Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Publication year range
1.
Immunity ; 39(4): 711-21, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24138882

ABSTRACT

Toll-like receptor 7 (TLR7) triggers antiviral immune responses by recognizing viral single-stranded RNA in endosomes, but the biosynthetic pathway of human TLR7 (hTLR7) remains unclear. Here, we show that hTLR7 is proteolytically processed and that the C-terminal fragment selectively accumulates in endocytic compartments. hTLR7 processing occurred at neutral pH and was dependent on furin-like proprotein convertases (PCs). Furthermore, TLR7 processing was required for its functional response to TLR7 agonists such as R837 or influenza virus. Notably, proinflammatory and differentiation stimuli increased the expression of furin-like PCs in immune cells, suggesting a positive feedback mechanism for TLR7 processing during infection. Because self-RNA can under certain conditions activate TLR7 and trigger autoimmunity, our results identify furin-like PCs as a possible target to attenuate TLR7-dependent autoimmunity and other immune pathologies.


Subject(s)
Furin/metabolism , Macrophages/metabolism , Proprotein Convertases/metabolism , Protein Processing, Post-Translational , Toll-Like Receptor 7/metabolism , Amino Acid Sequence , Autoimmunity , Cell Line , Endosomes/drug effects , Endosomes/immunology , Feedback, Physiological , Furin/genetics , Furin/immunology , Gene Expression Regulation , Genetic Vectors , Humans , Lentivirus/genetics , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Molecular Sequence Data , Orthomyxoviridae/immunology , Proprotein Convertases/genetics , Proprotein Convertases/immunology , Protein Structure, Tertiary , Quinolines/pharmacology , Signal Transduction , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology
2.
Proc Natl Acad Sci U S A ; 113(6): E772-81, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26798067

ABSTRACT

Invariant natural killer T (iNKT) cells recognize endogenous and exogenous lipid antigens presented in the context of CD1d molecules. The ability of iNKT cells to recognize endogenous antigens represents a distinct immune recognition strategy, which underscores the constitutive memory phenotype of iNKT cells and their activation during inflammatory conditions. However, the mechanisms regulating such "tonic" activation of iNKT cells remain unclear. Here, we show that the spatiotemporal distribution of CD1d molecules on the surface of antigen-presenting cells (APCs) modulates activation of iNKT cells. By using superresolution microscopy, we show that CD1d molecules form nanoclusters at the cell surface of APCs, and their size and density are constrained by the actin cytoskeleton. Dual-color single-particle tracking revealed that diffusing CD1d nanoclusters are actively arrested by the actin cytoskeleton, preventing their further coalescence. Formation of larger nanoclusters occurs in the absence of interactions between CD1d cytosolic tail and the actin cytoskeleton and correlates with enhanced iNKT cell activation. Importantly and consistently with iNKT cell activation during inflammatory conditions, exposure of APCs to the Toll-like receptor 7/8 agonist R848 increases nanocluster density and iNKT cell activation. Overall, these results define a previously unidentified mechanism that modulates iNKT cell autoreactivity based on the tight control by the APC cytoskeleton of the sizes and densities of endogenous antigen-loaded CD1d nanoclusters.


Subject(s)
Actin Cytoskeleton/metabolism , Antigen-Presenting Cells/metabolism , Antigens, CD1d/metabolism , Nanoparticles/chemistry , Natural Killer T-Cells/metabolism , Cell Line , Cell Membrane/metabolism , Diffusion , Galactosylceramides/metabolism , Humans , Inflammation/pathology , Lymphocyte Activation , Models, Biological , Monocytes/metabolism , Protein Transport , Spatio-Temporal Analysis
3.
J Infect Dis ; 217(6): 988-999, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29267892

ABSTRACT

Mucosal-associated invariant T (MAIT) cells represent an innate T-cell population that can recognize ligands generated by the microbial riboflavin synthesis pathway, presented via the major histocompatibility complex class I-related molecule (MR1). Streptococcus pneumoniae is a major human pathogen that is also associated with commensal carriage; thus, host control at the mucosal interface is critical. The recognition of pneumococci by MAIT cells has not been defined nor have the genomics and transcriptomics of the riboflavin operon. We observed robust recognition of pneumococci by MAIT cells, using both MR1-dependent and MR1-independent pathways. The pathway used was dependent on the antigen-presenting cell. The riboflavin operon was highly conserved across a range of 571 pneumococci from 39 countries, dating back to 1916, and different versions of the riboflavin operon were also identified in related Streptococcus species. These data indicate an important functional relationship between MAIT cells and pneumococci.


