Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 473
Filter
1.
Biol Direct ; 19(1): 58, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075541

ABSTRACT

Cluster of Differentiation 1 (CD1) proteins are widely expressed throughout jawed vertebrates and present lipid antigens to specific CD1-restricted T lymphocytes. CD1 molecules play an important role in immune defense with the presence or absence of particular CD1 proteins frequently associated with the functional characteristics of the immune system. Here, we show the evolution of CD1 proteins in the Rodentia family and the diversity among its members. Based on the analysis of CD1 protein-coding regions in rodent genomes and the reconstruction of protein structures, we found that Heterocephalus glaber represents a unique member of the suborder Hystricomorpha with significant changes in protein sequences and structures of the CD1 family. Multiple lines of evidence point to the absence of CD1d and CD1e and probably a dysfunctional CD1b protein in Heterocephalus glaber. In addition, the impact of CD1d loss on the CD1d/Natural killer T (NKT) cell axis in the naked mole-rat and its potential implications for immune system function are discussed in detail.


Subject(s)
Antigens, CD1 , Mole Rats , Animals , Mole Rats/genetics , Mole Rats/immunology , Antigens, CD1/genetics , Antigens, CD1/immunology , Evolution, Molecular , Phylogeny , Immune System , Multigene Family , Natural Killer T-Cells/immunology , Rodentia/genetics , Rodentia/immunology
2.
Cell Rep ; 43(6): 114352, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38870011

ABSTRACT

Addressing the mononuclear phagocyte system (MPS) and macrophage M1/M2 activation is important in diagnosing hematological disorders and inflammatory pathologies and designing therapeutic tools. CSF1R is a reliable marker to identify all circulating MPS cells and tissue macrophages in humans using a single surface protein. CSF1R permits the quantification and isolation of monocyte and dendritic cell (DC) subsets in conjunction with CD14, CD16, and CD1c and is stable across the lifespan and sexes in the absence of overt pathology. Beyond cell detection, measuring M1/M2 activation in humans poses challenges due to response heterogeneity, transient signaling, and multiple regulation steps for transcripts and proteins. MPS cells respond in a conserved manner to M1/M2 pathways such as interleukin-4 (IL-4), steroids, interferon-γ (IFNγ), and lipopolysaccharide (LPS), for which we propose an ad hoc modular gene expression tool. Signature analysis highlights macrophage activation mosaicism in experimental samples, an emerging concept that points to mixed macrophage activation states in pathology.


Subject(s)
Macrophage Activation , Macrophages , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Macrophage Activation/genetics , Macrophages/metabolism , Macrophages/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interferon-gamma/metabolism , Lipopolysaccharides/pharmacology , Female , Mosaicism , Male , Monocytes/metabolism , Lipopolysaccharide Receptors/metabolism , Interleukin-4/metabolism , Dendritic Cells/metabolism , Dendritic Cells/immunology , Receptors, IgG/metabolism , Receptors, IgG/genetics , Antigens, CD1/metabolism , Antigens, CD1/genetics , Mononuclear Phagocyte System/metabolism , Glycoproteins , Receptor, Macrophage Colony-Stimulating Factor
3.
BMC Cancer ; 23(1): 129, 2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36755259

ABSTRACT

BACKGROUND: The tumor microenvironment (TME) in breast cancer plays a vital role in occurrence, development, and therapeutic responses. However, immune and stroma constituents in the TME are major obstacles to understanding and treating breast cancer. We evaluated the significance of TME-related genes in breast cancer. METHODS: Invasive breast cancer (BRCA) samples were retrieved from the TCGA and GEO databases. Stroma and immune scores of samples as well as the proportion of tumor infiltrating immune cells (TICs) were calculated using the ESTIMATE and CIBERSORT algorithms. TME-related differentially expressed genes (DEGs) were analyzed by a protein interaction (PPI) network and univariate Cox regression to determine CD1C as a hub gene. Subsequently, the prognostic value of CD1C, its response to immunotherapy, and its mechanism in the TME were further studied. RESULTS: In BRCA, DEGs were determined to identify CD1C as a hub gene. The expression level of CD1C in BRCA patients was verified based on the TCGA database, polymerase chain reaction (PCR) results, and western blot analysis. Immunohistochemical staining (IHC) results revealed a correlation between prognosis, clinical features, and CD1C expression in BRCA. Enrichment analysis of GSEA and GSVA showed that CD1C participates in immune-associated signaling pathways. CIBERSORT showed that CD1C levels were associated with tumor immune infiltrating cells (TILs), such as different kinds of T cells. Gene co-expression analysis showed that CD1C and the majority of immune-associated genes were co-expressed in BRCA. In renal cell carcinoma, patients with a high expression of CD1C had a better immunotherapy effect. CONCLUSION: CD1C is an important part of the TME and participates in immune activity regulation in breast tumors. CD1C is expected to become a prognostic marker and a new treatment target for breast cancer.


