Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Clin Exp Immunol ; 215(1): 37-46, 2024 01 09.
Article in English | MEDLINE | ID: mdl-37583293

ABSTRACT

Staphylococcus aureus (SA) and its exotoxins activate eosinophils (Eos) and mast cells (MCs) via CD48, a GPI-anchored receptor belonging to the signaling lymphocytes activation molecules (SLAM) family. 2B4 (CD244), an immuno-regulatory transmembrane receptor also belonging to the SLAM family, is the high-affinity ligand for CD48. 2B4 is expressed on several leukocytes including NK cells, T cells, basophils, monocytes, dendritic cells (DCs), and Eos. In the Eos and MCs crosstalk carried out by physical and soluble interactions (named the 'allergic effector unit', AEU), 2B4-CD48 binding plays a central role. As CD48 and 2B4 share some structural characteristics and SA colonization accompanies most of the allergic diseases, we hypothesized that SA exotoxins (e.g. Staphylococcus enterotoxin B, SEB) can also bind and activate 2B4 and thereby possibly further aggravate inflammation. To check our hypothesis, we used in vitro, in silico, and in vivo methods. By enzyme-linked immunosorbent assay (ELISA), flow cytometry (FC), fluorescence microscopy, and microscale thermophoresis, we have shown that SEB can bind specifically to 2B4. By Eos short- and long-term activation assays, we confirmed the functionality of the SEB-2B4 interaction. Using computational modeling, we identified possible SEB-binding sites on human and mouse 2B4. Finally, in vivo, in an SEB-induced peritonitis model, 2B4-KO mice showed a significant reduction of inflammatory features compared with WT mice. Altogether, the results of this study confirm that 2B4 is an important receptor in SEB-mediated inflammation, and therefore a role is suggested for 2B4 in SA associated inflammatory conditions.


Subject(s)
Hypersensitivity , Staphylococcus aureus , Animals , Humans , Mice , CD48 Antigen/metabolism , Exotoxins , Inflammation , Signaling Lymphocytic Activation Molecule Family , Staphylococcus aureus/metabolism
2.
Blood ; 140(18): 1951-1963, 2022 11 03.
Article in English | MEDLINE | ID: mdl-35921533

ABSTRACT

Adult T-cell leukemia/lymphoma (ATLL) is one of the aggressive peripheral T-cell neoplasms with a poor prognosis. Accumulating evidence demonstrates that escape from adaptive immunity is a hallmark of ATLL pathogenesis. However, the mechanisms by which ATLL cells evade natural killer (NK)-cell-mediated immunity have been poorly understood. Here we show that CD48 expression in ATLL cells determines the sensitivity for NK-cell-mediated cytotoxicity against ATLL cells. We performed unbiased genome-wide clustered regularly interspaced short palindromic repeat (CRISPR) screening using 2 ATLL-derived cell lines and discovered CD48 as one of the best-enriched genes whose knockout conferred resistance to YT1-NK cell line-mediated cytotoxicity. The ability of CD48-knockout ATLL cells to evade NK-cell effector function was confirmed using human primary NK cells with reduced interferon-γ (IFNγ) induction and degranulation. We found that primary ATLL cells had reduced CD48 expression along with disease progression. Furthermore, other subgroups among aggressive peripheral T-cell lymphomas (PTCLs) also expressed lower concentrations of CD48 than normal T cells, suggesting that CD48 is a key molecule in malignant T-cell evasion of NK-cell surveillance. Thus, this study demonstrates that CD48 expression is likely critical for malignant T-cell lymphoma cell regulation of NK-cell-mediated immunity and provides a rationale for future evaluation of CD48 as a molecular biomarker in NK-cell-associated immunotherapies.


Subject(s)
Leukemia-Lymphoma, Adult T-Cell , Lymphoma, T-Cell, Peripheral , Adult , Humans , CD48 Antigen/genetics , CD48 Antigen/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Leukemia-Lymphoma, Adult T-Cell/genetics , Lymphoma, T-Cell, Peripheral/genetics , Killer Cells, Natural
3.
FASEB J ; 37(1): e22702, 2023 01.
Article in English | MEDLINE | ID: mdl-36520044

ABSTRACT

Neurodegenerative diseases result from the interplay of abnormal gene expression and various pathological factors. Therefore, a disease-specific integrative genetic approach is required to understand the complexities and causes of target diseases. Recent studies have identified the correlation between genes encoding several transmembrane proteins, such as the cluster of differentiation (CD) and Alzheimer's disease (AD) pathogenesis. In this study, CD48 and CD40 gene expression in AD, a neurodegenerative disease, was analyzed to infer this link. Total RNA sequencing was performed using an Alzheimer's disease mouse model brain and blood, and gene expression was determined using a genome-wide association study (GWAS). We observed a marked elevation of CD48 and CD40 genes in Alzheimer's disease. Indeed, the upregulation of both CD48 and CD40 genes was significantly increased in the severe Alzheimer's disease group. With the elevation of CD48 and CD40 genes in Alzheimer's disease, associations of protein levels were also markedly increased in tissues. In addition, overexpression of CD48 and CD40 genes triggered tau aggregation, and co-expression of these genes accelerated aggregation. The nuclear factor kappa B (NF-ĸB) signaling pathway was enriched by CD48 and CD40 gene expression: it was also associated with tau pathology. Our data suggested that the CD48 and CD40 genes are novel AD-related genes, and this approach may be useful as a diagnostic or therapeutic target for the disease.