Subject(s)
Cytokines/metabolism , Genes, MHC Class I/immunology , Mucosal-Associated Invariant T Cells/physiology , Streptococcus pneumoniae/genetics , Cells, Cultured , Cytokines/genetics , Genome, Bacterial , Humans , Immunity, Cellular , Macrophages , Operon , Riboflavin/biosynthesis , Streptococcus pneumoniae/classification , Up-Regulation
4.
Eur J Immunol ; 46(5): 1224-34, 2016 05.
Article in English | MEDLINE | ID: mdl-26873393

ABSTRACT

Invariant natural killer T (iNKT) cells recognize CD1d/glycolipid complexes and upon activation with synthetic agonists display immunostimulatory properties. We have previously described that the non-glycosidic CD1d-binding lipid, threitolceramide (ThrCer) activates murine and human iNKT cells. Here, we show that incorporating the headgroup of ThrCer into a conformationally more restricted 6- or 7-membered ring results in significantly more potent non-glycosidic analogs. In particular, ThrCer 6 was found to promote strong anti-tumor responses and to induce a more prolonged stimulation of iNKT cells than does the canonical α-galactosylceramide (α-GalCer), achieving an enhanced T-cell response at lower concentrations compared with α-GalCer both in vitro, using human iNKT-cell lines and in vivo, using C57BL/6 mice. Collectively, these studies describe novel non-glycosidic ThrCer-based analogs that have improved potency in iNKT-cell activation compared with that of α-GalCer, and are clinically relevant iNKT-cell agonists.


Subject(s)
Ceramides/immunology , Natural Killer T-Cells/immunology , Sugar Alcohols/immunology , Animals , Antigens, CD1d/immunology , Ceramides/chemical synthesis , Ceramides/chemistry , Ceramides/pharmacology , Cytokines/immunology , Galactosylceramides/immunology , Galactosylceramides/pharmacology , Humans , Immunotherapy , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/physiology , Neoplasms/immunology , Sugar Alcohols/chemical synthesis , Sugar Alcohols/chemistry , Sugar Alcohols/pharmacology
5.
J Immunol ; 194(11): 5417-25, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25917086

ABSTRACT

TLR7 mediates innate immune responses to viral RNA in endocytic compartments. Mouse and human (h)TLR7 undergo proteolytic cleavage, resulting in the generation of a C-terminal fragment that accumulates in endosomes and associates with the signaling adaptor MyD88 upon receptor triggering by TLR7 agonists. Although mouse TLR7 is cleaved in endosomes by acidic proteases, hTLR7 processing can occur at neutral pH throughout the secretory pathway through the activity of furin-like proprotein convertases. However, the mechanisms by which cleaved hTLR7 reaches the endosomal compartment remain unclear. In this study, we demonstrate that, after hTLR7 proteolytic processing, the liberated amino (N)-terminal fragment remains bound to the C terminus through disulfide bonds and provides key trafficking information that ensures correct delivery of the complex to endosomal compartments. In the absence of the N-terminal fragment, the C-terminal fragment is redirected to the cell surface, where it is functionally inactive. Our data reveal a novel role for the N terminus of hTLR7 as a molecular chaperone that provides processed hTLR7 with the correct targeting instructions to reach the endosomal compartment, hence ensuring its biological activity and preventing inadvertent cell surface responses to self-RNA.