Subject(s)
Antigens, CD1 , Breast Neoplasms , Glycoproteins , Female , Humans , Antigens, CD1/genetics , Breast , Breast Neoplasms/genetics , Glycoproteins/genetics , Prognosis , Tumor Microenvironment/genetics
4.
Arthritis Rheumatol ; 75(2): 279-292, 2023 02.
Article in English | MEDLINE | ID: mdl-36482877

ABSTRACT

OBJECTIVE: This study was undertaken to identify key disease pathways driving conventional dendritic cell (cDC) alterations in systemic sclerosis (SSc). METHODS: Transcriptomic profiling was performed on peripheral blood CD1c+ cDCs (cDC2s) isolated from 12 healthy donors and 48 patients with SSc, including all major disease subtypes. We performed differential expression analysis for the different SSc subtypes and healthy donors to uncover genes dysregulated in SSc. To identify biologically relevant pathways, we built a gene coexpression network using weighted gene correlation network analysis. We validated the role of key transcriptional regulators using chromatin immunoprecipitation (ChIP) sequencing and in vitro functional assays. RESULTS: We identified 17 modules of coexpressed genes in cDCs that correlated with SSc subtypes and key clinical traits, including autoantibodies, skin score, and occurrence of interstitial lung disease. A module of immunoregulatory genes was markedly down-regulated in patients with the diffuse SSc subtype characterized by severe fibrosis. Transcriptional regulatory network analysis performed on this module predicted nuclear receptor 4A (NR4A) subfamily genes (NR4A1, NR4A2, NR4A3) as the key transcriptional regulators of inflammation. Indeed, ChIP-sequencing analysis indicated that these NR4A members target numerous differentially expressed genes in SSc cDC2s. Inclusion of NR4A receptor agonists in culture-based experiments provided functional proof that dysregulation of NR4As affects cytokine production by cDC2s and modulates downstream T cell activation. CONCLUSION: NR4A1, NR4A2, and NR4A3 are important regulators of immunosuppressive and fibrosis-associated pathways in SSc cDCs. Thus, the NR4A family represents novel potential targets to restore cDC homeostasis in SSc.


Subject(s)
Nuclear Receptor Subfamily 4, Group A, Member 2 , Scleroderma, Systemic , Humans , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Gene Expression Regulation , Gene Expression , Scleroderma, Systemic/genetics , Fibrosis , Glycoproteins/metabolism , Antigens, CD1/genetics
5.
J Biol Chem ; 298(9): 102329, 2022 09.
Article in English | MEDLINE | ID: mdl-35921896

ABSTRACT

Antibodies engage Fc γ receptors (FcγRs) to elicit healing cellular immune responses following binding to a target antigen. Fc γ receptor IIIa/CD16a triggers natural killer cells to destroy target tissues with cytotoxic proteins and enhances phagocytosis mediated by macrophages. Multiple variables affect CD16a antibody-binding strength and the resulting immune response, including a genetic polymorphism. The predominant CD16a F158 allotype binds antibodies with less affinity than the less common V158 allotype. This polymorphism likewise affects cellular immune responses and clinical efficacy of antibodies relying on CD16a engagement, though it remains unclear how V/F158 affects CD16a structure. Another relevant variable shown to affect affinity is composition of the CD16a asparagine-linked (N)-glycans. It is currently not known how N-glycan composition affects CD16a F158 affinity. Here, we determined N-glycan composition affects the V158 and F158 allotypes similarly, and N-glycan composition does not explain differences in V158 and F158 binding affinity. Our analysis of binding kinetics indicated the N162 glycan slows the binding event, and shortening the N-glycans or removing the N162 glycan increased the speed of binding. F158 displayed a slower binding rate than V158. Surprisingly, we found N-glycan composition had a smaller effect on the dissociation rate. We also identified conformational heterogeneity of CD16a F158 backbone amide and N162 glycan resonances using NMR spectroscopy. Residues exhibiting chemical shift perturbations between V158 and F158 mapped to the antibody-binding interface. These data support a model for CD16a F158 with increased interface conformational heterogeneity, reducing the population of binding-competent forms available and decreasing affinity.


Subject(s)
Antibody Affinity , Antigens, CD1 , Polysaccharides , Receptors, IgG , Antigens, CD1/genetics , Antigens, CD1/immunology , Asparagine/genetics , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Polysaccharides/immunology , Receptors, IgG/chemistry , Receptors, IgG/genetics , Receptors, IgG/immunology
6.
Nat Commun ; 13(1): 78, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013257

ABSTRACT

T cells recognize mycobacterial glycolipid (mycolipid) antigens presented by CD1b molecules, but the role of CD4 and CD8 co-receptors in mycolipid recognition is unknown. Here we show CD1b-mycolipid tetramers reveal a hierarchy in which circulating T cells expressing CD4 or CD8 co-receptor stain with a higher tetramer mean fluorescence intensity than CD4-CD8- T cells. CD4+ primary T cells transduced with mycolipid-specific T cell receptors bind CD1b-mycolipid tetramer with a higher fluorescence intensity than CD8+ primary T cells. The presence of either CD4 or CD8 also decreases the threshold for interferon-γ secretion. Co-receptor expression increases surface expression of CD3ε, suggesting a mechanism for increased tetramer binding and activation. Targeted transcriptional profiling of mycolipid-specific T cells from individuals with active tuberculosis reveals canonical markers associated with cytotoxicity among CD8+ compared to CD4+ T cells. Thus, expression of co-receptors modulates T cell receptor avidity for mycobacterial lipids, leading to in vivo functional diversity during tuberculosis disease.


Subject(s)
Antigens, CD1/immunology , Glycolipids/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Antigens, CD1/genetics , CD3 Complex/genetics , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Cytotoxicity, Immunologic , Gene Expression , Glycolipids/metabolism , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Activation , Mycobacterium tuberculosis/growth & development , Primary Cell Culture , Protein Binding , Protein Multimerization , Transduction, Genetic , Tuberculosis/genetics , Tuberculosis/microbiology
7.
PLoS Negl Trop Dis ; 15(12): e0010018, 2021 12.
Article in English | MEDLINE | ID: mdl-34914694

ABSTRACT

T cell receptors (TCRs) encode the history of antigenic challenge within an individual and have the potential to serve as molecular markers of infection. In addition to peptide antigens bound to highly polymorphic MHC molecules, T cells have also evolved to recognize bacterial lipids when bound to non-polymorphic CD1 molecules. One such subset, germline-encoded, mycolyl lipid-reactive (GEM) T cells, recognizes mycobacterial cell wall lipids and expresses a conserved TCR-ɑ chain that is shared among genetically unrelated individuals. We developed a quantitative PCR assay to determine expression of the GEM TCR-ɑ nucleotide sequence in human tissues and blood. This assay was validated on plasmids and T cell lines. We tested blood samples from South African subjects with or without tuberculin reactivity or with active tuberculosis disease. We were able to detect GEM TCR-ɑ above the limit of detection in 92% of donors but found no difference in GEM TCR-ɑ expression among the three groups after normalizing for total TCR-ɑ expression. In a cohort of leprosy patients from Nepal, we successfully detected GEM TCR-ɑ in 100% of skin biopsies with histologically confirmed tuberculoid and lepromatous leprosy. Thus, GEM T cells constitute part of the T cell repertoire in the skin. However, GEM TCR-ɑ expression was not different between leprosy patients and control subjects after normalization. Further, these results reveal the feasibility of developing a simple, field deployable molecular diagnostic based on mycobacterial lipid antigen-specific TCR sequences that are readily detectable in human tissues and blood independent of genetic background.


Subject(s)
Leprosy/diagnosis , Lipids/immunology , Molecular Diagnostic Techniques/methods , Mycobacterium/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Tuberculosis/diagnosis , Antigens, CD1/genetics , Antigens, CD1/immunology , Cell Wall/genetics , Cell Wall/immunology , Cohort Studies , Humans , Leprosy/blood , Leprosy/immunology , Leprosy/microbiology , Mycobacterium/genetics , Mycobacterium/isolation & purification , Nepal , Polymerase Chain Reaction , Receptors, Antigen, T-Cell, alpha-beta/blood , Receptors, Antigen, T-Cell, alpha-beta/genetics , South Africa , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Tuberculosis/blood , Tuberculosis/immunology , Tuberculosis/microbiology
9.
Mol Biol Evol ; 38(6): 2273-2284, 2021 05 19.
Article in English | MEDLINE | ID: mdl-33528563