Subject(s)
Alzheimer Disease , CD40 Antigens , CD48 Antigen , Protein Aggregates , tau Proteins , Animals , Mice , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , CD40 Antigens/genetics , CD40 Antigens/metabolism , CD48 Antigen/genetics , CD48 Antigen/metabolism , Gene Expression , Genome-Wide Association Study , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Protein Aggregates/genetics , Protein Aggregates/physiology , tau Proteins/genetics , tau Proteins/metabolism
4.
Cytotherapy ; 25(10): 1080-1090, 2023 10.
Article in English | MEDLINE | ID: mdl-37516949

ABSTRACT

BACKGROUND AIMS: Decades after the identification of natural killer (NK) cells as potential effector cells against malignantly transformed cells, an increasing amount of research suggests that NK cells are a prospective choice of immunocytes for cancer immunotherapy in addition to T lymphocytes for cancer immunotherapy. Recent studies have led to a breakthrough in the combination of hematopoietic stem-cell transplantation with allogeneic NK cells infusion for the treatment of malignant tumors. However, the short lifespan of NK cells in patients is the major impediment, limiting their efficacy. Therefore, prolonging the survival of NK cells will promote the application of NK-cell immunotherapy. As we have known, NK cells use a "missing-self" mechanism to lyse target cells and exert their functions through a wide array of activating, co-stimulatory and inhibitory receptors. Our previous study has suggested that CD244 (2B4), one of the co-stimulatory receptors, can improve the function of chimeric antigen receptor NK cells. However, the underlying mechanism of how 2B4 engages in the function of NK cells requires further investigation. Overall, we established a feeder cell with the expression of CD48, the ligand of 2B4, to investigate the function of 2B4-CD48 axis in NK cells, and meanwhile, to explore whether the newly generated feeder cell can improve the function of ex vivo-expanded NK cells. METHODS: First, K562 cells overexpressing 4-1BBL and membrane-bound IL-21 (mbIL-21) were constructed (K562-41BBL-mbIL-21) and were sorted to generate the single clone. These widely used feeder cells (K562-41BBL-mbIL-21) were named as Basic Feeder hereinafter. Based on the Basic feeder, CD48 was overexpressed and named as CD48 Feeder. Then, the genetically modified feeder cells were used to expand primary NK cells from peripheral blood or umbilical cord blood. In vitro experiments were performed to compare proliferation ability, cytotoxicity, survival and activation/inhibition phenotypes of NK cells stimulated via different feeder cells. K562 cells were injected into nude mice subcutaneously with tail vein injection of NK cells from different feeder system for the detection of NK in vivo persistence and function. RESULTS: Compared with Basic Feeders, CD48 Feeders can promote the proliferation of primary NK cells from peripheral blood and umbilical cord blood and reduce NK cell apoptosis by activating the p-ERK/BCL2 pathway both in vitro and in vivo without affecting overall phenotypes. Furthermore, NK cells expanded via CD48 Feeders showed stronger anti-tumor capability and infiltration ability into the tumor microenvironment. CONCLUSIONS: In this preclinical study, the engagement of the 2B4-CD48 axis can inhibit the apoptosis of NK cells through the p-ERK/BCL2 signal pathway, leading to an improvement in therapeutic efficiency.


Subject(s)
Neoplasms , Receptors, Immunologic , Animals , Humans , Mice , Antigens, CD/metabolism , Apoptosis , CD48 Antigen/metabolism , Killer Cells, Natural , Lymphocyte Activation , Mice, Nude , Prospective Studies , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Signaling Lymphocytic Activation Molecule Family/metabolism , Tumor Microenvironment
5.
Ann Allergy Asthma Immunol ; 130(2): 245-253.e9, 2023 02.
Article in English | MEDLINE | ID: mdl-36280100

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can progress into a severe form of acute lung injury. The cosignaling receptor cluster of differentiation 48 (CD48) exists in membrane-bound (mCD48) and soluble (sCD48) forms and has been reported to be implicated in antiviral immunity and dysregulated in several inflammatory conditions. Therefore, CD48 dysregulation may be a putative feature in COVID-19-associated inflammation that deserves consideration. OBJECTIVE: To analyze CD48 expression in lung autopsies and peripheral blood leukocytes and sera of patients with COVID-19. The expression of the CD48 ligand 2B4 on the membrane of peripheral blood leukocytes was also assessed. METHODS: Twenty-eight lung tissue samples obtained from COVID-19 autopsies were assessed for CD48 expression using gene expression profiling immunohistochemistry (HTG autoimmune panel). Peripheral whole blood was collected from 111 patients with COVID-19, and the expression of mCD48 and of membrane-bound 2B4 was analyzed by flow cytometry. Serum levels of sCD48 were assessed by enzyme-linked immunosorbent assay. RESULTS: Lung tissue of patients with COVID-19 showed increased CD48 messenger RNA expression and infiltration of CD48+ lymphocytes. In the peripheral blood, mCD48 was considerably increased on all evaluated cell types. In addition, sCD48 levels were significantly higher in patients with COVID-19, independently of disease severity. CONCLUSION: Considering the changes of mCD48 and sCD48, a role for CD48 in COVID-19 can be assumed and needs to be further investigated.


Subject(s)
COVID-19 , Receptors, Immunologic , Humans , CD48 Antigen/metabolism , SARS-CoV-2 , Inflammation
6.
PLoS Pathog ; 15(4): e1007658, 2019 04.
Article in English | MEDLINE | ID: mdl-30947296

ABSTRACT

Throughout evolution, cytomegaloviruses (CMVs) have been capturing genes from their hosts, employing the derived proteins to evade host immune defenses. We have recently reported the presence of a number of CD48 homologs (vCD48s) encoded by different pathogenic viruses, including several CMVs. However, their properties and biological relevance remain as yet unexplored. CD48, a cosignaling molecule expressed on the surface of most hematopoietic cells, modulates the function of natural killer (NK) and other cytotoxic cells by binding to its natural ligand 2B4 (CD244). Here, we have characterized A43, the vCD48 exhibiting the highest amino acid sequence identity with host CD48. A43, which is encoded by owl monkey CMV, is a soluble molecule released from the cell after being proteolytically processed through its membrane proximal region. A43 is expressed with immediate-early kinetics, yielding a protein that is rapidly detected in the supernatant of infected cells. Remarkably, surface plasmon resonance assays revealed that this viral protein binds to host 2B4 with high affinity and slow dissociation rates. We demonstrate that soluble A43 is capable to abrogate host CD48:2B4 interactions. Moreover, A43 strongly binds to human 2B4 and prevents 2B4-mediated NK-cell adhesion to target cells, therefore reducing the formation of conjugates and the establishment of immunological synapses between human NK cells and CD48-expressing target cells. Furthermore, in the presence of this viral protein, 2B4-mediated cytotoxicity and IFN-γ production by NK cells are severely impaired. In summary, we propose that A43 may serve as a functional soluble CD48 decoy receptor by binding and masking 2B4, thereby impeding effective NK cell immune control during viral infections. Thus, our findings provide a novel example of the immune evasion strategies developed by viruses.


Subject(s)
CD48 Antigen/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Receptors, Immunologic/immunology , Signaling Lymphocytic Activation Molecule Family/immunology , CD48 Antigen/metabolism , Cells, Cultured , Cytomegalovirus Infections/virology , Humans , Killer Cells, Natural/metabolism , Killer Cells, Natural/virology , Lymphocyte Activation , Receptors, Immunologic/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism
7.
J Infect Dis ; 219(3): 470-479, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30202861

ABSTRACT

FimH-mediated bacterial invasion and polymorphonuclear neutrophil (PMN) transmigration across human brain microvascular endothelial cells (HBMECs) are required for the pathogenesis of Escherichia coli meningitis. However, the underlying mechanism remains unclear. This study demonstrated that the TnphoA mutant (22A33) and FimH-knockout mutant (ΔFimH) of E coli strain E44, which resulted in inactivation of FimH, were less invasive and less effective in promoting PMN transmigration than their wild-type strain. FimH protein induced PMN transmigration, whereas calmodulin inhibitor significantly blocked this effect. Moreover, immunofluorescence and co-immunoprecipitation analysis indicated that colocalized CD48 and α7 nAChR formed a complex on the surface of HBMECs that is associated with increased cofilin dephosphorylation, which could be remarkably enhanced by FimH+ E44. Our study concluded that FimH-induced E coli K1 invasion and PMN migration across HBMECs may be mediated by the CD48-α7nAChR complex in lipid rafts of HBMEC via Ca2+ signaling and cofilin dephosphorylation.


Subject(s)
Adhesins, Escherichia coli/metabolism , Brain/microbiology , CD48 Antigen/metabolism , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Escherichia coli/metabolism , Fimbriae Proteins/metabolism , Neutrophils/microbiology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Adhesins, Escherichia coli/genetics , Calcium/metabolism , Cell Movement , Escherichia coli/pathogenicity , Fimbriae Proteins/genetics , Gene Knockdown Techniques , Humans , Membrane Microdomains , Meningitis, Escherichia coli/microbiology , Meningitis, Escherichia coli/pathology , Signal Transduction , Virulence Factors/genetics , Virulence Factors/metabolism
8.
Eur J Immunol ; 47(6): 1051-1061, 2017 06.
Article in English | MEDLINE | ID: mdl-28386908

ABSTRACT

X-linked lymphoproliferative disease 1 (XLP1) is an inherited immunodeficiency, caused by mutations in SH2D1A encoding Signaling Lymphocyte Activation Molecule (SLAM)-associated protein (SAP). In XLP1, 2B4, upon engagement with CD48, has inhibitory instead of activating function. This causes a selective inability of cytotoxic effectors to kill EBV-infected cells, with dramatic clinical sequelae. Here, we investigated the NK cell education in XLP1, upon characterization of killer Ig-like receptor (KIR)/KIR-L genotype and phenotypic repertoire of self-HLA class I specific inhibitory NK receptors (self-iNKRs). We also analyzed NK-cell cytotoxicity against CD48+ or CD48- KIR-ligand matched or autologous hematopoietic cells in XLP1 patients and healthy controls. XLP1 NK cells may show a defective phenotypic repertoire with substantial proportion of cells lacking self-iNKR. These NK cells are cytotoxic and the inhibitory 2B4/CD48 pathway plays a major role to prevent killing of CD48+ EBV-transformed B cells and M1 macrophages. Importantly, self-iNKR defective NK cells kill CD48- targets, such as mature DCs. Self-iNKR- NK cells in XLP1 patients are functional even in resting conditions, suggesting a role of the inhibitory 2B4/CD48 pathway in the education process during NK-cell maturation. Killing of autologous mature DC by self-iNKR defective XLP1 NK cells may impair adaptive responses, further exacerbating the patients' immune defect.


Subject(s)
Killer Cells, Natural/immunology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/physiopathology , Receptors, Natural Killer Cell/immunology , Signaling Lymphocytic Activation Molecule Family/metabolism , CD48 Antigen/immunology , CD48 Antigen/metabolism , Genes, MHC Class I , Humans , Killer Cells, Natural/metabolism , Lymphocyte Activation , Potassium Channels, Inwardly Rectifying/immunology , Receptors, Immunologic/metabolism , Signal Transduction , Signaling Lymphocytic Activation Molecule Associated Protein/metabolism , Signaling Lymphocytic Activation Molecule Family/immunology
9.
Nanomedicine ; 14(1): 123-130, 2018 01.
Article in English | MEDLINE | ID: mdl-28939491

ABSTRACT

Liposomes functionalized with monoclonal antibodies or their antigen-binding fragments have attracted much attention as specific drug delivery devices for treatment of various diseases including cancer. The conjugation of antibodies to liposomes is usually achieved by covalent coupling using cross-linkers in a reaction that might adversely affect the characteristics of the final product. Here we present an alternative strategy for liposome functionalization: we created a recombinant Fab antibody fragment genetically fused on its C-terminus to the hydrophobic peptide derived from pulmonary surfactant protein D, which became inserted into the liposomal bilayer during liposomal preparation and anchored the Fab onto the liposome surface. The Fab-conjugated liposomes specifically recognized antigen-positive cells and efficiently delivered their cargo, the Alexa Fluor 647 dye, into target cells in vitro and in vivo. In conclusion, our approach offers the potential for straightforward development of nanomedicines functionalized with an antibody of choice without the need of harmful cross-linkers.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin Fab Fragments/immunology , Liposomes/chemistry , Lymphoma/immunology , Peptide Fragments/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , CD48 Antigen/metabolism , CD59 Antigens/metabolism , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Jurkat Cells , Lymphoma/metabolism , Lymphoma/pathology , Mice , Peptide Fragments/metabolism , Pulmonary Surfactant-Associated Protein D/immunology , Pulmonary Surfactant-Associated Protein D/metabolism , Tumor Cells, Cultured
10.
Mediators Inflamm ; 2017: 7521701, 2017.
Article in English | MEDLINE | ID: mdl-28337051

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous subset of cells that expands dramatically in many disease states and can suppress T-cell responses. MDSCs mainly include monocytic and granulocytic subpopulations that can be distinguished in mice by the expression of Ly6G and Ly6C cell surface markers. This identification system has been validated in experimental tumor models, but not in models of inflammation-associated conditions such as sepsis. We challenged growth factor independent 1 transcription repressor green fluorescent protein (Gfi1:GFP) knock-in reporter mice with cecal ligation and puncture surgery and found that CD11b+Ly6GlowLy6Chigh MDSCs in this sepsis model comprised both monocytic and granulocytic MDSCs. The evidence that conventional Ly6G/Ly6C marker analysis may not be suited to study of inflammation-induced MDSCs led to the development of a novel strategy of distinguishing granulocytic MDSCs from monocytic MDSCs in septic mice by expression of CD48. Application of this novel model should help achieve a more accurate understanding of the inflammation-induced MDSC activity.


Subject(s)
CD48 Antigen/metabolism , Myeloid-Derived Suppressor Cells/cytology , Sepsis/metabolism , Animals , CD48 Antigen/genetics , Cell Membrane/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Genes, Reporter , Granulocytes/cytology , Granulocytes/metabolism , Green Fluorescent Proteins/metabolism , Inflammation , Male , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Myeloid Cells/metabolism , Sepsis/physiopathology , Transcription Factors/metabolism
11.
Immunol Cell Biol ; 94(6): 583-92, 2016 07.
Article in English | MEDLINE | ID: mdl-26860368

ABSTRACT

Polyfunctionality and cytotoxic activity dictate CD8(+) T-cell efficacy in the eradication of infected and malignant cells. The induction of these effector functions depends on the specific interaction between the T-cell receptor (TCR) and its cognate peptide-MHC class I complex, in addition to signals provided by co-stimulatory or co-inhibitory receptors, which can further regulate these functions. Among these receptors, the role of 2B4 is contested, as it has been described as either co-stimulatory or co-inhibitory in modulating T-cell functions. We therefore combined functional, transcriptional and epigenetic approaches to further characterize the impact of disrupting the interaction of 2B4 with its ligand CD48, on the activity of human effector CD8(+) T-cell clones. In this setting, we show that the 2B4-CD48 axis is involved in the fine-tuning of CD8(+) T-cell effector function upon antigenic stimulation. Blocking this interaction resulted in reduced CD8(+) T-cell clone-mediated cytolytic activity, together with a subtle drop in the expression of genes involved in effector function regulation. Our results also imply a variable contribution of the 2B4-CD48 interaction to the modulation of CD8(+) T-cell functional properties, potentially linked to intrinsic levels of T-bet expression and TCR avidity. The present study thus provides further insights into the role of the 2B4-CD48 interaction in the fine regulation of CD8(+) T-cell effector function upon antigenic stimulation.


Subject(s)
CD48 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Signaling Lymphocytic Activation Molecule Family/metabolism , Antibody Affinity/immunology , Cytotoxicity, Immunologic/genetics , Epigenesis, Genetic , Humans , Immunomodulation , Protein Binding , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Box Domain Proteins/metabolism , Transcription, Genetic
12.
Allergy ; 71(6): 829-39, 2016 06.
Article in English | MEDLINE | ID: mdl-26836239

ABSTRACT

BACKGROUND: Staphylococcus aureus, one of the most important pathogens, is heavily associated with allergy. S. aureus and its toxins interact with eosinophils through CD48, a GPI-anchored receptor important in allergy mainly as expressed by the eosinophils (mCD48). CD48 can exist in a soluble form (sCD48). Our aim was to investigate SEB-induced regulation of eosinophil CD48 and the possible formation and role of sCD48 in SEB-mediated eosinophil activation in vitro and in vivo. METHODS: Human peripheral blood eosinophils were activated by SEB with or without inhibitors for phospholipases (PL) (-C or -D), or cycloheximide, or brefeldin A. We evaluated eosinophil activation (CD11b expression or EPO/IL-8 release), mCD48 (flow cytometry), sCD48 (ELISA), SEB binding to sCD48 (ELISA), and chemotaxis toward SEB. C57BL/6 mice were pre-injected (ip.) with sCD48, and then, peritonitis was induced by SEB injection; peritoneal lavages were collected after 48 h and analyzed by flow cytometry and ELISA. RESULTS: SEB-activated human eosinophils formed sCD48, directly correlating with CD11b expression, through cell-associated PL-C and -D. mCD48 remained stable due to up-regulation in CD48 transcription and cellular trafficking. sCD48 bound to SEB and down-regulated SEB stimulatory effects on eosinophils as assessed by EPO and IL-8 release and eosinophil chemotaxis toward SEB. sCD48 showed anti-inflammatory activity in a SEB-induced mouse peritonitis model. CONCLUSIONS: SEB regulates CD48 dynamics on eosinophils. Our data indicate sCD48 as a SEB-induced 'decoy' receptor derived from eosinophil and therefore as a potential anti-inflammatory tool in S. aureus-induced eosinophil inflammation often associated with allergy.


Subject(s)
CD48 Antigen/blood , Enterotoxins/metabolism , Eosinophils/immunology , Eosinophils/metabolism , Inflammation/etiology , Inflammation/metabolism , Staphylococcus aureus/physiology , Animals , CD48 Antigen/genetics , CD48 Antigen/metabolism , Chemotaxis/genetics , Chemotaxis/immunology , Disease Models, Animal , Female , Humans , Hypersensitivity/blood , Hypersensitivity/etiology , Hypersensitivity/metabolism , Inflammation/blood , Leukocyte Count , Mice , Peritonitis/immunology , Peritonitis/metabolism , Peritonitis/microbiology , Phospholipases , Protein Binding , Proteolysis , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Transcription, Genetic
13.
Cell Rep ; 43(3): 113800, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38386559

ABSTRACT

Infection of mice by mouse cytomegalovirus (MCMV) triggers activation and expansion of Ly49H+ natural killer (NK) cells, which are virus specific and considered to be "adaptive" or "memory" NK cells. Here, we find that signaling lymphocytic activation molecule family receptors (SFRs), a group of hematopoietic cell-restricted receptors, are essential for the expansion of Ly49H+ NK cells after MCMV infection. This activity is largely mediated by CD48, an SFR broadly expressed on NK cells and displaying augmented expression after MCMV infection. It is also dependent on the CD48 counter-receptor, 2B4, expressed on host macrophages. The 2B4-CD48 axis promotes expansion of Ly49H+ NK cells by repressing their phagocytosis by virus-activated macrophages through inhibition of the pro-phagocytic integrin lymphocyte function-associated antigen-1 (LFA-1) on macrophages. These data identify key roles of macrophages and the 2B4-CD48 pathway in controlling the expansion of adaptive NK cells following MCMV infection. Stimulation of the 2B4-CD48 axis may be helpful in enhancing adaptive NK cell responses for therapeutic purposes.


Subject(s)
Cytomegalovirus Infections , Receptors, Immunologic , Animals , Mice , Receptors, Immunologic/metabolism , CD48 Antigen/metabolism , Antigens, CD/metabolism , Lymphocyte Activation , Killer Cells, Natural , Receptors, Cell Surface/metabolism , Carrier Proteins/metabolism , Macrophages/metabolism , Phagocytosis
14.
J Immunol Res ; 2024: 6908968, 2024.
Article in English | MEDLINE | ID: mdl-38957433

ABSTRACT

Background: Kidney transplantation (KT) is the best treatment for end-stage renal disease. Although long and short-term survival rates for the graft have improved significantly with the development of immunosuppressants, acute rejection (AR) remains a major risk factor attacking the graft and patients. The innate immune response plays an important role in rejection. Therefore, our objective is to determine the biomarkers of congenital immunity associated with AR after KT and provide support for future research. Materials and Methods: A differential expression genes (DEGs) analysis was performed based on the dataset GSE174020 from the NCBI gene Expression Synthesis Database (GEO) and then combined with the GSE5099 M1 macrophage-related gene identified in the Molecular Signatures Database. We then identified genes in DEGs associated with M1 macrophages defined as DEM1Gs and performed gene ontology (GO) and Kyoto Encyclopedia of Genomes (KEGG) enrichment analysis. Cibersort was used to analyze the immune cell infiltration during AR. At the same time, we used the protein-protein interaction (PPI) network and Cytoscape software to determine the key genes. Dataset, GSE14328 derived from pediatric patients, GSE138043 and GSE9493 derived from adult patients, were used to verify Hub genes. Additional verification was the rat KT model, which was used to perform HE staining, immunohistochemical staining, and Western Blot. Hub genes were searched in the HPA database to confirm their expression. Finally, we construct the interaction network of transcription factor (TF)-Hub genes and miRNA-Hub genes. Results: Compared to the normal group, 366 genes were upregulated, and 423 genes were downregulated in the AR group. Then, 106 genes related to M1 macrophages were found among these genes. GO and KEGG enrichment analysis showed that these genes are mainly involved in cytokine binding, antigen binding, NK cell-mediated cytotoxicity, activation of immune receptors and immune response, and activation of the inflammatory NF-κB signaling pathway. Two Hub genes, namely CCR7 and CD48, were identified by PPI and Cytoscape analysis. They have been verified in external validation sets, originated from both pediatric patients and adult patients, and animal experiments. In the HPA database, CCR7 and CD48 are mainly expressed in T cells, B cells, macrophages, and tissues where these immune cells are distributed. In addition to immunoinfiltration, CD4+T, CD8+T, NK cells, NKT cells, and monocytes increased significantly in the AR group, which was highly consistent with the results of Hub gene screening. Finally, we predicted that 19 TFs and 32 miRNAs might interact with the Hub gene. Conclusions: Through a comprehensive bioinformatic analysis, our findings may provide predictive and therapeutic targets for AR after KT.


Subject(s)
CD48 Antigen , Graft Rejection , Kidney Transplantation , Macrophages , Protein Interaction Maps , Receptors, CCR7 , Humans , Graft Rejection/immunology , Graft Rejection/genetics , Kidney Transplantation/adverse effects , Macrophages/immunology , Macrophages/metabolism , Animals , Child , Rats , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , CD48 Antigen/genetics , CD48 Antigen/metabolism , Gene Expression Profiling , Biomarkers , Computational Biology/methods , Male , Gene Regulatory Networks , Databases, Genetic , Gene Ontology , Disease Models, Animal , Female , MicroRNAs/genetics
15.
Arch Pharm Res ; 45(1): 1-10, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34905179

ABSTRACT

The susceptibility of cancer cells to natural killer (NK) cell-mediated cytotoxicity depends on the balance of activating and inhibitory ligands expressed on their surface. Although many types of cancer cells are killed by NK cells, non-small-cell lung cancer (NSCLC) cells are relatively resistant to NK cell-mediated cytotoxicity. In this study, we showed that several NSCLC cell lines have differential sensitivity to NK cell-mediated cytotoxicity: NCI-H522 cells were highly sensitive, but A549, NCI-H23, NCI-H1915, and NCI-H1299 were resistant. Among activating ligands such as CD48, HLA-A/B/G, ICAM-1, MICA/B, and ULBPs, only CD48 rendered NCI-H522 cells susceptible to NK cell-mediated cytotoxicity, which was proved by using CD48 siRNA and neutralizing antibody. CD48-positive NCI-H522 cells established a more stable contact with NK cells than did CD48-negative A549 and CD48 siRNA cell-transfected NCI-H522 cells. Taken together, these data demonstrate that CD48-positive NSCLC cells might be susceptible to NK cell-mediated cytotoxicity, which provide information on how to stratify NSCLC patients potentially responsive to NK-cell therapy.


Subject(s)
CD48 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Killer Cells, Natural/physiology , Lung Neoplasms/immunology , Blotting, Western , CD48 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Flow Cytometry , Humans , Killer Cells, Natural/immunology , Lung Neoplasms/metabolism , Polymerase Chain Reaction
16.
Sci Immunol ; 7(74): eabn6373, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35930657

ABSTRACT

CD2 is largely described to promote T cell activation when engaged by its ligands, CD48 in mice and CD58 in humans, that are present on antigen-presenting cells (APCs). However, both CD48 and CD58 are also expressed on T cells. By generating new knockout mouse strains lacking CD2 or CD48 in the C57BL/6 background, we determined that whereas CD2 was necessary on T cells for T cell activation, its ligand CD48 was not required on APCs. Rather, CD48 was also needed on T cells. One exception was during cytotoxicity, which required CD48 on T cells and APCs. Fluorescence resonance energy transfer (FRET) studies in nonimmune cells provided evidence that cis interactions between CD2 and CD48 existed within individual cells. CD2-CD48 interactions on T cells enabled more robust T cell receptor (TCR) signals, including protein tyrosine phosphorylation. Using T cells from a CD2 knock-in mouse in which a tag was inserted at the carboxyl terminus of CD2, mass spectrometry analyses revealed that the role of CD2 in T cell activation correlated with its ability to interact with components of the TCR complex and the protein tyrosine kinase Lck. CD2-CD58 provided a similar function in human T cells. Thus, our data imply that T cell-intrinsic cis interactions of CD2 with its ligands are required for TCR signaling and T cell activation. Interactions with ligands on APCs contribute during cytotoxicity.


Subject(s)
Antigens, CD , Lymphocyte Activation , T-Lymphocytes , Animals , CD2 Antigens/chemistry , CD2 Antigens/metabolism , CD48 Antigen/metabolism , CD58 Antigens/metabolism , Humans , Ligands , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism
17.
Cell Mol Immunol ; 18(1): 194-205, 2021 01.
Article in English | MEDLINE | ID: mdl-31659245

ABSTRACT

The adaptive immune response relies on specific apoptotic programs to maintain homeostasis. Conventional effector T cell (Tcon) expansion is constrained by both forkhead box P3 (FOXP3)+-regulatory T cells (Tregs) and restimulation-induced cell death (RICD), a propriocidal apoptosis pathway triggered by repeated stimulation through the T-cell receptor (TCR). Constitutive FOXP3 expression protects Tregs from RICD by suppressing SLAM-associated protein (SAP), a key adaptor protein that amplifies TCR signaling strength. The role of transient FOXP3 induction in activated human CD4 and CD8 Tcons remains unresolved, but its expression is inversely correlated with acquired RICD sensitivity. Here, we describe a novel role for FOXP3 in protecting human Tcons from premature RICD during expansion. Unlike FOXP3-mediated protection from RICD in Tregs, FOXP3 protects Tcons through a distinct mechanism requiring de novo transcription that does not require SAP suppression. Transcriptome profiling and functional analyses of expanding Tcons revealed that FOXP3 enhances expression of the SLAM family receptor CD48, which in turn sustains basal autophagy and suppresses pro-apoptotic p53 signaling. Both CD48 and FOXP3 expression reduced p53 accumulation upon TCR restimulation. Furthermore, silencing FOXP3 expression or blocking CD48 decreased the mitochondrial membrane potential in expanding Tcons with a concomitant reduction in basal autophagy. Our findings suggest that FOXP3 governs a distinct transcriptional program in early-stage effector Tcons that maintains RICD resistance via CD48-dependent protective autophagy and p53 suppression.


Subject(s)
CD48 Antigen/metabolism , Cell Death , Forkhead Transcription Factors/metabolism , Lymphocyte Activation , Receptors, Antigen, T-Cell/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein/metabolism , T-Lymphocytes, Regulatory/immunology , Apoptosis , Autophagy , CD48 Antigen/genetics , Forkhead Transcription Factors/genetics , Humans , Receptors, Antigen, T-Cell/genetics , Signal Transduction , Signaling Lymphocytic Activation Molecule Associated Protein/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
18.
Stem Cell Reports ; 16(8): 1884-1893, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34297939

ABSTRACT

Immune cells are generated from hematopoietic stem cells (HSCs) in the bone marrow (BM). Immune stimulation can rapidly activate HSCs out of their quiescent state to accelerate the generation of immune cells. HSCs' activation follows various viral or bacterial stimuli, and we sought to investigate the hypersensitivity immune response. Surprisingly, the Ova-induced hypersensitivity peritonitis model finds no significant changes in BM HSCs. HSC markers cKIT, SCA1, CD48, CD150, and the Fgd5-mCherry reporter showed no significant difference from control. Functionally, hypersensitivity did not alter HSCs' potency, as assayed by transplantation. We further characterized the possible impact of hypersensitivity using RNA-sequencing of HSCs, finding minor changes at the transcriptome level. Moreover, hypersensitivity induced no significant change in the proliferative state of HSCs. Therefore, this study suggests that, in contrast to other immune stimuli, hypersensitivity has no impact on HSCs.


Subject(s)
Adaptive Immunity/immunology , Bone Marrow Cells/immunology , Hematopoietic Stem Cells/immunology , Hypersensitivity/immunology , Transcriptome/immunology , Animals , Ataxin-1/genetics , Ataxin-1/immunology , Ataxin-1/metabolism , Bone Marrow Cells/metabolism , CD48 Antigen/genetics , CD48 Antigen/immunology , CD48 Antigen/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/metabolism , Mice, Congenic , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , Proto-Oncogene Proteins c-kit/metabolism , RNA-Seq/methods , Transcriptome/genetics
19.
Sci Immunol ; 5(53)2020 11 20.
Article in English | MEDLINE | ID: mdl-33219153

ABSTRACT

Innate lymphoid cells (ILCs) develop from common lymphoid progenitors (CLPs), which further differentiate into the common ILC progenitor (CILP) that can give rise to both ILCs and natural killer (NK) cells. Murine ILC intermediates have recently been characterized, but the human counterparts and their developmental trajectories have not yet been identified, largely due to the lack of homologous surface receptors in both organisms. Here, we show that human CILPs (CD34+CD117+α4ß7+Lin-) acquire CD48 and CD52, which define NK progenitors (NKPs) and ILC precursors (ILCPs). Two distinct NK cell subsets were generated in vitro from CD34+CD117+α4ß7+Lin-CD48-CD52+ and CD34+CD117+α4ß7+Lin-CD48+CD52+ NKPs, respectively. Independent of NKPs, ILCPs exist in the CD34+CD117+α4ß7+Lin-CD48+CD52+ subset and give rise to ILC1s, ILC2s, and NCR+ ILC3s, whereas CD34+CD117+α4ß7+Lin-CD48+CD52- ILCPs give rise to a distinct subset of ILC3s that have lymphoid tissue inducer (LTi)-like properties. In addition, CD48-expressing CD34+CD117+α4ß7+Lin- precursors give rise to tissue-associated ILCs in vivo. We also observed that the interaction of 2B4 with CD48 induced differentiation of ILC2s, and together, these findings show that expression of CD48 by human ILCPs modulates ILC differentiation.


Subject(s)
CD48 Antigen/metabolism , Cell Differentiation/immunology , Killer Cells, Natural/physiology , Lymphoid Progenitor Cells/physiology , Signaling Lymphocytic Activation Molecule Family/metabolism , Animals , CD52 Antigen/metabolism , Cell Separation , Cells, Cultured , Flow Cytometry , Gene Knockout Techniques , Humans , Immunity, Innate , Mice , Primary Cell Culture , RNA-Seq , Signal Transduction/genetics , Signal Transduction/immunology , Signaling Lymphocytic Activation Molecule Family/genetics , Single-Cell Analysis , Species Specificity
20.
Viruses ; 12(8)2020 07 28.
Article in English | MEDLINE | ID: mdl-32731344

ABSTRACT

The genesis of gene families by the capture of host genes and their subsequent duplication is a crucial process in the evolution of large DNA viruses. CD48 is a cell surface molecule that interacts via its N-terminal immunoglobulin (Ig) domain with the cell surface receptor 2B4 (CD244), regulating leukocyte cytotoxicity. We previously reported the presence of five CD48 homologs (vCD48s) in two related cytomegaloviruses, and demonstrated that one of them, A43, binds 2B4 and acts as a soluble CD48 decoy receptor impairing NK cell function. Here, we have characterized the rest of these vCD48s. We show that they are highly glycosylated proteins that display remarkably distinct features: divergent biochemical properties, cellular locations, and temporal expression kinetics. In contrast to A43, none of them interacts with 2B4. Consistent with this, molecular modeling of the N-terminal Ig domains of these vCD48s evidences notable changes as compared to CD48, suggesting that they interact with alternative targets. Accordingly, we demonstrate that one of them, S30, tightly binds CD2, a crucial T- and NK-cell adhesion and costimulatory molecule. Thus, our findings show how a key host immune receptor gene captured by a virus can be subsequently remodeled to evolve new immunoevasins with altered binding properties.


Subject(s)
CD48 Antigen/genetics , CD48 Antigen/metabolism , Cytomegalovirus/genetics , Receptors, Cell Surface/metabolism , Animals , Cercopithecidae/virology , Cytomegalovirus/immunology , HEK293 Cells , Humans , Immune Evasion , Ligands , Models, Molecular , Protein Binding , Receptors, Immunologic/metabolism , Saimiri/virology , Sequence Homology , T-Lymphocytes/immunology , T-Lymphocytes/virology
SELECTION OF CITATIONS
SEARCH DETAIL