Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 694
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 21(10): 1232-1243, 2020 10.
Article in English | MEDLINE | ID: mdl-32929275

ABSTRACT

The CD2-CD58 recognition system promotes adhesion and signaling and counters exhaustion in human T cells. We found that CD2 localized to the outer edge of the mature immunological synapse, with cellular or artificial APC, in a pattern we refer to as a 'CD2 corolla'. The corolla captured engaged CD28, ICOS, CD226 and SLAM-F1 co-stimulators. The corolla amplified active phosphorylated Src-family kinases (pSFK), LAT and PLC-γ over T cell receptor (TCR) alone. CD2-CD58 interactions in the corolla boosted signaling by 77% as compared with central CD2-CD58 interactions. Engaged PD-1 invaded the CD2 corolla and buffered CD2-mediated amplification of TCR signaling. CD2 numbers and motifs in its cytoplasmic tail controlled corolla formation. CD8+ tumor-infiltrating lymphocytes displayed low expression of CD2 in the majority of people with colorectal, endometrial or ovarian cancer. CD2 downregulation may attenuate antitumor T cell responses, with implications for checkpoint immunotherapies.


Subject(s)
CD2 Antigens/metabolism , CD58 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Immunological Synapses/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , Cell Adhesion , Cells, Cultured , Humans , Immune Tolerance , Lymphocyte Activation , Protein Binding , Receptor Cross-Talk , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Single-Cell Analysis
2.
J Immunol ; 212(2): 245-257, 2024 01 15.
Article in English | MEDLINE | ID: mdl-38047900

ABSTRACT

CD8 T cells are emerging as important mediators in atherosclerosis and cardiovascular disease (CVD). Immune activation may play a particular role in people with HIV (PWH) who are at an increased risk of CVD, even after controlling for known CVD risk factors. Latent CMV infection is associated with increased CVD risk for both PWH and people without HIV, and human CMV-specific CD4 and CD8 T cells are enriched for an immunosenescent phenotype. We previously showed that CMV coinfection in PWH promotes vascular homing and activation of inflammatory CD4 T cells through the CD2-LFA-3 axis. However, the role of CD2/LFA3 costimulation of CD8 T cells in PWH with CMV has yet to be described. In the present study, we demonstrate that CD2 expression on CX3CR1+CD57+CD28- inflammescent CD8 T cells is increased on cells from CMV-seropositive PWH. In vitro CD2/LFA-3 costimulation enhances TCR-mediated activation of these inflammatory CD8 memory T cells. Finally, we show that LFA-3 is highly expressed in aortas of SIV-infected rhesus macaques and in atherosclerotic plaques of people without HIV. Our findings are consistent with a model in which CMV infection enhances CD2 expression on highly proinflammatory CD8 T cells that can then be stimulated by LFA-3 expressed in the vasculature, even in the absence of CD28 costimulation. This model, in which CMV infection exacerbates toxic cytokine and granzyme production by CD8 T cells within the vasculature, highlights a potential therapeutic target in atherosclerosis development and progression, especially for PWH.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Cytomegalovirus Infections , HIV Infections , Animals , Humans , CD28 Antigens/metabolism , HIV Infections/drug therapy , Cytomegalovirus , CD58 Antigens/metabolism , Macaca mulatta , CD8-Positive T-Lymphocytes , CD4-Positive T-Lymphocytes , Atherosclerosis/metabolism
3.
Hum Genet ; 143(1): 19-33, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37994973

ABSTRACT

CD58 plays roles in cell adhesion and co-stimulation with antigen presentation from major histocompatibility complex class II on antigen-presenting cells to T-cell antigen receptors on naïve T cells. CD58 reportedly contributes to the development of various human autoimmune diseases. Recently, genome-wide association studies (GWASs) identified CD58 as a susceptibility locus for autoimmune diseases such as systemic lupus erythematosus (SLE), multiple sclerosis (MS), and primary biliary cholangitis (PBC). However, the primary functional variant and molecular mechanisms of susceptibility to autoimmune diseases in the CD58 locus were not clarified. Here, rs10924104, located in the ZNF35-binding motif within the gene expression regulatory motif, was identified as the primary functional variant for SLE, MS, and PBC among genetic variants showing stronger linkage disequilibrium (LD) with GWAS-lead variants in the CD58 locus. Expression-quantitative trait locus (e-QTL) data for each distinct blood cell type and in vitro functional analysis using the CRISPR/Cas9 system corroborated the functional role of rs10924104 in the upregulation of CD58 transcription by the disease-risk allele. Additionally, the strength of disease susceptibility observed in the CD58 locus could be accounted for by the strength of LD between rs10924104 and each GWAS-lead variant. In conclusion, the present study demonstrated for the first time the existence of a shared autoimmune disease-related primary functional variant (i.e., rs10924104) that regulates the expression of CD58. Clarifying the molecular mechanism of disease susceptibility derived from such a shared genetic background is important for understanding human autoimmune diseases and human immunology.