ABSTRACT

Molecular studies of host-pathogen evolution have largely focused on the consequences of variation at protein-protein interaction surfaces. The potential for other microbe-associated macromolecules to promote arms race dynamics with host factors remains unclear. The cluster of differentiation 1 (CD1) family of vertebrate cell surface receptors plays a crucial role in adaptive immunity through binding and presentation of lipid antigens to T-cells. Although CD1 proteins present a variety of endogenous and microbial lipids to various T-cell types, they are less diverse within vertebrate populations than the related major histocompatibility complex (MHC) molecules. We discovered that CD1 genes exhibit a high level of divergence between simian primate species, altering predicted lipid-binding properties and T-cell receptor interactions. These findings suggest that lipid-protein conflicts have shaped CD1 genetic variation during primate evolution. Consistent with this hypothesis, multiple primate CD1 family proteins exhibit signatures of repeated positive selection at surfaces impacting antigen presentation, binding pocket morphology, and T-cell receptor accessibility. Using a molecular modeling approach, we observe that interspecies variation as well as single mutations at rapidly-evolving sites in CD1a drastically alter predicted lipid binding and structural features of the T-cell recognition surface. We further show that alterations in both endogenous and microbial lipid-binding affinities influence the ability of CD1a to undergo antigen swapping required for T-cell activation. Together these findings establish lipid-protein interactions as a critical force of host-pathogen conflict and inform potential strategies for lipid-based vaccine development.


Subject(s)
Antigens, CD1/genetics , Evolution, Molecular , Lipids/immunology , Models, Molecular , Primates/genetics , Animals , Multigene Family , Primates/immunology , Selection, Genetic
10.
Sci Rep ; 11(1): 2010, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479373

ABSTRACT

The cell wall of Mycobacterium tuberculosis is composed of diverse glycolipids which potentially interact with the human immune system. To overcome difficulties in obtaining pure compounds from bacterial extracts, we recently synthesized three forms of mycobacterial diacyltrehalose (DAT) that differ in their fatty acid composition, DAT1, DAT2, and DAT3. To study the potential recognition of DATs by human T cells, we treated the lipid-binding antigen presenting molecule CD1b with synthetic DATs and looked for T cells that bound the complex. DAT1- and DAT2-treated CD1b tetramers were recognized by T cells, but DAT3-treated CD1b tetramers were not. A T cell line derived using CD1b-DAT2 tetramers showed that there is no cross-reactivity between DATs in an IFN-γ release assay, suggesting that the chemical structure of the fatty acid at the 3-position determines recognition by T cells. In contrast with the lack of recognition of DAT3 by human T cells, DAT3, but not DAT1 or DAT2, activates Mincle. Thus, we show that the mycobacterial lipid DAT can be both an antigen for T cells and an agonist for the innate Mincle receptor, and that small chemical differences determine recognition by different parts of the immune system.


Subject(s)
Antigens, CD1/genetics , Host-Pathogen Interactions/genetics , Trehalose/genetics , Tuberculosis/enzymology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/pathology , Antigens, CD1/chemistry , Antigens, CD1/immunology , Humans , Interferon-gamma/chemistry , Interferon-gamma/genetics , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Lipids/chemistry , Lipids/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Trehalose/chemical synthesis , Trehalose/chemistry , Trehalose/immunology , Tuberculosis/genetics , Tuberculosis/immunology , Tuberculosis/microbiology
11.
Crit Rev Immunol ; 41(4): 1-21, 2021.
Article in English | MEDLINE | ID: mdl-35381140

ABSTRACT

Group 1 CD1-restricted T cells are members of the unconventional T cell family that recognize lipid antigens presented by CD1a, CD1b, and CD1c molecules. Although they developmentally mirror invariant natural killer T cells, they have diverse antigen specificity and functional capacity, with both anti-microbial and autoreactive targets. The role of group 1 CD1-restricted T cells has been best established in Mycobacterium tuberculosis (Mtb) infection in which a wide variety of lipid antigens have been identified and their ability to confer protection against Mtb infection in a CD1 transgenic mouse model has been shown. Group 1 CD1-restricted T cells have also been implicated in other infections, inflammatory conditions, and malignancies. In particular, autoreactive group 1 CD1-restricted T cells have been shown to play a role in several skin inflammatory conditions. The prevalence of group 1 CD1 autoreactive T cells in healthy individuals suggests the presence of regulatory mechanisms to suppress autoreactivity in homeostasis. The more recent use of group 1 CD1 tetramers and mouse models has allowed for better characterization of their phenotype, functional capacity, and underlying mechanisms of antigen-specific and autoreactive activation. These discoveries may pave the way for the development of novel vaccines and immunotherapies that target group 1 CD1-restricted T cells.


Subject(s)
Antigens, CD1 , Natural Killer T-Cells , Animals , Antigen Presentation , Antigens, CD1/genetics , Humans , Immunity , Lipids , Lymphocyte Count , Mice , Mice, Transgenic
12.
Front Immunol ; 11: 566710, 2020.
Article in English | MEDLINE | ID: mdl-33162982

ABSTRACT

Lipids, glycolipids and lipopeptides derived from Mycobacterium tuberculosis (Mtb) are presented to T cells by monomorphic molecules known as CD1. This is the case of the Mtb-specific sulfoglycolipid Ac2SGL, which is presented by CD1b molecules and is recognized by T cells found in tuberculosis (TB) patients and in individuals with latent infections. Our group, using filamentous phage display technology, obtained two specific ligands against the CD1b-Ac2SGL complex: (i) a single chain T cell receptor (scTCR) from a human T cell clone recognizing the CD1b-AcSGL complex; and (ii) a light chain domain antibody (dAbκ11). Both ligands showed lower reactivity to a synthetic analog of Ac2SGL (SGL12), having a shorter acyl chain as compared to the natural antigen. Here we put forward the hypothesis that the CD1b endogenous spacer lipid (EnSpacer) plays an important role in the recognition of the CD1b-Ac2SGL complex by specific T cells. To support this hypothesis we combined: (a) molecular binding assays for both the scTCR and the dAbκ11 antibody domain against a small panel of synthetic Ac2SGL analogs having different acyl chains, (b) molecular modeling of the CD1b-Ac2SGL/EnSpacer complex, and (c) modeling of the interactions of this complex with the scTCR. Our results contribute to understand the mechanisms of lipid presentation by CD1b molecules and their interactions with T-cell receptors and other specific ligands, which may help to develop specific tools targeting Mtb infected cells for therapeutic and diagnostic applications.


Subject(s)
Antigens, Bacterial/immunology , Antigens, CD1/immunology , Models, Molecular , Mycobacterium tuberculosis/immunology , Receptors, Antigen, T-Cell/immunology , Antigens, CD1/genetics , Humans , Recombinant Proteins/immunology
13.
EBioMedicine ; 55: 102774, 2020 May.
Article in English | MEDLINE | ID: mdl-32403085

ABSTRACT

BACKGROUND: Since numerous pathological conditions are evoked by unwanted dendritic cell (DC) activity, therapeutic agents modulating DC functions are of great medical interest. In regenerative medicine, cellular secretomes have gained increasing attention and valuable immunomodulatory properties have been attributed to the secretome of γ-irradiated peripheral blood mononuclear cells (PBMCs). Potential effects of the PBMC secretome (PBMCsec) on key DC functions have not been elucidated so far. METHODS: We used a hapten-mediated murine model of contact hypersensitivity (CH) to study the effects of PBMCsec on DCs in vivo. Effects of PBMCsec on human DCs were investigated in monocyte-derived DCs (MoDC) and ex vivo skin cultures. DCs were phenotypically characterised by transcriptomics analyses and flow cytometry. DC function was evaluated by cytokine secretion, antigen uptake, PBMC proliferation and T-cell priming. FINDINGS: PBMCsec significantly alleviated tissue inflammation and cellular infiltration in hapten-sensitized mice. We found that PBMCsec abrogated differentiation of MoDCs, indicated by lower expression of classical DC markers CD1a, CD11c and MHC class II molecules. Furthermore, PBMCsec reduced DC maturation, antigen uptake, lipopolysaccharides-induced cytokine secretion, and DC-mediated immune cell proliferation. Moreover, MoDCs differentiated with PBMCsec displayed diminished ability to prime naïve CD4+T-cells into TH1 and TH2 cells. Furthermore, PBMCsec modulated the phenotype of DCs present in the skin in situ. Mechanistically, we identified lipids as the main biomolecule accountable for the observed immunomodulatory effects. INTERPRETATION: Together, our data describe DC-modulatory actions of lipids secreted by stressed PBMCs and suggest PBMCsec as a therapeutic option for treatment of DC-mediated inflammatory skin conditions. FUNDING: This research project was supported by the Austrian Research Promotion Agency (Vienna, Austria; grant "APOSEC" 862068; 2015-2019) and the Vienna Business Agency (Vienna, Austria; grant "APOSEC to clinic" 2343727).


Subject(s)
Culture Media, Conditioned/chemistry , Dendritic Cells/radiation effects , Dermatitis, Contact/therapy , Immunologic Factors/pharmacology , Lipids/pharmacology , Skin/radiation effects , Adult , Animals , Antigens, CD1/genetics , Antigens, CD1/immunology , Biomarkers/analysis , CD11c Antigen/genetics , CD11c Antigen/immunology , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Dermatitis, Contact/etiology , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Dinitrofluorobenzene/administration & dosage , Female , Gamma Rays , Gene Expression , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Immunologic Factors/isolation & purification , Lipids/isolation & purification , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Monocytes/radiation effects , Primary Cell Culture , Skin/immunology , Skin/pathology , Th1 Cells/cytology , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/cytology , Th2 Cells/drug effects , Th2 Cells/immunology , Tissue Culture Techniques
14.
Hum Immunol ; 81(7): 361-365, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32467040

ABSTRACT

CD1 glycoproteins are a class of antigen presenting molecules that bind and present non-peptidic antigens (lipids and glycolipids) for immune recognition. CD1 polymorphisms, although limited, could have a critical role in antimicrobial, anticancer, and autoimmune responses and disease susceptibility. Ethnic differences and interactions between genetic and environmental factors make it attractive the study of these molecules in autoimmune inflammatory disorders, such as celiac disease (CD), in which a strong genetic predisposition (HLA-DQ2/DQ8) and pressure of environmental factors have a central role. CD1A, CD1D and CD1E polymorphisms in exon 2 were assessed in patients from Morocco affected by CD, using direct sequencing analysis, in order to investigate possible associations with the disease in a North African population. Differences in genotype and haplotype distribution of CD1E between celiac patients and controls were found: in particular, an increase of CD1E*02/02 homozygous (OR 2.93, CI 1.30-6.59, p = 0.007) and CD1A*02-E*02 estimated haplotypes in CD, compared with controls. Frequencies of CD1A and CD1D genotypes/alleles were not different between groups. CD1E*02/02, previously suggested as a potential immune protective genotype to malaria susceptibility, could be an additional gene involved in celiac risk in this geographic area.


Subject(s)
Antigens, CD1/genetics , Celiac Disease/epidemiology , Celiac Disease/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Genetic , Adolescent , Adult , Aged , Exons , Female , Gene Frequency , Genetic Testing/methods , Haplotypes , Homozygote , Humans , Male , Middle Aged , Morocco/epidemiology , Risk Factors , Young Adult
15.
PLoS Pathog ; 16(4): e1008443, 2020 04.
Article in English | MEDLINE | ID: mdl-32343740

ABSTRACT

Staphylococcus aureus (SA) is the causative agent of both skin/soft tissue infections as well as invasive bloodstream infections. Though vaccines have been developed to target both humoral and T cell-mediated immune responses against SA, they have largely failed due to lack of protective efficacy. Group 1 CD1-restricted T cells recognize lipid rather than peptide antigens. Previously found to recognize lipids derived from cell wall of Mycobacterium tuberculosis (Mtb), these cells were associated with protection against Mtb infection in humans. Using a transgenic mouse model expressing human group 1 CD1 molecules (hCD1Tg), we demonstrate that group 1 CD1-restricted T cells can recognize SA-derived lipids in both immunization and infection settings. Systemic infection of hCD1Tg mice showed that SA-specific group 1 CD1-restricted T cell response peaked at 10 days post-infection, and hCD1Tg mice displayed significantly decreased kidney pathology at this time point compared with WT control mice. Immunodominant SA lipid antigens recognized by group 1 CD1-restricted T cells were comprised mainly of cardiolipin and phosphatidyl glycerol, with little contribution from lysyl-phosphatidyl glycerol which is a unique bacterial lipid not present in mammals. Group 1 CD1-restricted T cell lines specific for SA lipids also conferred protection against SA infection in the kidney after adoptive transfer. They were further able to effectively control SA replication in vitro through direct antigen presentation by group 1 CD1-expressing BMDCs. Together, our data demonstrate a previously unknown role for group 1 CD1-restricted SA lipid-specific T cells in the control of systemic MRSA infection.


Subject(s)
Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD1/genetics , Antigens, CD1/immunology , Humans , Immunization , Kidney/immunology , Kidney/microbiology , Lipids/immunology , Mice , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology
16.
Biomed Res Int ; 2020: 4360930, 2020.
Article in English | MEDLINE | ID: mdl-32258122

ABSTRACT

The involvement of immune dysfunction in the pathogenesis of lung cancer has been extensively studied. However, the potential molecular mechanisms through which the tumor immune response affects drug resistance are still unclear. Accordingly, in this study, we evaluated deviations in the immune cell landscape among patients with different stages of lung adenocarcinoma to identify key microRNAs and their targets associated with patient outcomes. CIBERSORT was used for estimating the proportions of immune cells in various lung tissues. Significantly different adaptive and innate immune cell types, including memory B cells, CD8+ T cells, resting dendritic cells, and resting mast cells, were selected. Comparative studies and survival analyses were carried out. We found that potential genes and microRNAs involved in immune responses were associated with patient outcomes. Specifically, miR-582/CD1B, which are involved in resting and activated dendritic cells, may be potential novel biomarkers for immunotherapy. An independent dataset of miRNA microarray profiles was used to validate the expression of mature miR-582-5p in patients with advanced lung adenocarcinoma. Alternative treatments, including immunotherapies and chemotherapy, are urgently needed to improve outcomes in patients with lung cancer. Thus, our findings could provide insights into the selection of novel microRNAs targeting immune genes and could improve the efficacy of immunotherapy by disrupting tumor function and promoting immune infiltration in patients with advanced lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung/genetics , Antigens, CD1/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/pathology , Biomarkers, Tumor/genetics , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Resistance, Neoplasm/genetics , Female , Humans , Immunotherapy/adverse effects , Male , Neoplasm Staging , Progression-Free Survival , Signal Transduction/genetics , Treatment Outcome
17.
Blood Adv ; 4(1): 87-99, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31899802

ABSTRACT

Langerhans cell histiocytosis (LCH) is a myeloproliferative disorder that is characterized by the inflammatory lesions with pathogenic CD1a+CD207+ dendritic cells (DCs). BRAFV600E and other somatic activating MAPK gene mutations have been identified in differentiating bone marrow and blood myeloid cells, but the origin of the LCH lesion CD1a+CD207+ DCs and mechanisms of lesion formation remain incompletely defined. To identify candidate LCH CD1a+CD207+ DC precursor populations, gene-expression profiles of LCH lesion CD1a+CD207+ DCs were first compared with established gene signatures from human myeloid cell subpopulations. Interestingly, the CD1c+ myeloid DC (mDC) gene signature was most enriched in the LCH CD1a+CD207+ DC transcriptome. Additionally, the BRAFV600E allele was not only localized to CD1a+CD207- DCs and CD1a+CD207+ DCs, but it was also identified in CD1c+ mDCs in LCH lesions. Transcriptomes of CD1a+CD207- DCs were nearly indistinguishable from CD1a+CD207+ DCs (both CD1a+CD207low and CD1a+CD207high subpopulations). Transcription profiles of LCH lesion CD1a+CD207+ DCs and peripheral blood CD1c+ mDCs from healthy donors were compared to identify potential LCH DC-specific biomarkers: HLA-DQB2 expression was significantly increased in LCH lesion CD1a+CD207+ DCs compared with circulating CD1c+ mDCs from healthy donors. HLA-DQB2 antigen was identified on LCH lesion CD1a+CD207- DCs and CD1a+CD207+ DCs as well as on CD1c+(CD1a+CD207-) mDCs, but it was not identified in any other lesion myeloid subpopulations. HLA-DQB2 expression was specific to peripheral blood of patients with BRAFV600E+ peripheral blood mononuclear cells, and HLA-DQB2+CD1c+ blood cells were highly enriched for the BRAFV600E in these patients. These data support a model in which blood CD1c+HLA-DQB2+ mDCs with activated ERK migrate to lesion sites where they differentiate into pathogenic CD1a+CD207+ DCs.


Subject(s)
Histiocytosis, Langerhans-Cell , Leukocytes, Mononuclear , Antigens, CD/genetics , Antigens, CD1/genetics , Biomarkers , Dendritic Cells , Glycoproteins , Histiocytosis, Langerhans-Cell/diagnosis , Histiocytosis, Langerhans-Cell/genetics , Humans , Lectins, C-Type/genetics , Mannose-Binding Lectins/genetics , Myeloid Cells
18.
Proc Natl Acad Sci U S A ; 116(44): 22262-22268, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611396

ABSTRACT

Interleukin-4 (IL-4) is produced by a unique subset of invariant natural killer T (iNKT) cells (NKT2) in the thymus in the steady state, where it conditions CD8+ T cells to become "memory-like" among other effects. However, the signals that cause NKT2 cells to constitutively produce IL-4 remain poorly defined. Using histocytometry, we observed IL-4-producing NKT2 cells localized to the thymic medulla, suggesting that medullary signals might instruct NKT2 cells to produce IL-4. Moreover, NKT2 cells receive and require T cell receptor (TCR) stimulation for continuous IL-4 production in the steady state, since NKT2 cells lost IL-4 production when intrathymically transferred into CD1d-deficient recipients. In bone marrow chimeric recipients, only hematopoietic, not stromal, antigen-presenting cells (APCs), provided such stimulation. Furthermore, using different Cre-recombinase transgenic mouse strains to specifically target CD1d deficiency to various APCs, together with the use of diphtheria toxin receptor (DTR) transgenic mouse strains to deplete various APCs, we found that macrophages were the predominant cell to stimulate NKT2 IL-4 production. Thus, NKT2 cells appear to encounter and require different activating ligands for selection in the cortex and activation in the medulla.


Subject(s)
Interleukin-4/metabolism , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Thymus Gland/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens, CD1/genetics , Antigens, CD1/metabolism , Cells, Cultured , Interleukin-4/genetics , Lymphocyte Activation , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Thymus Gland/cytology
19.
J Immunol ; 203(7): 1882-1896, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31492741

ABSTRACT

Comparative analyses suggest that the MHC was derived from a prevertebrate "primordial immune complex" (PIC). PIC duplicated twice in the well-studied two rounds of genome-wide duplications (2R) early in vertebrate evolution, generating four MHC paralogous regions (predominantly on human chromosomes [chr] 1, 6, 9, 19). Examining chiefly the amphibian Xenopus laevis, but also other vertebrates, we identified their MHC paralogues and mapped MHC class I, AgR, and "framework" genes. Most class I genes mapped to MHC paralogues, but a cluster of Xenopus MHC class Ib genes (xnc), which previously was mapped outside of the MHC paralogues, was surrounded by genes syntenic to mammalian CD1 genes, a region previously proposed as an MHC paralogue on human chr 1. Thus, this gene block is instead the result of a translocation that we call the translocated part of the MHC paralogous region (MHCtrans) Analyses of Xenopus class I genes, as well as MHCtrans, suggest that class I arose at 1R on the chr 6/19 ancestor. Of great interest are nonrearranging AgR-like genes mapping to three MHC paralogues; thus, PIC clearly contained several AgR precursor loci, predating MHC class I/II. However, all rearranging AgR genes were found on paralogues derived from the chr 19 precursor, suggesting that invasion of a variable (V) exon by the RAG transposon occurred after 2R. We propose models for the evolutionary history of MHC/TCR/Ig and speculate on the dichotomy between the jawless (lamprey and hagfish) and jawed vertebrate adaptive immune systems, as we found genes related to variable lymphocyte receptors also map to MHC paralogues.


Subject(s)
Antigens, CD1/genetics , Databases, Genetic , Histocompatibility Antigens Class I/genetics , Xenopus Proteins/genetics , Animals , Antigens, CD1/immunology , Histocompatibility Antigens Class I/immunology , Xenopus Proteins/immunology , Xenopus laevis
20.
Mol Genet Genomic Med ; 7(10): e00960, 2019 10.
Article in English | MEDLINE | ID: mdl-31464097

ABSTRACT

BACKGROUND: Guillain-Barre Syndrome (GBS) is considered a complex disorder with significant environmental effect and genetic susceptibility. Genetic polymorphisms in CD1E, CD1A, IL-17, and/or ICAM1 had been proposed as susceptibility genetic variants for GBS mainly in Caucasian population. This study explores the association between selected polymorphisms in these genes and GBS susceptibility in confirmed GBS cases reported in mestizo population from northern Peru during the most recent GBS outbreak of May 2018. METHODS: A total of nine nonrelated cases and 11 controls were sequenced for the polymorphic regions of CD1A, CD1E, IL-17, and ICAM1. RESULTS: We found a significant protective association between heterozygous GA genotype in ICAM1 (241Gly/Arg) and GBS (p < .047). IL-17 was monomorphic in both controls and patients. No significant differences were found in the frequency of SNPs in CD1A and CD1E between the group with GBS patients and healthy controls. CONCLUSION: ICAM1 polymorphisms might be considered as potential genetic markers of GBS susceptibility. Further studies with larger sample size will be required to validate these findings.


Subject(s)
Antigens, CD1/genetics , Guillain-Barre Syndrome/pathology , Intercellular Adhesion Molecule-1/genetics , Interleukin-17/genetics , Aged , Body Mass Index , Case-Control Studies , Female , Genetic Predisposition to Disease , Genotype , Guillain-Barre Syndrome/genetics , Humans , Male , Middle Aged , Peru , Polymorphism, Single Nucleotide , Regression Analysis , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL