T cell responses causing accelerated skin-graft rejection without influencing tumour growth. Although both skin grafts and tumours were able to expand HY-specific Treg cells in draining lymph node (dLN), the proportion of tumour-infiltrating Treg cells was significantly higher than that within skin grafts, correlating with accelerated tumour growth. Moreover, there was a higher level of HY antigen presentation by host APC in tumour-dLN than in graft-dLN. Finally, tumour tissues expressed a significant higher level of IDO, TGFß, IL10 and Arginase I than skin grafts, indicating that malignant but not normal tissue represents a stronger immunosuppressive environment. These comparisons provide important insight into the in vivo mechanisms that conspire to compromise tumour-specific adaptive immunity and identify new targets for cancer immunotherapy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , Neoplasms, Experimental/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cell Line, Tumor , Cell Separation , Female , Flow Cytometry , Graft Rejection/immunology , H-Y Antigen/immunology , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Mice, Inbred C57BL , Skin Transplantation/immunology
11.
J Immunol ; 186(8): 4557-64, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21389255

ABSTRACT

Although polyclonal regulatory T cells (Tregs) that once expressed Foxp3 (ex-Tregs) derived from Foxp3(+) Tregs have been described in homeostatic and autoimmune settings, little is known regarding the influence of the tumor environment on ex-Treg development. After adoptive transfer of HY-specific green Tregs (peripheral or thymic) to Rag2(-/-) B6 female mice bearing syngeneic HY-expressing MB49 tumors, a significant fraction rapidly lost expression of Foxp3. On the second transfer to a Rag2(-/-) B6 male environment, these ex-Tregs expanded strongly, whereas Tregs that maintained expression of Foxp3 expression did not. Both FACS and quantitative real-time-PCR analysis revealed that ex-Tregs upregulated genes characteristic of a Th1 effector-memory phenotype including IFN-γ and downregulated a panel of Treg-specific genes. Peripheral HY-specific green Tregs were adoptively transferred to Rag2(-/-) B6 male mice, to dissect the factors regulating ex-Treg differentiation. Development of ex-Tregs was more efficient in the mesenteric lymph node (mLN) than peripheral lymph node environment, correlating with a much greater level of IL-6 mRNA in mLN. In addition, the preferential development of ex-Tregs in mLN was significantly impaired by cotransfer of HY-specific naive CD4 T cells. Collectively, our study not only demonstrates the plasticity of Ag-specific Tregs in the context of the tumor environment, but also defines key molecular and cellular events that modulate ex-Treg differentiation.


Subject(s)
Antigens/immunology , Forkhead Transcription Factors/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cell Line, Tumor , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Female , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , H-Y Antigen/immunology , H-Y Antigen/metabolism , Interleukin-2/genetics , Interleukin-2/metabolism , Male , Mice , Mice, Knockout , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
J Immunol ; 186(10): 5719-28, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21490154

ABSTRACT

We have previously shown that intranasal (i.n.) administration of a single MHC class II-restricted HY peptide to female mice induces tolerance to up to five additional epitopes expressed on test male grafts, a phenomenon known as linked suppression. In this study, we investigated the molecular mechanisms involved both in the induction phase following peptide administration and during linked suppression after grafting. We report that following initial i.n. administration, peptide is widely disseminated and is presented by functionally immature dendritic cells. These fail to cause optimal stimulation of the responding HY-specific CD4(+) T cells that express genes characteristic of regulatory T cells. Following i.n. peptide plus LPS administration, causing immunization, HY-specific CD4(+) T cells express genes characteristic of activated T cells. We further find that following male skin grafting, HY-specific CD8(+) T cells from peptide-treated tolerant mice display both quantitative and qualitative differences compared with similar cells from untreated mice that reject their grafts. In tolerant mice there are fewer HY-specific CD8(+) cells and they express several genes characteristic of exhausted T cells. Furthermore, associated with specific chemokine receptor and integrin expression, HY-specific CD8(+) T cells show more limited migration from the graft draining lymph node into other tissues.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Survival/immunology , H-Y Antigen/immunology , Peptide Fragments/immunology , Transplantation Tolerance , Administration, Intranasal , Adoptive Transfer , Animals , Cell Movement , Cytokines/genetics , Dendritic Cells/immunology , Female , Flow Cytometry , Gene Expression , H-Y Antigen/administration & dosage , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Molecular Sequence Data , Peptide Fragments/administration & dosage , Polymerase Chain Reaction , Skin Transplantation/immunology
13.
Proc Natl Acad Sci U S A ; 107(45): 19461-6, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20978210

ABSTRACT

CD31 is an Ig-like molecule expressed by leukocytes and endothelial cells with an established role in the regulation of leukocyte trafficking. Despite genetic deletion of CD31 being associated with exacerbation of T cell-mediated autoimmunity, the contribution of this molecule to T-cell responses is largely unknown. Here we report that tumor and allograft rejection are significantly enhanced in CD31-deficient mice, which are also resistant to tolerance induction. We propose that these effects are dependent on an as yet unrecognized role for CD31-mediated homophilic interactions between T cells and antigen-presenting cells (APCs) during priming. We show that loss of CD31 interactions leads to enhanced primary clonal expansion, increased killing capacity, and diminished regulatory functions by T cells. Immunomodulation by CD31 signals correlates with a partial inhibition of proximal T-cell receptor (TCR) signaling, specifically Zap-70 phosphorylation. However, CD31-deficient mice do not develop autoimmunity due to increased T-cell death following activation, and we show that CD31 triggering induces Erk-mediated prosurvival activity in T cells either in conjunction with TCR signaling or autonomously. We conclude that CD31 functions as a nonredundant comodulator of T-cell responses, which specializes in sizing the ensuing immune response by setting the threshold for T-cell activation and tolerance, while preventing memory T-cell death.


Subject(s)
Immune Tolerance , Platelet Endothelial Cell Adhesion Molecule-1/immunology , T-Lymphocytes/immunology , Animals , Cell Survival , Clone Cells , Cytotoxicity, Immunologic , Genes, Immunoglobulin , Immunologic Memory , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/genetics
14.
Eur J Immunol ; 41(10): 2997-3005, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21805470

ABSTRACT

The cornea is an immune privileged tissue. Since arginase has been found to modulate T-cell function by depleting arginine, we investigated the expression of arginase in the cornea and its possible role in immune privilege using a murine transplant model. We found that both the endothelium and epithelium of murine corneas express functional arginase I, capable of down-regulating T-cell proliferation in an in vitro culture system. The administration of the specific arginase inhibitor N-hydroxy-nor-L-Arg to recipient mice resulted in an accelerated rejection of allogeneic C57BL/6 (B6) corneal grafts. In contrast, in vivo blockade of arginase activity had no effect in altering the course of rejection of primary skin grafts that express little, if any, arginase. In addition, the inhibition of arginase did not alter systemic T-cell proliferation. These data show that arginase is functional in the cornea and contributes to the immune privilege of the eye, and that modulation of arginase contributes to graft survival.


Subject(s)
Arginase/antagonists & inhibitors , Arginine/metabolism , Cornea/immunology , Corneal Transplantation , Graft Survival , Animals , Arginase/metabolism , Arginine/administration & dosage , Arginine/analogs & derivatives , Arginine/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Endothelium, Corneal/immunology , Endothelium, Corneal/metabolism , Epithelium, Corneal/immunology , Epithelium, Corneal/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Polymerase Chain Reaction , Skin Transplantation , Transplantation, Homologous
15.
J Cell Biol ; 221(11)2022 11 07.
Article in English | MEDLINE | ID: mdl-36129440

ABSTRACT

Activation of T cells relies on the availability of intracellular cholesterol for an effective response after stimulation. We investigated the contribution of cholesterol derived from extracellular uptake by the low-density lipoprotein (LDL) receptor in the immunometabolic response of T cells. By combining proteomics, gene expression profiling, and immunophenotyping, we described a unique role for cholesterol provided by the LDLR pathway in CD8+ T cell activation. mRNA and protein expression of LDLR was significantly increased in activated CD8+ compared to CD4+ WT T cells, and this resulted in a significant reduction of proliferation and cytokine production (IFNγ, Granzyme B, and Perforin) of CD8+ but not CD4+ T cells from Ldlr -/- mice after in vitro and in vivo stimulation. This effect was the consequence of altered cholesterol routing to the lysosome resulting in a lower mTORC1 activation. Similarly, CD8+ T cells from humans affected by familial hypercholesterolemia (FH) carrying a mutation on the LDLR gene showed reduced activation after an immune challenge.


Subject(s)
CD8-Positive T-Lymphocytes , Cholesterol , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 1 , Receptors, LDL , Animals , CD8-Positive T-Lymphocytes/metabolism , Cholesterol/metabolism , Cytokines/metabolism , Granzymes/metabolism , Humans , Hyperlipoproteinemia Type II , Interferon-gamma/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Perforin , RNA, Messenger/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism
16.
JCI Insight ; 7(10)2022 05 23.
Article in English | MEDLINE | ID: mdl-35472029

ABSTRACT

Voltage-gated hydrogen channel 1 (Hvcn1) is a voltage-gated proton channel, which reduces cytosol acidification and facilitates the production of ROS. The increased expression of this channel in some cancers has led to proposing Hvcn1 antagonists as potential therapeutics. While its role in most leukocytes has been studied in depth, the function of Hvcn1 in T cells remains poorly defined. We show that Hvcn1 plays a nonredundant role in protecting naive T cells from intracellular acidification during priming. Despite sharing overall functional impairment in vivo and in vitro, Hvcn1-deficient CD4+ and CD8+ T cells display profound differences during the transition from naive to primed T cells, including in the preservation of T cell receptor (TCR) signaling, cellular division, and death. These selective features result, at least in part, from a substantially different metabolic response to intracellular acidification associated with priming. While Hvcn1-deficient naive CD4+ T cells reprogram to rescue the glycolytic pathway, naive CD8+ T cells, which express high levels of this channel in the mitochondria, respond by metabolically compensating mitochondrial dysfunction, at least in part via AMPK activation. These observations imply heterogeneity between adaptation of naive CD4+ and CD8+ T cells to intracellular acidification during activation.


Subject(s)
Hydrogen , Protons , Hydrogen-Ion Concentration , Lymphocyte Count , Signal Transduction
17.
Hum Mol Genet ; 18(15): 2825-38, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19423552

ABSTRACT

A previously reported blood pressure (BP) quantitative trait locus on rat Chromosome 1 was isolated in a short congenic segment spanning 804.6 kb. The 804.6 kb region contained only two genes, LOC306664 and LOC306665. LOC306664 is predicted to translate into A Disintegrin-like and Metalloproteinase with Thrombospondin Motifs-16 (Adamts16). LOC306665 is a novel gene. All predicted exons of both LOC306664 and LOC306665 were sequenced. Non-synonymous variants were identified in only one of these genes, LOC306664. These variants were naturally existing polymorphisms among inbred, outbred and wild rats. The full-length rat transcript of Adamts16 was detected in multiple tissues. Similar to ADAMTS16 in humans, expression of Adamts16 was prominent in the kidney. Renal transcriptome analysis suggested that a network of genes related to BP was differential between congenic and S rats. These genes were also differentially expressed between kidney cell lines with or without knock-down of Adamts16. Adamts16 is conserved between rats and humans. It is a candidate gene within the homologous region on human Chromosome 5, which is linked to systolic and diastolic BP in the Quebec Family Study. Multiple variants, including an Ala to Pro variant in codon 90 (rs2086310) of human ADAMTS16, were associated with human resting systolic BP (SBP). Replication study in GenNet confirmed the association of two variants of ADAMTS16 with SBP, including rs2086310. Overall, our report represents a high resolution positional cloning and translational study for Adamts16 as a candidate gene controlling BP.


Subject(s)
ADAM Proteins/genetics , Genetic Variation , Hypertension/congenital , Hypertension/genetics , ADAMTS Proteins , ADAMTS1 Protein , Animals , Blood Pressure , Chromosome Mapping , Female , Genetic Linkage , Humans , Hypertension/physiopathology , Male , Quantitative Trait Loci , Rats
18.
Eur J Immunol ; 40(6): 1758-67, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20213737

ABSTRACT

Complement activation is known to have deleterious effects on organ transplantation. On the other hand, the complement system is also known to have an important role in regulating immune responses. The balance between these two opposing effects is critical in the context of transplantation. Here, we report that female mice deficient in C1q (C1qa(-/-)) or C3 (C3(-/-)) reject male syngeneic grafts (HY incompatible) at an accelerated rate compared with WT mice. Intranasal HY peptide administration, which induces tolerance to syngeneic male grafts in WT mice, fails to induce tolerance in C1qa(-/-) or C3(-/-) mice. The rejection of the male grafts correlated with the presence of HY D(b)Uty-specific CD8(+) T cells. Consistent with this, peptide-treated C1qa(-/-) and C3(-/-) female mice rejecting male grafts exhibited more antigen-specific CD8(+)IFN-gamma(+) and CD8(+)IL-10(+) cells compared with WT females. This suggests that accumulation of IFN-gamma- and IL-10-producing T cells may play a key role in mediating the ongoing inflammatory process and graft rejection. Interestingly, within the tolerized male skin grafts of peptide-treated WT mice, IFN-gamma, C1q and C3 mRNA levels were higher compared to control female grafts. These results suggest that C1q and C3 facilitate the induction of intranasal tolerance.


Subject(s)
Complement C1q/immunology , Complement C3/immunology , Graft Rejection/immunology , H-Y Antigen/immunology , Transplantation, Homologous/immunology , Administration, Intranasal , Animals , CD8-Positive T-Lymphocytes/immunology , Complement C1q/deficiency , Complement C3/deficiency , Cytokines/biosynthesis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Female , H-Y Antigen/administration & dosage , Interferon-gamma/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Skin Transplantation/immunology
19.
Am J Pathol ; 176(3): 1494-504, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20093484

ABSTRACT

The metalloprotease ADAMTS9 participates in melanoblast development and is a tumor suppressor in esophageal and nasopharyngeal cancer. ADAMTS9 null mice die before gastrulation, but, ADAMTS9+/- mice were initially thought to be normal. However, when congenic with the C57Bl/6 strain, 80% of ADAMTS9+/- mice developed spontaneous corneal neovascularization. beta-Galactosidase staining enabled by a lacZ cassette targeted to the ADAMTS9 locus showed that capillary endothelial cells (ECs) in embryonic and adult tissues and in capillaries growing into heterotopic tumors expressed ADAMTS9. Heterotopic B.16-F10 melanomas elicited greater vascular induction in ADAMTS9+/- mice than in wild-type littermates, suggesting a potential inhibitory role in tumor angiogenesis. Treatment of cultured human microvascular ECs with ADAMTS9 small-interfering RNA resulted in enhanced filopodial extension, decreased cell adhesion, increased cell migration, and enhanced formation of tube-like structures on Matrigel. Conversely, overexpression of catalytically active, but not inactive, ADAMTS9 in ECs led to fewer tube-like structures, demonstrating that the proteolytic activity of ADAMTS9 was essential. However, unlike the related metalloprotease ADAMTS1, which exerts anti-angiogenic effects by cleavage of thrombospondins and sequestration of vascular endothelial growth factor165, ADAMTS9 neither cleaved thrombospondins 1 and 2, nor bound vascular endothelial growth factor165. Taken together, these data identify ADAMTS9 as a novel, constitutive, endogenous angiogenesis inhibitor that operates cell-autonomously in ECs via molecular mechanisms that are distinct from those used by ADAMTS1.


Subject(s)
ADAM Proteins/metabolism , Endothelial Cells/enzymology , Endothelial Cells/pathology , Microvessels/enzymology , Microvessels/pathology , Neovascularization, Pathologic/enzymology , ADAM Proteins/genetics , ADAMTS9 Protein , Aging/metabolism , Animals , Biocatalysis , Cell Movement , Corneal Neovascularization/enzymology , Corneal Neovascularization/pathology , Embryo, Mammalian/enzymology , Embryo, Mammalian/pathology , Enzyme Activation , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplasms/blood supply , Neoplasms/enzymology , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Organ Specificity , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Thrombospondin 1/metabolism , Thrombospondins/metabolism , Vascular Endothelial Growth Factor A/metabolism
20.
Immunology ; 131(4): 556-69, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20722761

ABSTRACT

In this study we investigated the impact of several factors on the expansion of natural regulatory T (nTreg) cells by tumours, including antigen specificity, transforming growth factor-ß (TGF-ß) signalling and the antigen-presenting cell subsets responsible for expansion. We found that antigen non-specific expansion of nTreg cells is tumour cell line-dependent. Although both antigen-specific and non-specific pathways can contribute to expansion, the migration of activated nTreg cells to tumour tissues is strictly antigen-dependent. Intact TGF-ß signalling on nTreg cells is also essential for tumour-induced expansion. Finally, for stimulation of resting antigen-specific CD4 T cells, CD11c(+) cells purified from tumour-draining lymph nodes were more potent than CD11b(+) cells, suggesting that dendritic cells are the key antigen-presenting cell subset involved in cross-presentation of tumour antigens. This study not only provides an in vivo system in which cross-talk between nTreg cells and tumours can be explored but also reveals novel aspects of tumour immune evasion.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , Cell Proliferation , Cross-Priming/immunology , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Tumor Escape/immunology , Animals , Antigen-Presenting Cells/pathology , CD11c Antigen/immunology , Cell Line, Tumor , Cell Movement/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Knockout , Neoplasms/pathology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/pathology
SELECTION OF CITATIONS
SEARCH DETAIL