Subject(s)
Autoimmune Diseases , Lupus Erythematosus, Systemic , Multiple Sclerosis , Humans , Autoimmune Diseases/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Lupus Erythematosus, Systemic/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Polymorphism, Single Nucleotide , CD58 Antigens/metabolism
4.
J Transl Med ; 21(1): 539, 2023 08 12.
Article in English | MEDLINE | ID: mdl-37573318

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies worldwide because of rapid progression and high incidence of metastasis or recurrence. Accumulating evidence shows that CD58-expressing tumor cell is implicated in development of various cancers. The present study aimed to reveal the functional significance of CD58 in HCC progression and the underlying mechanisms. METHODS: Immunohistochemical staining (IHC), and western blotting were used to detect the expression of CD58 in HCC tissues and cells. The levels of sCD58 (a soluble form of CD58) in the cell supernatants and serum were assessed by ELISA. CCK-8, colony formation, and xenograft assays were used to detect the function of CD58 on proliferation in vitro and in vivo. Transwell assay and sphere formation assay were performed to evaluate the effect of CD58 and sCD58 on metastasis and self-renewal ability of HCC cells. Western blotting, immunofluorescence (IF), TOP/FOP Flash reporter assay, and subcellular fractionation assay were conducted to investigate the molecular regulation between CD58/sCD58 and AKT/GSK-3ß/ß-catenin axis in HCC cells. RESULTS: CD58 was significantly upregulated in HCC tissues. Elevation of CD58 expression correlated with more satellite foci and vascular invasion, and poorer tumor-free and overall survival in HCC patients. Higher sCD58 levels were in HCC patients' serum compared to healthy individuals. Functionally, CD58 promotes the proliferation of HCC cells in vitro and in vivo. Meanwhile, CD58 and sCD58 induce metastasis, self-renewal and pluripotency in HCC cells in vitro. Mechanistically, CD58 activates the AKT/GSK-3ß/ß-catenin signaling pathway by increasing phosphorylation of AKT or GSK3ß signaling, promoting expression of Wnt/ß-catenin target proteins and TCF/LEF-mediated transcriptional activity. Furthermore, AKT activator SC-79 or inhibitor LY294002 abolished the inhibitory effect of CD58 silencing on the proliferation, metastasis, and stemness of HCC cells. CONCLUSIONS: Taken together, CD58 promotes HCC progression and metastasis via activating the AKT/GSK-3ß/ß-catenin pathway, suggesting that CD58 is a novel prognostic biomarker and therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , beta Catenin/metabolism , Carcinogens , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Glycogen Synthase Kinase 3 beta , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , CD58 Antigens/metabolism
5.
Molecules ; 28(19)2023 Sep 27.
Article in English | MEDLINE | ID: mdl-37836662

ABSTRACT

Human cell surface receptor CD58, also known as lymphocyte function-associated antigen 3 (LFA-3), plays a critical role in the early stages of immune response through interacting with CD2. Recent research identified CD58 as a surface marker of colorectal cancer (CRC), which can upregulate the Wnt pathway and promote self-renewal of colorectal tumor-initiating cells (CT-ICs) by degradation of Dickkopf 3. In addition, it was also shown that knockdown of CD58 significantly impaired tumor growth. In this study, we developed a structure-based virtual screening pipeline using Autodock Vina and binding analysis and identified a group of small molecular compounds having the potential to bind with CD58. Five of them significantly inhibited the growth of the SW620 cell line in the following in vitro studies. Their proposed binding models were further verified by molecular dynamics (MD) simulations, and some pharmaceutically relevant chemical and physical properties were predicted. The hits described in this work may be considered interesting leads or structures for the development of new and more efficient CD58 inhibitors.


Subject(s)
Colorectal Neoplasms , Early Detection of Cancer , Humans , CD2 Antigens/chemistry , CD2 Antigens/metabolism , CD58 Antigens/chemistry , CD58 Antigens/metabolism , Molecular Dynamics Simulation , Colorectal Neoplasms/drug therapy
6.
Blood ; 136(21): 2437-2441, 2020 11 19.
Article in English | MEDLINE | ID: mdl-32589698

