Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
BMC Cancer ; 23(1): 1005, 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37858067

ABSTRACT

BACKGROUND: SH2 domain containing 1A (SH2D1A) expression has been linked to cancer progression. However, the functions of SH2D1A in hepatocellular carcinoma (HCC) have not been reported. METHODS: The effects of SH2D1A on the proliferation, migration, and invasion of HCC cells and the related pathways were re-explored in cell models with SH2D1A overexpression using the CCK-8, migration and invasion assays and western blotting. The functions and mechanisms of genes co-expressed with SH2D1A were analyzed using gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. The relationship between SH2D1A expression and immune microenvironment features in HCC was explored. RESULTS: Elevated SH2D1A expression promoted cell proliferation, migration, and invasion, which was related to the overexpression of p-Nf-κB and BCL2A1 protein levels in HCC. SH2D1A expression was related to the immune, stromal, and ESTIMATE scores, and the abundance of immune cells, such as B cells, CD8+ T cells, and T cells. SH2D1A expression was significantly related to the expression of immune cell markers, such as PDCD1, CD8A, and CTLA4 in HCC. CONCLUSION: SH2D1A overexpression was found to promote cell growth and metastasis via the Nf-κB signaling pathway and may be related to the immune microenvironment in HCC. The findings indicate that SH2D1A can function as a biomarker in HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Signaling Lymphocytic Activation Molecule Associated Protein , Humans , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , CD8-Positive T-Lymphocytes , Computational Biology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Neoplastic Processes , NF-kappa B , Signaling Lymphocytic Activation Molecule Associated Protein/genetics , Signaling Lymphocytic Activation Molecule Associated Protein/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
2.
Front Immunol ; 12: 747738, 2021.
Article in English | MEDLINE | ID: mdl-34987501

ABSTRACT

X-linked lymphoproliferative disease (XLP1) is a combined immunodeficiency characterized by severe immune dysregulation caused by mutations in the SH2D1A/SAP gene. Loss or dysfunction of SH2D1A is associated with the inability in clearing Epstein-Barr-Virus (EBV) infections. Clinical manifestation is diverse and ranges from life-threatening hemophagocytic lymphohistiocytosis (HLH) and fulminant infectious mononucleosis (FIM) to lymphoma and antibody deficiency. Rare manifestations include aplastic anemia, chronic gastritis and vasculitis. Herein, we describe the case of a previously healthy eight-year old boy diagnosed with XLP1 presenting with acute non-EBV acute meningoencephalitis with thrombotic occlusive vasculopathy. The patient developed multiple cerebral aneurysms leading to repeated intracerebral hemorrhage and severe cerebral damage. Immunological examination was initiated after development of a susceptibility to infections with recurrent bronchitis and one episode of severe pneumonia and showed antibody deficiency with pronounced IgG1-3-4 subclass deficiency. We could identify a novel hemizygous SH2D1A point mutation affecting the start codon. Basal levels of SAP protein seemed to be detectable in CD8+ and CD4+ T- and CD56+ NK-cells of the patient what indicated an incomplete absence of SAP. In conclusion, we could demonstrate a novel SH2D1A mutation leading to deficient SAP protein expression and a rare clinical phenotype of non-EBV associated acute meningoencephalitis with thrombotic occlusive vasculopathy.


Subject(s)
Epstein-Barr Virus Infections/immunology , Lymphoproliferative Disorders/immunology , Meningoencephalitis/immunology , Signaling Lymphocytic Activation Molecule Associated Protein/immunology , Thrombosis/immunology , Child , Epstein-Barr Virus Infections/diagnosis , Humans , Lymphoproliferative Disorders/diagnosis , Male , Meningoencephalitis/diagnosis , Mutation , Signaling Lymphocytic Activation Molecule Associated Protein/genetics , Thrombosis/diagnosis
3.
J Immunol ; 205(12): 3263-3276, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33199538

ABSTRACT

Signaling lymphocytic activation molecule-associated protein (SAP), a critical intracellular signaling molecule for T-B lymphocyte interactions, drives T follicular helper (Tfh) cell development in germinal centers (GCs). High-affinity islet autoantibodies predict type 1 diabetes (T1D) but do not cause ß cell destruction. This paradox intimates Tfh cells as key pathologic effectors, consistent with an observed Tfh signature in T1D. To understand how fully developed Tfh (GC Tfh) contribute to different autoimmune processes, we investigated the role of SAP in T1D and autoantibody-mediated arthritis. Whereas spontaneous arthritis depended on SAP in the autoantibody-mediated K/BxN model, organized insulitis and diabetes onset were unabated, despite a blocked anti-insulin vaccine response in SAP-deficient NOD mice. GC Tfh and GC B cell development were blocked by loss of SAP in K/BxN mice. In contrast, although GC B cell formation was markedly reduced in SAP-deficient NOD mice, T cells with a GC Tfh phenotype were found at disease sites. CXCR3+ CCR6- (Tfh1) subset bias was observed among GC Tfh cells infiltrating the pancreas of NOD mice, which was enhanced by loss of SAP NOD T cells override SAP requirement to undergo activation and proliferation in response to Ag presentation, demonstrating the potential for productive cognate T-B lymphocyte interactions in T1D-prone mice. We find that SAP is essential when autoantibody-driven immune complexes promote inflammation but is not required for effective organ-specific autoimmune attack. Thus, Tfh induced in classic GC reactions are dispensable for T1D, but the autoimmune process in the NOD model retains pathogenic Tfh without SAP.


Subject(s)
B-Lymphocytes/immunology , Cell Communication/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Signaling Lymphocytic Activation Molecule Associated Protein/immunology , Th1 Cells/immunology , Animals , Autoantibodies/genetics , Autoantibodies/immunology , B-Lymphocytes/pathology , Cell Communication/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Mice , Mice, Inbred NOD , Mice, Transgenic , Signaling Lymphocytic Activation Molecule Associated Protein/genetics , Th1 Cells/pathology
4.
Immunohorizons ; 4(4): 153-164, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32276922

ABSTRACT

SLAM-associated protein (SAP) is an adaptor molecule that facilitates critical effector functions in immune cells, and its deficiency causes X-linked lymphoproliferative disease type 1 in which effector responses directed against EBV are severely compromised. The primary objective of this study was to phenotypically and functionally characterize a rare, CD8 T cell-restricted bimodal SAP expression pattern observed in healthy, human donors with the widely used 1C9-SAP mAb clone. We initially observed this pattern during the clinical validation of our flow cytometry-based assay to diagnose X-linked lymphoproliferative disease type 1 in our laboratory. For this validation study, we used multiparameter flow cytometry to identify cytosolic SAP expression in lymphocyte subsets, and CD8 T cells from the donors displaying the rare SAP expression pattern mentioned above were separately further evaluated by intracellular cytokine and CD107a staining to examine polyfunctionality following PMA/ionomycin and HLA class I allele-restricted EBV peptide epitope-induced T cell activation. Our data revealed that SAP 1C9-hi CD8 T cells clearly displayed higher polyfunctional responses versus SAP 1C9-lo CD8 T cells following PMA/ionomycin stimulation. Furthermore, polyfunctional EBV-specific CD8 T cell responses segregated with the SAP 1C9-hi CD8 T cells and not the SAP 1C9-lo CD8 T cells. Additionally, and rather intriguingly, short- and long-term T cell stimulation selectively diminished the signal for the 1C9-hi subset. Overall, our data suggest that although rare, this unique SAP expression pattern merits further evaluation as it has the potential to provide some insight into fundamental processes as they might relate to host-pathogen dynamics.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , CD8-Positive T-Lymphocytes/immunology , Phenotype , Signaling Lymphocytic Activation Molecule Associated Protein/immunology , Signaling Lymphocytic Activation Molecule Associated Protein/metabolism , Adult , Blood Donors , Cells, Cultured , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/pharmacology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Female , Flow Cytometry , Herpesvirus 4, Human/immunology , Host-Pathogen Interactions/immunology , Humans , Immunoglobulin G/immunology , Ionomycin/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Signal Transduction/drug effects , Signal Transduction/immunology , Tetradecanoylphorbol Acetate/pharmacology
5.
J Immunotoxicol ; 16(1): 155-163, 2019 12.
Article in English | MEDLINE | ID: mdl-31403359

ABSTRACT

Exposure to the widely-used phthalate plasticizer di-(2-ethylhexyl)-phthalate (DEHP) has been shown to be closely related to an increased prevalence of allergic diseases in infants and juveniles. Earlier work in our laboratory found that DEHP-related anaphylactic responses could be ascribed to T-follicular helper (Tfh) cell hyperfunction directly. The Tfh cell, a newly identified CD4+ TH cell subset, until recently has been considered as a key player in humoral immunity. Tfh cells can respond to stimulation through various receptors. Signaling lymphocytic activation molecule family member-1 (SLAMF1, CD150) is a surface co-stimulatory receptor that can bind to an intracytoplasmic adaptor signaling lymphocytic activation molecule-associated protein (SAP) to initiate downstream signaling cascades, regulating some events of immune response. The present study explored the role of SLAMF1 in Tfh cell differentiation and cytokine secretion under the condition of DEHP exposure. Using a weanling mice model of DEHP gavage with ovalbumin (OVA) sensitization, it was found that DEHP acted as an immunoadjuvant to elevate SLAMF1 and SAP expression in host Tfh cells. Ex vivo studies of effects from DEHP exposure on Tfh cells from OVA-sensitized hosts showed that DEHP acted in an adjuvant-like manner to promote the expression of adaptor protein SAP, transcription factors Bcl-6 and c-MAF, and cytokines interleukin (IL)-21 and IL-4 in Tfh cells. Transfection of these Tfh cells with Slamf1 small interfering RNA prior to exposure to the DEHP attenuated the over-expression of these molecules that was caused by the DEHP. In conclusion, this study demonstrated that DEHP, via a SLAMF1-mediated pathway, can impact on Tfh cell differentiation and their ability to form select cytokines.


Subject(s)
Anaphylaxis/immunology , Cell Differentiation/drug effects , Diethylhexyl Phthalate/toxicity , Plasticizers/toxicity , T-Lymphocytes, Helper-Inducer/drug effects , Administration, Oral , Anaphylaxis/chemically induced , Animals , Animals, Newborn , Cell Differentiation/immunology , Child , Diethylhexyl Phthalate/administration & dosage , Disease Models, Animal , Humans , Interleukin-4/immunology , Interleukin-4/metabolism , Interleukins/immunology , Interleukins/metabolism , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Ovalbumin/immunology , Plasticizers/administration & dosage , Signal Transduction/drug effects , Signal Transduction/immunology , Signaling Lymphocytic Activation Molecule Associated Protein/immunology , Signaling Lymphocytic Activation Molecule Associated Protein/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1/immunology , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Weaning
7.
Proc Natl Acad Sci U S A ; 114(48): 12797-12802, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29133396

ABSTRACT

Follicular helper T cells (TFHs) are a key component of adaptive immune responses as they help antibody production by B cells. Differentiation and function of TFH cells are controlled by the master gene BCL6, but it is largely unclear how this transcription repressor specifies the TFH program. Here we asked whether BCL6 controlled helper function through down-regulation of specific microRNAs (miRNAs). We first assessed miRNA expression in TFH cells and defined a TFH-specific miRNA signature. We report that hsa-miR-31-5p (miR-31) is down-regulated in TFH; we showed that BCL6 suppresses miR-31 expression by binding to its promoter; and we demonstrated that miR-31 inhibits the expression of molecules that control T-helper function, such as CD40L and SAP. These findings identify a BCL6-initiated inhibitory circuit that stabilizes the follicular helper T cell program at least in part through the control of miRNA transcription. Although BCL6 controls TFH activity in human and mouse, the role of miR-31 is restricted to human TFH cell differentiation, reflecting a species specificity of the miR-31 action. Our findings highlight miR-31 as a possible target to modulate human T cell dependent antibody responses in the settings of infection, vaccination, or immune dysregulation.


Subject(s)
B-Lymphocytes/immunology , CD40 Ligand/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Signaling Lymphocytic Activation Molecule Associated Protein/genetics , T-Lymphocytes, Helper-Inducer/immunology , Adaptive Immunity , Animals , B-Lymphocytes/cytology , CD40 Ligand/immunology , Cell Differentiation , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Germinal Center/cytology , Germinal Center/immunology , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/immunology , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-bcl-6/immunology , Signal Transduction , Signaling Lymphocytic Activation Molecule Associated Protein/immunology , Species Specificity , T-Lymphocytes, Helper-Inducer/cytology
8.
Rheumatology (Oxford) ; 56(7): 1206-1216, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28387859

ABSTRACT

Objectives: The aim was to investigate whether the signalling lymphocyte activation molecule (SLAM) signalling pathways contribute to LN and whether SLAM receptors could be valuable biomarkers of disease activity. Methods: Peripheral blood mononuclear cells from 30National Research Ethics Service SLE patients with biopsy-proven LN were analysed by flow cytometry. Clinical measures of disease activity were assessed. The expression of the SLAM family receptors on T-cell subpopulations [CD4, CD8 and double negative (DN) T cells] was measured and compared between lupus patients with active renal disease and those in remission. Results: The frequency of CD8 T cells expressing SLAMF3, SLAMF5 and SLAMF7 was significantly lower in LN patients who were in remission. In contrast, these subsets were similar in patients with active renal disease and in healthy individuals. Patients with active nephritis had an increased percentage of circulating monocytes, consistent with a potential role played by these cells in glomerular inflammation. Changes in the frequency of DN T cells positive for SLAMF2, SLAMF4 and SLAMF7 were observed in lupus patients irrespective of the disease activity. We detected alterations in the cellular expression of the SLAM family receptors, but these changes were less obvious and did not reveal any specific pattern. The percentage of DN T cells expressing SLAMF6 could predict the clinical response to B-cell depletion in patients with LN. Conclusion: Our study demonstrates altered expression of the SLAM family receptors in SLE T lymphocytes. This is consistent with the importance of the SLAM-associated pathways in lupus pathogenesis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lupus Nephritis/drug therapy , Lupus Nephritis/immunology , Rituximab/therapeutic use , Signaling Lymphocytic Activation Molecule Associated Protein/metabolism , Adult , Antigens, CD/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Biomarkers/metabolism , Biopsy, Needle , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cohort Studies , Disease Progression , Female , Humans , Immunohistochemistry , Infusions, Intravenous , Leukocytes, Mononuclear/metabolism , Lupus Nephritis/pathology , Lymphocyte Activation/immunology , Male , Middle Aged , Predictive Value of Tests , Risk Assessment , Severity of Illness Index , Signaling Lymphocytic Activation Molecule Associated Protein/immunology , Statistics, Nonparametric , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...