Subject(s)
Endosomes/metabolism , Molecular Chaperones/metabolism , Peptide Fragments/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Carrier Proteins/metabolism , Cell Line , HEK293 Cells , Humans , Interleukin-8/metabolism , Myeloid Differentiation Factor 88/metabolism , Proprotein Convertases/metabolism , Protein Processing, Post-Translational/genetics , Protein Transport/physiology , Proteolysis , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
6.
Proc Natl Acad Sci U S A ; 111(52): E5678-87, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25512546

ABSTRACT

Autophagy is an evolutionarily conserved cellular homeostatic pathway essential for development, immunity, and cell death. Although autophagy modulates MHC antigen presentation, it remains unclear whether autophagy defects impact on CD1d lipid loading and presentation to invariant natural killer T (iNKT) cells and on iNKT cell differentiation in the thymus. Furthermore, it remains unclear whether iNKT and conventional T cells have similar autophagy requirements for differentiation, survival, and/or activation. We report that, in mice with a conditional deletion of the essential autophagy gene Atg7 in the T-cell compartment (CD4 Cre-Atg7(-/-)), thymic iNKT cell development--unlike conventional T-cell development--is blocked at an early stage and mature iNKT cells are absent in peripheral lymphoid organs. The defect is not due to altered loading of intracellular iNKT cell agonists; rather, it is T-cell-intrinsic, resulting in enhanced susceptibility of iNKT cells to apoptosis. We show that autophagy increases during iNKT cell thymic differentiation and that it developmentally regulates mitochondrial content through mitophagy in the thymus of mice and humans. Autophagy defects result in the intracellular accumulation of mitochondrial superoxide species and subsequent apoptotic cell death. Although autophagy-deficient conventional T cells develop normally, they show impaired peripheral survival, particularly memory CD8(+) T cells. Because iNKT cells, unlike conventional T cells, differentiate into memory cells while in the thymus, our results highlight a unique autophagy-dependent metabolic regulation of adaptive and innate T cells, which is required for transition to a quiescent state after population expansion.


Subject(s)
Autophagy/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Immunologic Memory , Natural Killer T-Cells/immunology , Thymus Gland/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Autophagy/genetics , Autophagy-Related Protein 7 , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Cell Survival/genetics , Cell Survival/immunology , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/immunology , Natural Killer T-Cells/cytology , Superoxides/immunology , Thymus Gland/cytology
7.
J Immunol ; 193(5): 2118-26, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25063864

ABSTRACT

The mechanisms behind destruction of the adrenal glands in autoimmune Addison's disease remain unclear. Autoantibodies against steroid 21-hydroxylase, an intracellular key enzyme of the adrenal cortex, are found in >90% of patients, but these autoantibodies are not thought to mediate the disease. In this article, we demonstrate highly frequent 21-hydroxylase-specific T cells detectable in 20 patients with Addison's disease. Using overlapping 18-aa peptides spanning the full length of 21-hydroxylase, we identified immunodominant CD8(+) and CD4(+) T cell responses in a large proportion of Addison's patients both ex vivo and after in vitro culture of PBLs ≤20 y after diagnosis. In a large proportion of patients, CD8(+) and CD4(+) 21-hydroxylase-specific T cells were very abundant and detectable in ex vivo assays. HLA class I tetramer-guided isolation of 21-hydroxylase-specific CD8(+) T cells showed their ability to lyse 21-hydroxylase-positive target cells, consistent with a potential mechanism for disease pathogenesis. These data indicate that strong CTL responses to 21-hydroxylase often occur in vivo, and that reactive CTLs have substantial proliferative and cytolytic potential. These results have implications for earlier diagnosis of adrenal failure and ultimately a potential target for therapeutic intervention and induction of immunity against adrenal cortex cancer.


Subject(s)
Addison Disease/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Immunity, Cellular , Peptides/immunology , Steroid 21-Hydroxylase/immunology , Addison Disease/pathology , Adolescent , Adrenal Cortex Neoplasms/immunology , Adrenal Cortex Neoplasms/pathology , Adult , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Humans , Middle Aged
8.
Proc Natl Acad Sci U S A ; 110(49): E4753-61, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24248359

ABSTRACT

Lipid transfer proteins, such as molecules of the saposin family, facilitate extraction of lipids from biological membranes for their loading onto CD1d molecules. Although it has been shown that prosaposin-deficient mice fail to positively select invariant natural killer T (iNKT) cells, it remains unclear whether saposins can facilitate loading of endogenous iNKT cell agonists in the periphery during inflammatory responses. In addition, it is unclear whether saposins, in addition to loading, also promote dissociation of lipids bound to CD1d molecules. To address these questions, we used a combination of cellular assays and demonstrated that saposins influence CD1d-restricted presentation to human iNKT cells not only of exogenous lipids but also of endogenous ligands, such as the self-glycosphingolipid ß-glucopyranosylceramide, up-regulated by antigen-presenting cells following bacterial infection. Furthermore, we demonstrated that in human myeloid cells CD1d-loading of endogenous lipids after bacterial infection, but not at steady state, requires trafficking of CD1d molecules through an endo-lysosomal compartment. Finally, using BIAcore assays we demonstrated that lipid-loaded saposin B increases the off-rate of lipids bound to CD1d molecules, providing important insights into the mechanisms by which it acts as a "lipid editor," capable of fine-tuning loading and unloading of CD1d molecules. These results have important implications in understanding how to optimize lipid-loading onto antigen-presenting cells, to better harness iNKT cells central role at the interface between innate and adaptive immunity.


Subject(s)
Antigens, CD1d/metabolism , Immunity, Innate/immunology , Lipid Metabolism/physiology , Natural Killer T-Cells/immunology , Saposins/metabolism , Antigen-Presenting Cells/immunology , Bacteria/immunology , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Immunoprecipitation , Scintillation Counting
9.
J Exp Med ; 204(5): 1131-44, 2007 May 14.
Article in English | MEDLINE | ID: mdl-17485514

ABSTRACT

CD1d-restricted lymphocytes recognize a broad lipid range. However, how CD1d-restricted lymphocytes translate T cell receptor (TCR) recognition of lipids with similar group heads into distinct biological responses remains unclear. Using a soluble invariant NKT (iNKT) TCR and a newly engineered antibody specific for alpha-galactosylceramide (alpha-GalCer)-human CD1d (hCD1d) complexes, we measured the affinity of binding of iNKT TCR to hCD1d molecules loaded with a panel of alpha-GalCer analogues and assessed the rate of dissociation of alpha-GalCer and alpha-GalCer analogues from hCD1d molecules. We extended this analysis by studying iNKT cell synapse formation and iNKT cell activation by the same panel of alpha-GalCer analogues. Our results indicate the unique role of the lipid chain occupying the hCD1d F' channel in modulating TCR binding affinity to hCD1d-lipid complexes, the formation of stable immunological synapse, and cell activation. These data are consistent with previously described conformational changes between empty and loaded hCD1d molecules (Koch, M., V.S. Stronge, D. Shepherd, S.D. Gadola, B. Mathew, G. Ritter, A.R. Fersht, G.S. Besra, R.R. Schmidt, E.Y. Jones, and V. Cerundolo. 2005. Nat. Immunol 6:819-826), suggesting that incomplete occupation of the hCD1d F' channel results in conformational differences at the TCR recognition surface. This indirect effect provides a general mechanism by which lipid-specific lymphocytes are capable of recognizing both the group head and the length of lipid antigens, ensuring greater specificity of antigen recognition.


Subject(s)
Antigens, CD1/metabolism , Glycolipids/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Models, Molecular , Receptors, Antigen, T-Cell/metabolism , Antigens, CD1d , Calcium/metabolism , Cell Line , Enzyme-Linked Immunosorbent Assay , Galactosylceramides/immunology , Galactosylceramides/metabolism , Humans , Molecular Structure , Protein Binding
10.
Eur J Immunol ; 42(7): 1886-92, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22585405

ABSTRACT

Invariant natural killer T (iNKT) cells are a specialised subset of T cells that are restricted to the MHC class I like molecule, CD1d. The ligands for iNKT cells are lipids, with the canonical superagonist being α-galactosylceramide, a non-mammalian glycosphingolipid. Trafficking of CD1d through the lysosome is required for the development of murine iNKT cells. Niemann-Pick type C (NPC) disease is a lysosomal storage disorder caused by dysfunction in either of two lysosomal proteins, NPC1 or NPC2, resulting in the storage of multiple lipids, including glycosphingolipids. In the NPC1 mouse model, iNKT cells are virtually undetectable, which is likely due to the inability of CD1d to be loaded with the selecting ligand due to defective lysosomal function and/or CD1d trafficking. However, in this study we have found that in NPC1 patients iNKT cells are present at normal frequencies, with no phenotypic or functional differences. In addi-tion, antigen-presenting cells derived from NPC1 patients are functionally competent to present several different CD1d/iNKT-cell ligands. This further supports the hypothesis that there are different trafficking requirements for the development of murine and human iNKT cells, and a functional lysosomal/late-endosomal compartment is not required for human iNKT-cell development.


Subject(s)
Antigens, CD1d/immunology , Lysosomes/immunology , Natural Killer T-Cells/immunology , Niemann-Pick Disease, Type C/immunology , Animals , Cell Line , Cell Survival/immunology , Disease Models, Animal , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Humans , Interferon-gamma/immunology , Interleukin-4/immunology , Mice , Natural Killer T-Cells/cytology
11.
J Immunol ; 186(3): 1348-60, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21191069

ABSTRACT

NKT cells with an invariant Ag receptor (iNKT cells) represent a highly conserved and unique subset of T lymphocytes having properties of innate and adaptive immune cells. They have been reported to regulate a variety of immune responses, including the response to cancers and the development of autoimmunity. The development and activation of iNKT cells is dependent on self-Ags presented by the CD1d Ag-presenting molecule. It is widely believed that these self-Ags are glycosphingolipids (GSLs), molecules that contain ceramide as the lipid backbone. In this study, we used a variety of methods to show that mammalian Ags for mouse iNKT cells need not be GSLs, including the use of cell lines deficient in GSL biosynthesis and an inhibitor of GSL biosynthesis. Presentation of these Ags required the expression of CD1d molecules that could traffic to late endosomes, the site where self-Ag is acquired. Extracts of APCs contain a self-Ag that could stimulate iNKT cells when added to plates coated with soluble, rCD1d molecules. The Ag(s) in these extracts are resistant to sphingolipid-specific hydrolase digestion, consistent with the results using live APCs. Lyosphosphatidylcholine, a potential self-Ag that activated human iNKT cell lines, did not activate mouse iNKT cell hybridomas. Our data indicate that there may be more than one type of self-Ag for iNKT cells, that the self-Ags comparing mouse and human may not be conserved, and that the search to identify these molecules should not be confined to GSLs.


Subject(s)
Autoantigens/physiology , Glycosphingolipids , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Animals , Antigen Presentation/immunology , CHO Cells , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Glucosyltransferases/immunology , Glucosyltransferases/metabolism , Glycosphingolipids/immunology , Humans , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/cytology , Spleen/immunology , Spleen/metabolism
12.
Biomolecules ; 13(3)2023 03 22.
Article in English | MEDLINE | ID: mdl-36979508

ABSTRACT

Core-shell superparamagnetic iron oxide nanoparticles hold great promise as a theranostic platform in biological systems. Herein, we report the biological effect of multifunctional cyclodextrin-appended SPIONs (CySPION) in mutant Npc1-deficient CHO cells compared to their wild type counterparts. CySPIONs show negligible cytotoxicity while they are strongly endocytosed and localized in the lysosomal compartment. Through their bespoke pH-sensitive chemistry, these nanoparticles release appended monomeric cyclodextrins to mobilize over-accumulated cholesterol and eject it outside the cells. CySPIONs show a high rate of transport across blood-brain barrier models, indicating their promise as a therapeutic approach for cholesterol-impaired diseases affecting the brain.


Subject(s)
Cyclodextrins , Nanoparticles , Cricetinae , Animals , Cricetulus , Precision Medicine , Blood-Brain Barrier , Nanoparticles/therapeutic use , Cholesterol
13.
J Biol Chem ; 286(18): 15973-9, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21454514

ABSTRACT

Invariant natural killer T (iNKT) cells are a population of T lymphocytes that play an important role in regulating immunity to infection and tumors by recognizing endogenous and exogenous CD1d-bound lipid molecules. Using soluble iNKT T cell receptor (TCR) molecules, we applied single molecule force spectroscopy for the investigation of the iNKT TCR affinity for human CD1d molecules loaded with glycolipids differing in the length of the phytosphingosine chain using either recombinant CD1d molecules or lipid-pulsed THP1 cells. In both settings, the dissociation of the iNKT TCR from human CD1d molecules loaded with the lipid containing the longer phytosphingosine chain required higher unbinding forces compared with the shorter phytosphingosine lipid. Our findings are discussed in the context of previous results obtained by surface plasmon resonance measurements. We present new insights into the energy landscape and the kinetic rate constants of the iNKT TCR/human CD1d-glycosphingolipid interaction and emphasize the unique potential of single molecule force spectroscopy on living cells.


Subject(s)
Antigens, CD1d/chemistry , Glycosphingolipids/chemistry , Killer Cells, Natural/chemistry , Receptors, Antigen, T-Cell/chemistry , Antigens, CD1d/genetics , Antigens, CD1d/immunology , Cell Line , Glycosphingolipids/genetics , Glycosphingolipids/immunology , Humans , Killer Cells, Natural/immunology , Kinetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Spectrum Analysis
14.
J Exp Med ; 203(3): 699-710, 2006 Mar 20.
Article in English | MEDLINE | ID: mdl-16520393

ABSTRACT

Invariant human TCR Valpha24-Jalpha18+/Vbeta11+ NKT cells (iNKT) are restricted by CD1d-alpha-glycosylceramides. We analyzed crystal structures and binding characteristics for an iNKT TCR plus two CD1d-alpha-GalCer-specific Vbeta11+ TCRs that use different TCR Valpha chains. The results were similar to those previously reported for MHC-peptide-specific TCRs, illustrating the versatility of the TCR platform. Docking TCR and CD1d-alpha-GalCer structures provided plausible insights into their interaction. The model supports a diagonal orientation of TCR on CD1d and suggests that complementarity determining region (CDR)3alpha, CDR3beta, and CDR1beta interact with ligands presented by CD1d, whereas CDR2beta binds to the CD1d alpha1 helix. This docking provides an explanation for the dominant usage of Vbeta11 and Vbeta8.2 chains by human and mouse iNKT cells, respectively, for recognition of CD1d-alpha-GalCer.


Subject(s)
Antigens, CD1/chemistry , Complementarity Determining Regions/chemistry , Galactosylceramides/chemistry , Killer Cells, Natural , Receptors, Antigen, T-Cell, alpha-beta/chemistry , T-Lymphocytes , Animals , Antigen Presentation/immunology , Antigens, CD1/immunology , Antigens, CD1d , Complementarity Determining Regions/immunology , Crystallography, X-Ray , Galactosylceramides/immunology , Humans , Killer Cells, Natural/immunology , Mice , Models, Molecular , Protein Binding/immunology , Protein Structure, Quaternary , Receptors, Antigen, T-Cell, alpha-beta/immunology , Structure-Activity Relationship , T-Lymphocytes/immunology
15.
J Immunol ; 184(4): 1829-1839, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20053942

ABSTRACT

Although several cancer immunotherapy strategies are based on the use of analog peptides and on the modulation of the TCR affinity of adoptively transferred T cells, it remains unclear whether tumor-specific T cell activation by strong and weak TCR stimuli evoke different Ca(2+) signatures from the Ca(2+) intracellular stores and whether the amplitude of Ca(2+) release from the endoplasmic reticulum (ER) can be further modulated by coreceptor binding to peptide/MHC. In this study, we combined functional, structural, and kinetic measurements to correlate the intensity of Ca(2+) signals triggered by the stimulation of the 1G4 T cell clone specific to the tumor epitope NY-ESO-1(157-165). Two analogs of the NY-ESO-1(157-165) peptide, having similar affinity to HLA-A2 molecules, but a 6-fold difference in binding affinity for the 1G4 TCR, resulted in different Ca(2+) signals and T cell activation. 1G4 stimulation by the stronger stimulus emptied the ER of stored Ca(2+), even in the absence of CD8 binding, resulting in sustained Ca(2+) influx. In contrast, the weaker stimulus induced only partial emptying of stored Ca(2+), resulting in significantly diminished and oscillatory Ca(2+) signals, which were enhanced by CD8 binding. Our data define the range of TCR/peptide MHC affinities required to induce depletion of Ca(2+) from intracellular stores and provide insights into the ability of T cells to tailor the use of the CD8 coreceptor to enhance Ca(2+) release from the ER. This, in turn, modulates Ca(2+) influx from the extracellular environment, ultimately controlling T cell activation.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Epitopes, T-Lymphocyte/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Clone Cells , Crystallography, X-Ray , Cytoplasmic Granules/immunology , Cytoplasmic Granules/metabolism , Cytotoxicity Tests, Immunologic , Endoplasmic Reticulum/immunology , Epitopes, T-Lymphocyte/metabolism , Humans , Lymphocyte Activation/immunology , Neoplasm Proteins/metabolism , Peptide Fragments/metabolism , Protein Binding/immunology , Protein Isoforms/immunology , Protein Isoforms/metabolism , Receptors, Antigen, T-Cell/physiology
16.
Nat Commun ; 13(1): 5320, 2022 09 09.
Article in English | MEDLINE | ID: mdl-36085278

ABSTRACT

Mycobacterium tuberculosis (Mtb) survives and replicates within host macrophages (MΦ) and subverts multiple antimicrobial defense mechanisms. Previously, we reported that lipids shed by pathogenic mycobacteria inhibit NPC1, the lysosomal membrane protein deficient in the lysosomal storage disorder Niemann-Pick disease type C (NPC). Inhibition of NPC1 leads to a drop in lysosomal calcium levels, blocking phagosome-lysosome fusion leading to mycobacterial survival. We speculated that the production of specific cell wall lipid(s) that inhibit NPC1 could have been a critical step in the evolution of pathogenicity. We therefore investigated whether lipid extracts from clinical Mtb strains from multiple Mtb lineages, Mtb complex (MTBC) members and non-tubercular mycobacteria (NTM) inhibit the NPC pathway. We report that inhibition of the NPC pathway was present in all clinical isolates from Mtb lineages 1, 2, 3 and 4, Mycobacterium bovis and the NTM, Mycobacterium abscessus and Mycobacterium avium. However, lipid extract from Mycobacterium canettii, which is considered to resemble the common ancestor of the MTBC did not inhibit the NPC1 pathway. We conclude that the evolution of NPC1 inhibitory mycobacterial cell wall lipids evolved early and post divergence from Mycobacterium canettii-related mycobacteria and that this activity contributes significantly to the promotion of disease.


Subject(s)
Mycobacterium Infections , Mycobacterium bovis , Humans , Lipids , Mycobacterium , Niemann-Pick C1 Protein
17.
Wellcome Open Res ; 7: 267, 2022.
Article in English | MEDLINE | ID: mdl-37065726

ABSTRACT

Background: Niemann-Pick disease type C1 (NPC1) is a neurodegenerative lysosomal storage disorder characterized by the accumulation of multiple lipids in the late endosome/lysosomal system and reduced acidic store calcium. The lysosomal system regulates key aspects of iron homeostasis, which prompted us to investigate whether there are hematological abnormalities and iron metabolism defects in NPC1. Methods: Iron-related hematological parameters, systemic and tissue metal ion and relevant hormonal and proteins levels, expression of specific pro-inflammatory mediators and erythrophagocytosis were evaluated in an authentic mouse model and in a large cohort of NPC patients. Results: Significant changes in mean corpuscular volume and corpuscular hemoglobin were detected in Npc1 -/- mice from an early age. Hematocrit, red cell distribution width and hemoglobin changes were observed in late-stage disease animals. Systemic iron deficiency, increased circulating hepcidin, decreased ferritin and abnormal pro-inflammatory cytokine levels were also found. Furthermore, there is evidence of defective erythrophagocytosis in Npc1 -/- mice and in an in vitro NPC1 cellular model. Comparable hematological changes, including low normal serum iron and transferrin saturation and low cerebrospinal fluid ferritin were confirmed in NPC1 patients. Conclusions: These data suggest loss of iron homeostasis and hematological abnormalities in NPC1 may contribute to the pathophysiology of this disease.

18.
J Immunother Cancer ; 9(6)2021 06.
Article in English | MEDLINE | ID: mdl-34088742

ABSTRACT

BACKGROUND: NY-ESO-1 is a tumor-specific, highly immunogenic, human germ cell antigen of the MAGE-1 family that is a promising vaccine and cell therapy candidate in clinical trial development. The mouse genome does not encode an NY-ESO-1 homolog thereby not subjecting transgenic T-cells to thymic tolerance mechanisms that might impair in-vivo studies. We hypothesized that an NY-ESO-1 T cell receptor (TCR) transgenic mouse would provide the unique opportunity to study avidity of TCR response against NY-ESO-1 for tumor vaccine and cellular therapy development against this clinically relevant and physiological human antigen. METHODS: To study in vitro and in vivo the requirements for shaping an effective T cell response against the clinically relevant NY-ESO-1, we generated a C57BL/6 HLA-A*0201 background TCR transgenic mouse encoding the 1G4 TCR specific for the human HLA-A2 restricted, NY-ESO-1157-165 SLLMWITQC (9C), initially identified in an NY-ESO-1 positive melanoma patient. RESULTS: The HLA-A*0201 restricted TCR was positively selected on both CD4+ and CD8+ cells. Mouse 1G4 T cells were not activated by endogenous autoimmune targets or a large library of non-cognate viral antigens. In contrast, their activation by HLA-A2 NY-ESO-1157-165 complexes was evident by proliferation, CD69 upregulation, interferon-γ production, and interleukin-2 production, and could be tuned using a twofold higher affinity altered peptide ligand, NY-ESO-1157-165V. NY-ESO-1157-165V recombinant vaccination of syngeneic mice adoptively transferred with m1G4 CD8+ T cells controlled tumor growth in vivo. 1G4 transgenic mice suppressed growth of syngeneic methylcholanthrene (MCA) induced HHD tumor cells expressing the full-length human NY-ESO-1 protein but not MCA HHD tumor cells lacking NY-ESO-1. CONCLUSIONS: The 1G4 TCR mouse model for the physiological human TCR against the clinically relevant antigen, NY-ESO-1, is a valuable tool with the potential to accelerate clinical development of NY-ESO-1-targeted T-cell and vaccine therapies.


Subject(s)
HLA-A2 Antigen/metabolism , Neoplasm Proteins/administration & dosage , Peptide Fragments/administration & dosage , Receptors, Antigen, T-Cell/genetics , Thymoma/drug therapy , Thymus Neoplasms/drug therapy , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/immunology , Thymoma/genetics , Thymoma/immunology , Thymus Neoplasms/genetics , Thymus Neoplasms/immunology , Xenograft Model Antitumor Assays
20.
Proc Natl Acad Sci U S A ; 104(51): 20490-5, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-18077358

ABSTRACT

Invariant natural killer T (iNKT) cells are a subset of nonconventional T cells recognizing endogenous and/or exogenous glycolipid antigens in the context of CD1d molecules. It remains unclear whether innate stimuli can modify the profile of endogenous lipids recognized by iNKT cells on the surface of antigen-presenting cells (APCs). We report that activation of human APCs by Toll-like receptor ligands (TLR-L) modulates the lipid biosynthetic pathway, resulting in enhanced recognition of CD1d-associated lipids by iNKT cells, as defined by IFN-gamma secretion. APC-derived soluble factors further increase CD1d-restricted iNKT cell activation. Finally, using soluble tetrameric iNKT T cell receptors (TCR) as a staining reagent, we demonstrate specific up-regulation of CD1d-bound ligand(s) on TLR-mediated APC maturation. The ability of innate stimuli to modulate the lipid profile of APCs resulting in iNKT cell activation and APC maturation underscores the role of iNKT cells in assisting priming of antigen-specific immune responses.


Subject(s)
Antigen-Presenting Cells/immunology , Glycosphingolipids/metabolism , Killer Cells, Natural/immunology , Receptors, Antigen, T-Cell/agonists , T-Lymphocyte Subsets/immunology , Antigens, CD1/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Humans , Interferon-gamma/metabolism , Ligands , Lymphocyte Activation
SELECTION OF CITATIONS
SEARCH DETAIL