ABSTRACT

A unique feature of Hodgkin lymphoma (HL) is the presence of CD4+ T cells that surround, protect, and promote survival of tumor cells. The adhesion molecules involved in this so-called T-cell rosetting are important components of the immunological synapse (IS). However, it is unknown whether this synapse is fully assembled and leads to T-cell activation by enabling interaction between the T-cell receptor (TCR) and human leukocyte antigen class II (HLA-II). We established a novel rosetting model by coculturing HLA-II-matched peripheral blood mononuclear cells with HL cell lines and showed IS formation with activation of rosetting T cells. HLA-II downregulation by class II transactivator knockout did not affect the extent of rosetting, but almost completely abrogated T-cell activation. Intriguingly, the level of CD58 expression correlated with the extent of rosette formation, and CD58 knockout or CD2 blockade reduced both rosette formation and T-cell activation. The extension of our findings to primary HL tissue by immunohistochemistry and proximity ligation assays showed interaction of CD2 with CD58 and of TCR-associated CD4 with HLA-II. In conclusion, T-cell rosetting in HL is established by formation of the IS, and activation of rosetting T cells critically depends on the interaction of both TCR-HLA-II and CD2-CD58.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD58 Antigens/immunology , Histocompatibility Antigens Class II/immunology , Hodgkin Disease/immunology , Immunological Synapses/immunology , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/immunology , Rosette Formation , CD2 Antigens/antagonists & inhibitors , CD58 Antigens/biosynthesis , CD58 Antigens/genetics , CRISPR-Cas Systems , Cell Adhesion , Cell Line, Tumor , Coculture Techniques , Gene Knockout Techniques , Humans , Protein Interaction Mapping
7.
J Pharmacol Sci ; 149(3): 124-138, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35641025

ABSTRACT

Protein-protein interactions (PPI) of co-stimulatory molecules CD2-CD58 are important in the early stage of an immune response, and increased expression of these co-stimulatory molecules is observed in the synovial region of joints in rheumatoid arthritis (RA) patients. A CD2 epitope region that binds to CD58 was grafted on to sunflower trypsin inhibitor (SFTI) template structure to inhibit CD2-CD58 PPI. The peptide was incorporated with an organic moiety dibenzofuran (DBF) in its structure. The designed peptidomimetic was studied for its ability to inhibit CD2-CD58 interactions in vitro, and its thermal and enzymatic stability was evaluated. Stability studies indicated that the grafted peptidomimetic was stable against trypsin cleavage. In vivo studies using the collagen-induced arthritis (CIA) model in mice indicated that the peptidomimetic was able to slow down the progress of arthritis, an autoimmune disease in the mice model. These studies suggest that with the grafting of organic functional groups in the stable peptide template SFTI stabilizes the peptide structure, and these peptides can be used as a template to design stable peptides for therapeutic purposes.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Helianthus , Peptidomimetics , Animals , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , CD58 Antigens/chemistry , CD58 Antigens/metabolism , Helianthus/chemistry , Helianthus/metabolism , Humans , Immunity , Immunomodulation , Mice , Peptides/pharmacology , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacology , Peptidomimetics/pharmacology , Peptidomimetics/therapeutic use , Trypsin Inhibitors/therapeutic use
8.
J Immunol ; 204(10): 2722-2733, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32229536

ABSTRACT

Cytotoxic CD4 T cells are linked to cardiovascular morbidities and accumulate in both HIV and CMV infections, both of which are associated with increased risk of cardiovascular disease (CVD). In this study, we identify CMV coinfection as a major driver of the cytotoxic phenotype, characterized by elevated CD57 expression and reduced CD28 expression, in circulating CD4 T cells from people living with HIV infection, and investigate potential mechanisms linking this cell population to CVD. We find that human CD57+ CD4 T cells express high levels of the costimulatory receptor CD2 and that CD2/LFA-3 costimulation results in a more robust and polyfunctional effector response to TCR signals, compared with CD28-mediated costimulation. CD57+ CD4 T cells also express the vascular endothelium-homing receptor CX3CR1 and migrate toward CX3CL1-expressing endothelial cells in vitro. IL-15 promotes the cytotoxic phenotype, elevates CX3CR1 expression, and enhances the trafficking of CD57+ CD4 T cells to endothelium and may therefore be important in linking these cells to cardiovascular complications. Finally, we demonstrate the presence of activated CD57+ CD4 T cells and expression of CX3CL1 and LFA-3 in atherosclerotic plaque tissues from HIV-uninfected donors. Our findings are consistent with a model in which cytotoxic CD4 T cells contribute to CVD in HIV/CMV coinfection and in atherosclerosis via CX3CR1-mediated trafficking and CD2/LFA-3-mediated costimulation. This study identifies several targets for therapeutic interventions and may help bridge the gap in understanding how CMV infection and immunity are linked to increased cardiovascular risk in people living with HIV infection.


Subject(s)
Blood Vessels/physiology , CD4-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/physiology , HIV Infections/immunology , HIV-1/physiology , Plaque, Atherosclerotic/immunology , CD28 Antigens/metabolism , CD57 Antigens/metabolism , CD58 Antigens/metabolism , CX3C Chemokine Receptor 1/metabolism , Cell Movement , Chemokine CX3CL1/metabolism , Coinfection , Cytotoxicity, Immunologic , Humans , Receptors, CXCR3/metabolism , Risk
9.
PLoS Genet ; 15(2): e1007961, 2019 02.
Article in English | MEDLINE | ID: mdl-30730892

ABSTRACT

Genome-wide association studies have identified more than 200 genetic variants to be associated with an increased risk of developing multiple sclerosis (MS). Still, little is known about the causal molecular mechanisms that underlie the genetic contribution to disease susceptibility. In this study, we investigated the role of the single-nucleotide polymorphism (SNP) rs1414273, which is located within the microRNA-548ac stem-loop sequence in the first intron of the CD58 gene. We conducted an expression quantitative trait locus (eQTL) analysis based on public RNA-sequencing and microarray data of blood-derived cells of more than 1000 subjects. Additionally, CD58 transcripts and mature hsa-miR-548ac molecules were measured using real-time PCR in peripheral blood samples of 32 MS patients. Cell culture experiments were performed to evaluate the efficiency of Drosha-mediated stem-loop processing dependent on genotype and to determine the target genes of this underexplored microRNA. Across different global populations and data sets, carriers of the MS risk allele showed reduced CD58 mRNA levels but increased hsa-miR-548ac levels. We provide evidence that the SNP rs1414273 might alter Drosha cleavage activity, thereby provoking partial uncoupling of CD58 gene expression and microRNA-548ac production from the shared primary transcript in immune cells. Moreover, the microRNA was found to regulate genes, which participate in inflammatory processes and in controlling the balance of protein folding and degradation. We thus uncovered new regulatory implications of the MS-associated haplotype of the CD58 gene locus, and we remind that paradoxical findings can be encountered in the analysis of eQTLs upon data aggregation. Our study illustrates that a better understanding of RNA processing events might help to establish the functional nature of genetic variants, which predispose to inflammatory and neurological diseases.


Subject(s)
CD58 Antigens/genetics , MicroRNAs/genetics , Multiple Sclerosis/genetics , Polymorphism, Single Nucleotide , CD58 Antigens/metabolism , Cohort Studies , Computer Simulation , Female , Genetic Association Studies , Genome-Wide Association Study , HeLa Cells , Humans , Introns , Male , MicroRNAs/chemistry , MicroRNAs/metabolism , Middle Aged , Models, Genetic , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Nucleic Acid Conformation , Quantitative Trait Loci , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
J Virol ; 94(5)2020 02 14.
Article in English | MEDLINE | ID: mdl-31801856

ABSTRACT

The human cytomegalovirus (HCMV) endoplasmic reticulum (ER)-resident glycoprotein UL148 is posited to play roles in immune evasion and regulation of viral cell tropism. UL148 prevents cell surface presentation of the immune cell costimulatory ligand CD58 while promoting maturation and virion incorporation of glycoprotein O, a receptor binding subunit for an envelope glycoprotein complex involved in entry. Meanwhile, UL148 activates the unfolded protein response (UPR) and causes large-scale reorganization of the ER. In order to determine whether the seemingly disparate effects of UL148 are related or discrete, we generated six charged cluster-to-alanine (CCTA) mutants within the UL148 ectodomain and compared them to wild-type UL148, both in the context of infection studies using recombinant viruses and in ectopic expression experiments, assaying for effects on ER remodeling and CD58 surface presentation. Two mutants, targeting charged clusters spanning residues 79 to 83 (CC3) and 133 to 136 (CC4), retained the potential to impede CD58 surface presentation. Of the six mutants, only CC3 retained the capacity to reorganize the ER, but it showed a partial phenotype. Wild-type UL148 accumulates in a detergent-insoluble form during infection. However, all six CCTA mutants were fully soluble, which implies a relationship between insolubility and organelle remodeling. Additionally, we found that the chimpanzee cytomegalovirus UL148 homolog suppresses surface presentation of CD58 but fails to reorganize the ER, while the homolog from rhesus cytomegalovirus shows neither activity. Collectively, our findings illustrate various degrees of functional divergence between homologous primate cytomegalovirus immunevasins and suggest that the capacity to cause ER reorganization is unique to HCMV UL148.IMPORTANCE In myriad examples, viral gene products cause striking effects on cells, such as activation of stress responses. It can be challenging to decipher how such effects contribute to the biological roles of the proteins. The HCMV glycoprotein UL148 retains CD58 within the ER, thereby preventing it from reaching the cell surface, where it functions to stimulate cell-mediated antiviral responses. Intriguingly, UL148 also triggers the formation of large, ER-derived membranous structures and activates the UPR, a set of signaling pathways involved in adaptation to ER stress. We demonstrate that the potential of UL148 to reorganize the ER and to retain CD58 are separable by mutagenesis and, possibly, by evolution, since chimpanzee cytomegalovirus UL148 retains CD58 but does not remodel the ER. Our findings imply that ER reorganization contributes to other roles of UL148, such as modulation of alternative viral glycoprotein complexes that govern the virus' ability to infect different cell types.


Subject(s)
CD58 Antigens/metabolism , Cytomegalovirus/physiology , Endoplasmic Reticulum/metabolism , Glycoproteins/metabolism , Viral Fusion Proteins/metabolism , Amino Acid Sequence , Cytomegalovirus/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum Stress , Humans , Immune Evasion , Unfolded Protein Response , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/genetics , Viral Tropism
11.
Cancer Invest ; 39(10): 854-870, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34569407

ABSTRACT

The novel anti-neoplastic glycopeptide T11TS retards glioma both in in-vitro clinical samples and in-vivo models. This study investigates the correlation between altering the glioma microenvironment with glioma arrest and death. Flow cytometry, immunoblotting, ELISA, and co-immunoprecipitation were employed to investigate glioma cell arrest and death. Results include a decline in phosphorylation of Akt and attenuation of p21 phosphorylation (Thr145,Ser146) and disassociation of p-Akt-Mdm2 and p-Akt-BAD facilitating death by Akt>BAD. T11TS influence phosphorylation patterns in two focal axes Akt>p21 and Akt>Mdm2>p53. The current article provides crucial insight in deciphering the mechanism of T11TS induced glioma cell arrest and death.


Subject(s)
Brain Neoplasms/drug therapy , CD58 Antigens/pharmacology , Glioma/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , CD58 Antigens/therapeutic use , Cell Cycle Checkpoints/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Glioma/metabolism , Glioma/pathology , Male , PTEN Phosphohydrolase/analysis , Phosphorylation , Proto-Oncogene Proteins c-mdm2/analysis , Rats , Rats, Wistar , Tumor Microenvironment , Tumor Suppressor Protein p53/analysis , bcl-Associated Death Protein/metabolism
12.
J Comput Aided Mol Des ; 35(3): 337-353, 2021 03.
Article in English | MEDLINE | ID: mdl-33532888

ABSTRACT

The immune system has very intricate mechanisms of fighting against the invading infections which are accomplished by a sequential event of molecular interactions in the body. One of the crucial phenomena in this process is the recognition of T-cells by the antigen-presenting cells (APCs), which is initiated by the rapid interaction between both cell surface receptors, i.e., CD2 located on T-cells and CD58 located on APCs. Under various pathological conditions, which involve undesired immune response, inhibiting the CD2-CD58 interactions becomes a therapeutically relevant opportunity. Herein we present an extensive work to identify novel inhibiting agents of the CD2-CD58 interactions. Classical molecular dynamics (MD) simulations of the CD2-CD58 complex highlighted a series of crucial CD58 residues responsible for the interactions with CD2. Based on such results, a pharmacophore map, complementary to the CD2-binding site of CD58, was created and employed for virtual screening of ~ 300,000 available compounds. On the ~ 6000 compounds filtered from pharmacophore mapping, ADME screening leads to ~ 350 molecules. Molecular docking was then performed on these molecules, and fifteen compounds emerged with significant binding energy (< - 50 kcal/mol) for CD58. Finally, short MD simulations were performed in triplicate on each complex (i) to provide a microscopic view of the ligand binding and (ii) to rule out possibly weak binders of CD58 from the identified hits. At last, we suggest eight compounds for in vitro testing that were identified as promising hits to bind CD58 with a high binding affinity.


Subject(s)
CD2 Antigens/chemistry , CD58 Antigens/chemistry , Organic Chemicals/chemistry , Amino Acid Sequence , Binding Sites , Databases, Chemical , Drug Evaluation, Preclinical , Humans , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Structure-Activity Relationship , T-Lymphocytes
13.
Nucleic Acids Res ; 47(1): 421-431, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30418605

ABSTRACT

RNA splicing is a highly regulated process dependent on sequences near splice sites. Insertions of Alu retrotransposons can disrupt splice sites or bind splicing regulators. We hypothesized that some common inherited polymorphic Alu insertions are responsible for splicing QTLs (sQTL). We focused on intronic Alu variants mapping within 100 bp of an alternatively used exon and screened for those that alter splicing. We identify five loci, 21.7% of those assayed, where the polymorphic Alu alters splicing. While in most cases the Alu promotes exon skipping, at one locus the Alu increases exon inclusion. Of particular interest is an Alu polymorphism in the CD58 gene. Reduced CD58 expression is associated with risk for developing multiple sclerosis. We show that the Alu insertion promotes skipping of CD58 exon 3 and results in a frameshifted transcript, indicating that the Alu may be the causative variant for increased MS risk at this locus. Using RT-PCR analysis at the endogenous locus, we confirm that the Alu variant is a sQTL for CD58. In summary, altered splicing efficiency is a common functional consequence of Alu polymorphisms including at least one instance where the variant is implicated in disease risk. This work broadens our understanding of splicing regulatory sequences around exons.


Subject(s)
Alu Elements/genetics , CD58 Antigens/genetics , Quantitative Trait Loci/genetics , RNA Splicing/genetics , Alternative Splicing/genetics , Exons/genetics , Genetic Variation , Humans , Introns/genetics , RNA Splice Sites/genetics , RNA, Messenger/genetics
14.
Eur J Immunol ; 49(9): 1441-1443, 2019 09.
Article in English | MEDLINE | ID: mdl-31332790

ABSTRACT

Activated allergen-specific Th2 and Th1 cells release cytokines that transform neutrophils into functional APCs characterized by the expression of HLA-DR and CD58 as well as enhanced survival and antigen uptake, irrespectively of the presence of IL-10, which reduces allergen uptake by neutrophils.


Subject(s)
Antigen Presentation/immunology , Cytokines/immunology , Lymphocyte Activation/immunology , Neutrophils/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Allergens/immunology , CD4-Positive T-Lymphocytes/immunology , CD58 Antigens/immunology , HLA-DR Antigens/immunology , Humans
15.
Immunol Invest ; 49(1-2): 106-119, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31505972

ABSTRACT

The prognosis of autoimmune thyroid diseases (AITDs), including Graves' disease (GD) and Hashimoto's disease (HD), varies among patients. The interaction of CD58 and its ligand (CD2) promotes the differentiation of regulatory T cells and suppresses the immune response. To clarify the association of CD58 expression with the pathogenesis and prognosis of AITDs, we genotyped polymorphisms in the CD58 gene including rs12044852A/C (SNP1), rs2300747A/G (SNP2), rs1335532C/T (SNP3), rs1016140G/T (SNP4), rs1414275C/T (SNP5) and rs11588376C/T (SNP6). The CD58 SNPs were genotyped in 177 GD patients, 193 HD patients and 116 healthy volunteers (control subjects). We used the Polymerase chain reaction-Restriction Fragment Length Polymorphism (PCR-RFLP) method for the genotyping of SNP1 and SNPs3-6 and the TaqMan® SNP genotyping assay for the genotyping of SNP2. The frequencies of the AA genotype in SNP1 tend to be high in all patients with AITDs than in control subjects, although it was not significant. The GG genotype of SNP2, the CC genotype of SNP3, the TT genotype of SNP4, the CC genotype of SNP5 and the CC genotype of SNP6 were all significantly more frequent in patients with AITDs than in control subjects. The proportion of CD58+ cells in monocytes was significantly lower in healthy individuals with each of these risk genotypes of AITDs and lower in GD and HD patients than that in healthy controls. In conclusion, CD58 SNPs are involved in AITD susceptibility through the reduction in CD58 expression, which probably suppresses regulatory T cells.


Subject(s)
CD58 Antigens/genetics , Graves Disease/genetics , Hashimoto Disease/genetics , Adult , Aged , Female , Genetic Predisposition to Disease/genetics , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Prognosis
16.
J Appl Microbiol ; 129(3): 753-767, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32145053

ABSTRACT

AIMS: To examine the modulation of the interacting partners of the calcineurin (CaN)-NFAT pathway in T cells during Cryptococcus neoformans fungal infection and post-T11TS immunotherapy. METHODS AND RESULTS: Wistar rats were infected with C. neoformans and followed by immunotherapy with immune-potentiator T11TS. T cells were analysed by flow cytometry, immunoblotting and nuclear translocation study. The signalling proteins LCK, FYN, LAT, PLCγ1 and CaN in T cells were regulated by C. neoformans infection resulting in reduced nuclear translocation of NFAT and IL-2 expression. Following T11TS immunotherapy, the expressions of the above-mentioned proteins were boosted and thus resulting in the clearance of C. neoformans from lung and spleen. CONCLUSIONS: The precise mechanism of suppression of the T-cell function by C. neoformans is still unknown. Previously, we have shown that T11TS positively regulates the function of T cells to abrogate glioma and other immunosuppressive conditions. T11TS immunotherapy increased the expression of the above signalling partners of the CaN-NFAT pathway in T cells and improved nuclear retention of NFAT. As a result, an increased IL-2 expression leads to activation and proliferation of T cells. SIGNIFICANCE AND IMPACT OF THE STUDY: Our results demonstrate the role of T11TS in restoring the CaN-NFAT signalling pathway in T cells. It identifies T11TS as an immunotherapeutic agent with potential clinical outcomes to counteract C. neoformans infection.


Subject(s)
CD58 Antigens/therapeutic use , Calcineurin/metabolism , Cryptococcosis/therapy , NFATC Transcription Factors/metabolism , T-Lymphocytes/immunology , Animals , Cryptococcosis/immunology , Cryptococcosis/microbiology , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/immunology , Immunotherapy/methods , Interleukin-2/metabolism , Lymphocyte Activation/drug effects , Rats , Rats, Wistar , Sheep , Signal Transduction/drug effects , T-Lymphocytes/metabolism
17.
J Gen Virol ; 100(1): 26-34, 2019 01.
Article in English | MEDLINE | ID: mdl-30480508

ABSTRACT

For an effective T-cell activation and response, co-stimulation is required in addition to the antigen-specific signal from their antigen receptors. The CD2/CD58 interaction is considered as one of the most important T-cell co-stimulatory pathways for T-cell activation and proliferation, and its role in regulating intestinal T-cell function in acute and chronic SIV -infected macaques is poorly documented. Here, we demonstrated a significant reduction of CD58 expression in both T- and B-cell populations during acute SIV infection along with high plasma viral load and a loss of intestinal CD4+ T cells compared to SIV-uninfected control macaques. The reduction of CD58 expression in T cells was correlated with the reduced expression of T-cell-mediated IL-2 and TNFα production. Together, these results indicate that reduction in the CD2/CD58 interaction pathway in mucosal lymphocytes might play a crucial role in mucosal T-cell dysfunction during acute SIV/HIV infection.


Subject(s)
CD58 Antigens/biosynthesis , Gene Expression , Interleukin-2/metabolism , Intestinal Mucosa/pathology , Intraepithelial Lymphocytes/immunology , Simian Acquired Immunodeficiency Syndrome/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , B-Lymphocytes/immunology , Lymphocyte Activation , Macaca , Plasma/virology , Simian Immunodeficiency Virus/isolation & purification , Viral Load
18.
Scand J Immunol ; 89(2): e12733, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30450625

ABSTRACT

Cryptococcus neoformans, the encapsulated yeast acquired through inhalation, remains localized in lungs, but harbours the CNS in immunocompromised individuals. Several treatment regimes have failed combating this disease totally, but long-term usage of drugs leads to organ damage. As T11-target structure (T11TS) has documented profound immune potentiation, we aimed to investigate the role of microglia, pivotal immune cells of brain in ameliorating cryptococcosis, with T11TS immunotherapy. Murine model with C neoformans infection was prepared by intraperitoneal injection and the brains of rats examined 7 days post-infections for histopathology by PAS and Alcian blue staining corroborated with organ fungal burden evidencing restorative T11TS action on Cryptococcal meningitis. Immunotherapy with three doses of T11TS, a CD2 ligand, in C neoformans infected rats, upregulates toll-like receptors 2, -4 and -9 of microglia, indicating increased phagocytosis of the fungus. Flowcytometric analysis revealed increased numbers of T11TS treated brain infiltrating CD4+ and CD8+ T-lymphocytes along with increased MHC I and MHC II on microglia, activating the infiltrating lymphocytes aiding the killing mechanism. Present study also indicated that T11TS increased production of Th1 inflammatory cytokines conducive to fungal elimination while the inhibitory Th2 cytokines were dampened. This preclinical study is first of its kind to show that T11TS effected profound immune stimulation of microglial activity of C neoformans infected rats eradicating residual fungal burden from the brain and can be a useful therapeutic strategy in fighting against this deadly disease.


Subject(s)
Brain/drug effects , CD58 Antigens/therapeutic use , Cryptococcus neoformans/physiology , Immunologic Factors/therapeutic use , Immunotherapy/methods , Meningitis, Cryptococcal/therapy , Microglia/immunology , Animals , Brain/immunology , Brain/microbiology , Cattle , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Humans , Immunity, Innate/drug effects , Inflammation Mediators/metabolism , Male , Meningitis, Cryptococcal/immunology , Microglia/pathology , Rats , Rats, Wistar , T-Lymphocytes/immunology , Toll-Like Receptors/metabolism
19.
J Cell Physiol ; 233(2): 759-770, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28608562

ABSTRACT

Malignant glioma is the most fatal of astrocytic lineage tumors despite therapeutic advances. Onset and progression of gliomas is accompanied by severe debilitation of T-cell defense and T-cell survival. One of the chief contributors to T-cell survival downstream of activation is the PI3K-AKT pathway. Our prior studies showed that the novel immunotherapeutic molecule T11-target structure (T11TS) blocks T-cell apoptosis in glioma. We also showed activation of immunological synapse components and calcineurin-NFAT pathway following T11TS immunotherapy of glioma-bearing rats. This lead to investigations whether such T-cell activation upon T11TS therapy translates into activation of downstream PI3K/AKT signals which may be related to observed blockade of T-cell apoptosis. For the purpose, we assessed by flowcytometry and immunoblotting, expressions of PI3K, PDK1, AKT, p-AKT, and PTEN in splenic T-cells of normal, experimentally-induced glioma-bearing rats and glioma-bearing rats receiving first, second and third doses of T11TS. We also determined comparative nuclear translocation of NF-κB across groups. We found significant increases in T-cell expressions of PDK1, PI3K, and p-AKT in T11TS-treated animal groups compared to sharp downregulations in glioma. AKT levels remained unchanged across groups. PTEN levels declined sharply after T11TS immunotherapy. T11TS also caused enhanced NF-κB translocation to the T-cell nucleus compared to glioma group. Results showed heightened activation of the PI3K-AKT pathway in glioma-bearing rats following T11TS immunotherapy. These results illustrate the novel role of T11TS immunotherapy in ameliorating the PI3K pathway in T-cells in glioma-bearing animals to enhance T-cell survival, according greater defense against glioma. The study thus has far-reaching clinical outcomes.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , CD58 Antigens/pharmacology , Glioma/drug therapy , Immunotherapy/methods , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , T-Lymphocytes/drug effects , Tumor Escape/drug effects , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Active Transport, Cell Nucleus , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/immunology , Brain Neoplasms/pathology , CD28 Antigens/immunology , CD28 Antigens/metabolism , Cell Survival , Ethylnitrosourea , Female , Glioma/enzymology , Glioma/immunology , Glioma/pathology , Male , NF-kappa B/metabolism , PTEN Phosphohydrolase/metabolism , Phosphorylation , Rats , Signal Transduction/drug effects , T-Lymphocytes/enzymology , T-Lymphocytes/immunology
20.
Crit Rev Immunol ; 37(2-6): 463-481, 2017.
Article in English | MEDLINE | ID: mdl-29773030

ABSTRACT

Two signals are required for induction of cell proliferation and cytokine production in resting T cells. Occupancy of the T cell receptor by antigen/MHC complexes delivers the first signal to the T cell, while the second signal is provided by interaction with costimulatory ligands on APC. CD2, LFA-1, and CD28 are the major costimulatory and adhesive molecules on T cells and bind to the LFA-3, ICAM-1 and B7 ligands, respectively, on APC. LFA-3 plays a central role for naive and memory T helper cells during the early phase of an immune response. The LFA-3/CD2 pathway initiates strong antigen-independent cell adhesion, substantial expansion of naive T helper cells, and induction of large amounts of IFN-γ in memory cells. The release of IFN-γ may upregulate expression of ICAM-1 and B7 on APC and allows multiple adhesion pathways to amplify the immune response. The LFA- 1/ICAM-l pathway stimulates adhesion and cell proliferation more efficiently in memory T helper cells than in naive cells. Further, the results suggest that naive T helper cells express functionally inactive LFA-1 molecules on the cell surface, which may have a physiological role in keeping these cells in a resting state. B7 costimulation superinduces IL-2 production in both naive and memory T helper cells and generates long-lasting cell proliferation. This permits transition from an autocrine to a paracrine immune response. Coexpression of B7/LFA-3 provides an optimal APC function and enables a vigorous T cell response to minute amounts of antigen. AP-1 and NF-κB transcription factors are involved in the induction of several cytokine gene promoters and play a central role in the regulation of IL-2 gene transcription. LFA-3 costimulation only moderately enhances AP-1 DNA-binding activity and does not influence the NF-κB activity induced by TCR engagement, whereas B7 costimulation induces large amounts of NF-κB and AP-1 activity in T helper cells. The costimulatory ligands represent a family of adhesion molecules with considerable redundancy. Interfamily redundancy of LFA-3, B7, and ICAM ligands offers an opportunity to regulate distinct T cell response profiles in various microenvironments at separate time points of an immune response.


Subject(s)
B7 Antigens/metabolism , CD58 Antigens/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , B7 Antigens/immunology , CD58 Antigens/immunology , Cell Adhesion/immunology , Cell Proliferation , Cytokines/immunology , Cytokines/metabolism , Humans , Immunity, Cellular , Immunologic Memory/immunology , Intercellular Adhesion Molecule-1/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL