Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Biochemistry (Mosc) ; 89(5): 853-861, 2024 May.
Article in English | MEDLINE | ID: mdl-38880646

ABSTRACT

Tumor necrosis factor (TNF) is one of many cytokines - protein molecules responsible for communication between the cells of immune system. TNF was discovered and given its grand name because of its striking antitumor effects in experimental systems, but its main physiological functions in the context of whole organism turned out to be completely unrelated to protection against tumors. This short review discusses "man-made" mouse models generated by early genome-editing technologies, which enabled us to establish true functions of TNF in health and certain diseases as well as to unravel potential strategies for improving therapy of TNF-dependent diseases.


Subject(s)
Tumor Necrosis Factor-alpha , Animals , Humans , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Mice , Gene Editing/methods , Neoplasms/immunology , Neoplasms/genetics , Neoplasms/therapy
2.
Int J Mol Sci ; 25(1)2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38203754

ABSTRACT

Wound healing is a complex process involving a coordinated series of events aimed at restoring tissue integrity and function. Regulatory B cells (Bregs) are a subset of B lymphocytes that play an essential role in fine-tuning immune responses and maintaining immune homeostasis. Recent studies have suggested that Bregs are important players in cutaneous immunity. This review summarizes the current understanding of the role of Bregs in skin immunity in health and pathology, such as diabetes, psoriasis, systemic sclerosis, cutaneous lupus erythematosus, cutaneous hypersensitivity, pemphigus, and dermatomyositis. We discuss the mechanisms by which Bregs maintain tissue homeostasis in the wound microenvironment through the promotion of angiogenesis, suppression of effector cells, and induction of regulatory immune cells. We also mention the potential clinical applications of Bregs in promoting wound healing, such as the use of adoptive Breg transfer.


Subject(s)
B-Lymphocytes, Regulatory , Dermatitis, Atopic , Psoriasis , Humans , Skin , Wound Healing
3.
Front Immunol ; 14: 1178445, 2023.
Article in English | MEDLINE | ID: mdl-37731503

ABSTRACT

Regulatory B lymphocytes (Bregs) are B cells with well-pronounced immunosuppressive properties, allowing them to suppress the activity of effector cells. A broad repertoire of immunosuppressive mechanisms makes Bregs an attractive tool for adoptive cell therapy for diseases associated with excessive activation of immune reactions. Such therapy implies Breg extraction from the patient's peripheral blood, ex vivo activation and expansion, and further infusion into the patient. At the same time, the utility of Bregs for therapeutic approaches is limited by their small numbers and extremely low survival rate, which is typical for all primary B cell cultures. Therefore, extracting CD19+ cells from the patient's peripheral blood and specifically activating them ex vivo to make B cells acquire a suppressive phenotype seems to be far more productive. It will allow a much larger number of B cells to be obtained initially, which may significantly increase the likelihood of successful immunosuppression after adoptive Breg transfer. This comparative study focuses on finding ways to efficiently manipulate B cells in vitro to differentiate them into Bregs. We used CD40L, CpG, IL4, IL21, PMA, and ionomycin in various combinations to generate immunosuppressive phenotype in B cells and performed functional assays to test their regulatory capacity. This work shows that treatment of primary B cells using CD40L + CpG + IL21 mix was most effective in terms of induction of functionally active regulatory B lymphocytes with high immunosuppressive capacity ex vivo.


Subject(s)
B-Lymphocytes, Regulatory , CD40 Ligand , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Immunosuppression Therapy , Phenotype
4.
Int J Mol Sci ; 24(18)2023 Sep 07.
Article in English | MEDLINE | ID: mdl-37762093

ABSTRACT

Single-nucleotide polymorphism rs71327024 located in the human 3p21.31 locus has been associated with an elevated risk of hospitalization upon SARS-CoV-2 infection. The 3p21.31 locus contains several genes encoding chemokine receptors potentially relevant to severe COVID-19. In particular, CXCR6, which is prominently expressed in T lymphocytes, NK, and NKT cells, has been shown to be involved in the recruitment of immune cells to non-lymphoid organs in chronic inflammatory and respiratory diseases. In COVID-19, CXCR6 expression is reduced in lung resident memory T cells from patients with severe disease as compared to the control cohort with moderate symptoms. We demonstrate here that rs71327024 is located within an active enhancer that augments the activity of the CXCR6 promoter in human CD4+ T lymphocytes. The common rs71327024(G) variant makes a functional binding site for the c-Myb transcription factor, while the risk rs71327024(T) variant disrupts c-Myb binding and reduces the enhancer activity. Concordantly, c-Myb knockdown in PMA-treated Jurkat cells negates rs71327024's allele-specific effect on CXCR6 promoter activity. We conclude that a disrupted c-Myb binding site may decrease CXCR6 expression in T helper cells of individuals carrying the minor rs71327024(T) allele and thus may promote the progression of severe COVID-19 and other inflammatory pathologies.


Subject(s)
COVID-19 , Humans , COVID-19/genetics , Hospitalization , Promoter Regions, Genetic , Receptors, CXCR6/genetics , SARS-CoV-2 , T-Lymphocytes, Helper-Inducer
5.
Biochemistry (Mosc) ; 88(1): 13-21, 2023 Jan.
Article in English | MEDLINE | ID: mdl-37068869

ABSTRACT

B lymphocytes play an important role in the regulation of immune response in both normal and pathological conditions. Traditionally, the main functions of B cells were considered to be antibody production and antigen presentation, but in recent decades there have been discovered several subpopulations of regulatory B lymphocytes (Bregs), which maintain immunological tolerance and prevent overactivation of the immune system. Memory (mBregs, CD19+CD24hiCD27+) and transitional (tBregs, CD19+CD24hiCD38hi) subpopulations of Bregs are usually considered in the context of studying the role of these B cells in various human pathologies. However, the mechanisms by which these Breg subpopulations exert their immunosuppressive activity remain poorly understood. In this work, we used bioinformatic analysis of open-source RNA sequencing data to propose potential mechanisms of B cell-mediated immunosuppression. Analysis of differential gene expression before and after activation of these subpopulations allowed us to identify six candidate molecules that may determine the functionality of mBregs and tBregs. IL4I1-, SIRPA-, and SLAMF7-dependent mechanisms of immunosuppression may be characteristic of both Breg subsets, while NID1-, CST7-, and ADORA2B-dependent mechanisms may be predominantly characteristic of tBregs. In-depth understanding of the molecular mechanisms of anti-inflammatory immune response of B lymphocytes is an important task for both basic science and applied medicine and could facilitate the development of new approaches to the therapy of complex diseases.


Subject(s)
B-Lymphocytes, Regulatory , Humans , B-Lymphocytes, Regulatory/metabolism , B-Lymphocytes, Regulatory/pathology , Immune Tolerance , Immunosuppressive Agents/metabolism , Immunosuppression Therapy , L-Amino Acid Oxidase/metabolism
7.
Cells ; 11(12)2022 06 19.
Article in English | MEDLINE | ID: mdl-35741098

ABSTRACT

TNF and LTα are structurally related cytokines of the TNF superfamily. Their genes are located in close proximity to each other and to the Ltb gene within the TNF/LT locus inside MHC. Unlike Ltb, transcription of Tnf and of Lta is tightly controlled, with the Tnf gene being an immediate early gene that is rapidly induced in response to various inflammatory stimuli. Genes of the TNF/LT locus play a crucial role in lymphoid tissue organogenesis, although some aspects of their specific contribution remain controversial. Here, we present new findings and discuss the distinct contribution of TNF produced by ILC3 cells to Peyer's patch organogenesis.


Subject(s)
Lymphotoxin-alpha , Peyer's Patches , Animals , Lymphoid Tissue , Mice , Mice, Knockout , Organogenesis/genetics , Tumor Necrosis Factors/metabolism
8.
Sci Rep ; 11(1): 14120, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34239022

ABSTRACT

Transforming growth factor beta (TGF-ß) is the main cytokine responsible for the induction of the epithelial-mesenchymal transition of breast cancer cells, which is a hallmark of tumor transformation to the metastatic phenotype. Recently, research demonstrated that the chemokine CCL2 gene expression level directly correlates with the TGF-ß activity in breast cancer patients. CCL2 attracts tumor-associated macrophages and is, therefore, considered as an important inductor of breast cancer progression; however, the precise mechanisms underlying its regulation by TGF-ß are unknown. Here, we studied the behavior of the CCL2 gene in MDA-MB-231 and HCC1937 breast cancer cells representing mesenchymal-like phenotype activated by TGF-ß. Using bioinformatics, deletion screening and point mutagenesis, we identified binding sites in the CCL2 promoter and candidate transcription factors responsible for its regulation by TGF-ß. Among these factors, only the knock-down of EGR1 and RXRA made CCL2 promoter activity independent of TGF-ß. These factors also demonstrated binding to the CCL2 promoter in a TGF-ß-dependent manner in a chromatin immunoprecipitation assay, and point mutations in the EGR1 and RXRA binding sites totally abolished the effect of TGF-ß. Our results highlight the key role of EGR1 and RXRA transcription factors in the regulation of CCL2 gene in response to TGF-ß pathway.


Subject(s)
Chemokine CCL2/metabolism , Early Growth Response Protein 1/metabolism , Retinoid X Receptor alpha/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Triple Negative Breast Neoplasms/metabolism , Base Sequence , Binding Sites , Cell Line, Tumor , Chemokine CCL2/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Point Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
9.
Biochim Biophys Acta Mol Basis Dis ; 1866(3): 165626, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31785408

ABSTRACT

Toll-like receptor 4 (TLR4) is an innate immunity receptor predominantly expressed on myeloid cells and involved in the development of various diseases, many of them with complex genetics. Here we present data on functionality of single nucleotide polymorphism rs7873784 located in the 3'-untranslated region (3'-UTR) of TLR4 gene and associated with various pathologies involving chronic inflammation. We demonstrate that TLR4 3'-UTR strongly enhanced the activity of TLR4 promoter in U937 human monocytic cell line while minor rs7873784(C) allele created a binding site for transcription factor PU.1 (encoded by SPI1 gene), a known regulator of TLR4 expression. Increased binding of PU.1 further augmented the TLR4 transcription while PU.1 knockdown or complete disruption of the PU.1 binding site abrogated the effect. We hypothesize that additional functional PU.1 site may increase TLR4 expression in individuals carrying minor C variant of rs7873784 and modulate the development of certain pathologies, such as rheumatoid arthritis and type-2 diabetes mellitus.


Subject(s)
Arthritis, Rheumatoid/genetics , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide/genetics , Proto-Oncogene Proteins/genetics , Toll-Like Receptor 4/genetics , Trans-Activators/genetics , 3' Untranslated Regions/genetics , Alleles , Cell Line , Cell Line, Tumor , HEK293 Cells , Humans , Promoter Regions, Genetic/genetics , U937 Cells
10.
Cancer Lett ; 467: 96-106, 2019 12 28.
Article in English | MEDLINE | ID: mdl-31326556

ABSTRACT

Tumor-associated antigen (TAA)-specific autoantibodies have been widely implicated in cancer diagnosis. However, cancer cell lines that are typically exploited as candidate TAA sources in immunoproteomic studies may fail to accurately represent the autoantigen-ome of lower-grade neoplasms. Here, we established an integrated strategy for the identification of disease-relevant TAAs in thyroid neoplasia, which combined NRASQ61R oncogene expression in non-tumorous thyroid Nthy-ori 3-1 cells with a multi-dimensional proteomic technique DISER that consisted of profiling NRASQ61R-induced proteins using 2-dimensional difference gel electrophoresis (2D-DIGE) coupled with serological proteome analysis (SERPA) of the TAA repertoire of patients with thyroid encapsulated follicular-patterned/RAS-like phenotype (EFP/RLP) tumors. We identified several candidate cell-based (nicotinamide phosphoribosyltransferase NAMPT, glutamate dehydrogenase GLUD1, and glutathione S-transferase omega-1 GSTO1) and autoantibody (fumarate hydratase FH, calponin-3 CNN3, and pyruvate kinase PKM autoantibodies) biomarkers, including NRASQ61R-induced TAA phosphoglycerate kinase 1 PGK1. Meta-profiling of the reactivity of the identified autoantibodies across an independent SERPA series implicated the PKM autoantibody as a histological phenotype-independent biomarker of thyroid malignancy (11/38 (29%) patients with overtly malignant and uncertain malignant potential (UMP) tumors vs 0/22 (p = 0.0046) and 0/20 (p = 0.011) patients with non-invasive EFP/RLP tumors and healthy controls, respectively). PGK1 and CNN3 autoantibodies were identified as EFP/RLP-specific biomarkers, potentially suitable for further discriminating tumors with different malignant potential (PGK1: 7/22 (32%) patients with non-invasive EFP/RLP tumors vs 0/38 (p = 0.00044) and 0/20 (p = 0.0092) patients with other tumors and healthy controls, respectively; СNN3: 9/29 (31%) patients with malignant and borderline EFP/RLP tumors vs 0/31 (p = 0.00068) and 0/20 (p = 0.0067) patients with other tumors and healthy controls, respectively). The combined use of PKM, CNN3, and PGK1 autoantibodies allowed the reclassification of malignant/UMP tumor risk in 19/41 (46%) of EFP/RLP tumor patients. Taken together, we established an experimental pipeline DISER for the concurrent identification of cell-based and TAA biomarkers. The combination of DISER with in vitro oncogene expression allows further targeted identification of oncogene-induced TAAs. Using this integrated approach, we identified candidate autoantibody biomarkers that might be of value for differential diagnostic purposes in thyroid neoplasia.


Subject(s)
Autoantibodies/metabolism , GTP Phosphohydrolases/genetics , Membrane Proteins/genetics , Proteomics/methods , Thyroid Neoplasms/diagnosis , Biomarkers, Tumor/metabolism , Case-Control Studies , Cell Line, Tumor , Early Detection of Cancer , Female , GTP Phosphohydrolases/immunology , Humans , Membrane Proteins/immunology , Mutation , Thyroid Neoplasms/immunology
11.
Genes (Basel) ; 10(2)2019 01 23.
Article in English | MEDLINE | ID: mdl-30678091

ABSTRACT

Genome-wide association studies (GWASes) revealed several single-nucleotide polymorphisms (SNPs) in the human 17q12-21 locus associated with autoimmune diseases. However, follow-up studies are still needed to identify causative SNPs directly mediating autoimmune risk in the locus. We have chosen six SNPs in high linkage disequilibrium with the GWAS hits that showed the strongest evidence of causality according to association pattern and epigenetic data and assessed their functionality in a local genomic context using luciferase reporter system. We found that rs12946510, rs4795397, rs12709365, and rs8067378 influenced the reporter expression level in leukocytic cell lines. The strongest effect visible in three distinct cell types was observed for rs12946510 that is predicted to alter MEF2A/C and FOXO1 binding sites.


Subject(s)
Autoimmune Diseases/genetics , Chromosomes, Human, Pair 17/genetics , Polymorphism, Single Nucleotide , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Humans , Jurkat Cells , Linkage Disequilibrium , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Quantitative Trait Loci
12.
Int J Mol Sci ; 19(12)2018 Dec 09.
Article in English | MEDLINE | ID: mdl-30544846

ABSTRACT

Interleukin 33 (IL-33) is a cytokine constitutively expressed by various cells of barrier tissues that contribute to the development of inflammatory immune responses. According to its function as an alarmin secreted by lung and airway epithelium, IL-33 plays a significant role in pathogenesis of allergic disorders. IL-33 is strongly involved in the pathogenesis of asthma, anaphylaxis, allergy and dermatitis, and genetic variations in IL33 locus are associated with increased susceptibility to asthma. Genome-wide association studies have identified risk "T" allele of the single-nucleotide polymorphism rs4742170 located in putative IL33 enhancer area as susceptible variant for development of specific wheezing phenotype in early childhood. Here, we demonstrate that risk "T" rs4742170 allele disrupts binding of glucocorticoid receptor (GR) transcription factor to IL33 putative enhancer. The IL33 promoter/enhancer constructs containing either 4742170 (T) allele or point mutations in the GR-binding site, were significantly more active and did not respond to cortisol in a pulmonary epithelial cell line. At the same time, the constructs containing rs4742170 (C) allele with a functional GR-binding site were less active and further inhibitable by cortisol. The latter effect was GR-dependent as it was completely abolished by GR-specific siRNA. This mechanism may explain the negative effect of the rs4742170 (T) risk allele on the development of wheezing phenotype that strongly correlates with allergic sensitization in childhood.


Subject(s)
Alleles , Enhancer Elements, Genetic/genetics , Interleukin-33/genetics , Introns/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Glucocorticoid/metabolism , Respiratory Sounds/genetics , Base Sequence , Binding Sites , Cell Line, Tumor , Child, Preschool , Humans , Hydrocortisone/pharmacology , Phenotype , Phosphorylation/drug effects , Promoter Regions, Genetic
13.
Int J Mol Sci ; 19(10)2018 Sep 25.
Article in English | MEDLINE | ID: mdl-30257479

ABSTRACT

Cytokine interleukin 33 (IL-33) is constitutively expressed by epithelial barrier cells, and promotes the development of humoral immune responses. Along with other proinflammatory mediators released by the epithelium of airways and lungs, it plays an important role in a number of respiratory pathologies. In particular, IL-33 significantly contributes to pathogenesis of allergy and asthma; genetic variations in the IL33 locus are associated with increased susceptibility to asthma. Large-scale genome-wide association studies have identified minor "G" allele of the single-nucleotide polymorphism rs928413, located in the IL33 promoter area, as a susceptible variant for early childhood and atopic asthma development. Here, we demonstrate that the rs928413(G) allele creates a binding site for the cAMP response element-binding protein 1 (CREB1) transcription factor. In a pulmonary epithelial cell line, activation of CREB1, presumably via the p38 mitogen-activated protein kinases (MAPK) cascade, activates the IL33 promoter containing the rs928413(G) allele specifically and in a CREB1-dependent manner. This mechanism may explain the negative effect of the rs928413 minor "G" allele on asthma development.


Subject(s)
Asthma/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Interleukin-33/genetics , Polymorphism, Single Nucleotide , Alleles , Asthma/metabolism , Cell Line, Tumor , Child , Epithelial Cells/metabolism , Genetic Predisposition to Disease , Humans , Lung/cytology , Lung/metabolism , MAP Kinase Signaling System , Promoter Regions, Genetic , Protein Binding , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Transcriptional Activation
14.
Biochim Biophys Acta Mol Basis Dis ; 1864(10): 3211-3220, 2018 10.
Article in English | MEDLINE | ID: mdl-30006149

ABSTRACT

CD58 is expressed on the surface of antigen-presenting cells, including B-cells, and provides co-stimulation to regulatory T-cells (Treg) through CD2 receptor binding. Tregs appear to be essential suppressors of tissue-specific autoimmune responses. Thereby, CD58 plays protective role in multiple sclerosis (MS) and CD58 was identified among several loci associated with MS susceptibility. Minor (C) variant of the single-nucleotide polymorphism (SNP) rs1335532 is associated with lower MS risk according to genome-wide association studies (GWAS) and its presence correlates with higher CD58 mRNA levels in MS patients. We found that genomic region containing rs1335532 has enhancer properties and can significantly boost the CD58 promoter activity in lymphoblast cells. Using bioinformatics and pull-down assay we found that the protective (C) rs1335532 allele created functional binding site for ASCL2 transcription factor, a target of the Wnt signaling pathway. Both in B-lymphoblastoid cell lines and in primary B-cells, as well as in a monocytic cell line, activation of Wnt signaling resulted in an increased CD58 promoter activity in the presence of the protective but not the risk allele of rs1335532, whereas ASCL2 knockdown abrogated this effect. In summary, our results suggest that ASCL2 mediates the protective function of rs1335532 minor (C) allele in MS.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , CD58 Antigens/genetics , Multiple Sclerosis/genetics , Polymorphism, Single Nucleotide , Up-Regulation , Alleles , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites , CD58 Antigens/chemistry , Cell Line, Tumor , Computational Biology/methods , Enhancer Elements, Genetic , Female , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Humans , Male , Multiple Sclerosis/metabolism , Promoter Regions, Genetic , Wnt Signaling Pathway
15.
Article in English | MEDLINE | ID: mdl-29535976

ABSTRACT

Toll-like receptor 4 (TLR4) initiates immune response against Gram-negative bacteria upon specific recognition of lipid A moiety of lipopolysaccharide (LPS), the major component of their cell wall. Some natural differences between LPS variants in their ability to interact with TLR4 may lead to either insufficient activation that may not prevent bacterial growth, or excessive activation which may lead to septic shock. In this study we evaluated the biological activity of LPS isolated from pathogenic strain of Campylobacter jejuni, the most widespread bacterial cause of foodborne diarrhea in humans. With the help of hydrophobic chromatography and MALDI-TOF mass spectrometry we showed that LPS from a C. jejuni strain O2A consists of both hexaacyl and tetraacyl forms. Since such hypoacylation can result in a reduced immune response in humans, we assessed the activity of LPS from C. jejuni in mouse macrophages by measuring its capacity to activate TLR4-mediated proinflammatory cytokine and chemokine production, as well as NFκB-dependent reporter gene transcription. Our data support the hypothesis that LPS acylation correlates with its bioactivity.


Subject(s)
Campylobacter jejuni/immunology , Campylobacter jejuni/metabolism , Foodborne Diseases/microbiology , Lipopolysaccharides/pharmacology , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/immunology , Animals , Campylobacter jejuni/pathogenicity , Cytokines/metabolism , Interferon Regulatory Factor-3/genetics , Interleukin-1beta/metabolism , Interleukin-6 , Lipid A/immunology , Lipid A/isolation & purification , Lipid A/pharmacology , Lipopolysaccharides/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , RNA, Small Interfering , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/metabolism
16.
Biochim Biophys Acta Gene Regul Mech ; 1860(12): 1169-1178, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29107083

ABSTRACT

Many types of chemotherapeutic agents induce of DNA-damage that is accompanied by activation of p53 tumor suppressor, a key regulator of tumor development and progression. In our previous study we demonstrated that p53 could repress CXCR5 chemokine receptor gene in MCF-7 breast cancer cells via attenuation of NFkB activity. In this work we aimed to determine individual roles of p53 family members in the regulation of CXCR5 gene expression under genotoxic stress. DNA-alkylating agent methyl methanesulfonate caused a reduction in CXCR5 expression not only in parental MCF-7 cells but also in MCF-7-p53off cells with CRISPR/Cas9-mediated inactivation of the p53 gene. Since p53 knockout was associated with elevated expression of its p63 and p73 homologues, we knocked out p63 using CRISPR/Cas9 system and knocked down p73 using specific siRNA. The CXCR5 promoter activity, CXCR5 expression and CXCL13-directed migration in MCF-7 cells with inactivation of all three p53 family genes were completely insensitive to genotoxic stress, while pairwise p53+p63 or p53+p73 inactivation resulted in partial effects. Using deletion analysis and site-directed mutagenesis, we demonstrated that effects of NFkB on the CXCR5 promoter inversely correlated with p63 and p73 levels. Thus, all three p53 family members mediate the effects of genotoxic stress on the CXCR5 promoter using the same mechanism associated with attenuation of NFkB activity. Understanding of this mechanism could facilitate prognosis of tumor responses to chemotherapy.


Subject(s)
DNA Damage , Gene Expression Regulation, Neoplastic , Membrane Proteins/physiology , Receptors, CXCR5/genetics , Tumor Protein p73/physiology , Tumor Suppressor Protein p53/physiology , CRISPR-Cas Systems , Female , Humans , MCF-7 Cells , Methyl Methanesulfonate/pharmacology , NF-kappa B/physiology , Promoter Regions, Genetic
17.
Sci Rep ; 7(1): 2339, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28539628

ABSTRACT

The Src-family tyrosine kinase Lck is an enzyme associated with the CD4 and CD8 co-receptors and promoting signaling through the T cell receptor (TCR) complex. The levels of Lck expression and activity change during the development and differentiation of T cells. Here we show that Lck expression is higher in Th1 cells as compared to Th2 cells. Ectopic overexpression of Lck in Th2 cells results in increased expression of CD4 co-receptor and enhanced S73 phosphorylation of transcription factor c-Jun. Our findings indicate that TCR-mediated signaling in Th2 cells may be directly attenuated by Lck protein expression level.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Antigen, T-Cell/metabolism , Th2 Cells/metabolism , Animals , Cells, Cultured , Gene Expression Profiling , HEK293 Cells , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mice, Inbred C57BL , Phosphorylation , Serine/metabolism , Signal Transduction , Th1 Cells/metabolism
18.
PLoS One ; 12(2): e0172681, 2017.
Article in English | MEDLINE | ID: mdl-28234966

ABSTRACT

We studied functional effect of rs12722489 single nucleotide polymorphism located in the first intron of human IL2RA gene on transcriptional regulation. This polymorphism is associated with multiple autoimmune conditions (rheumatoid arthritis, multiple sclerosis, Crohn's disease, and ulcerative colitis). Analysis in silico suggested significant difference in the affinity of estrogen receptor (ER) binding site between alternative allelic variants, with stronger predicted affinity for the risk (G) allele. Electrophoretic mobility shift assay showed that purified human ERα bound only G variant of a 32-bp genomic sequence containing rs12722489. Chromatin immunoprecipitation demonstrated that endogenous human ERα interacted with rs12722489 genomic region in vivo and DNA pull-down assay confirmed differential allelic binding of amplified 189-bp genomic fragments containing rs12722489 with endogenous human ERα. In a luciferase reporter assay, a kilobase-long genomic segment containing G but not A allele of rs12722489 demonstrated enhancer properties in MT-2 cell line, an HTLV-1 transformed human cell line with a regulatory T cell phenotype.


Subject(s)
Estrogen Receptor alpha/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Polymorphism, Single Nucleotide , Response Elements , T-Lymphocytes, Regulatory/metabolism , Alleles , Base Sequence , Binding Sites , Cell Line, Transformed , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Estrogen Receptor alpha/metabolism , Gene Expression Regulation , Genes, Reporter , Human T-lymphotropic virus 1/genetics , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Introns , Luciferases/genetics , Luciferases/metabolism , Protein Binding , T-Lymphocytes, Regulatory/cytology
19.
Cytokine ; 89: 127-135, 2017 01.
Article in English | MEDLINE | ID: mdl-26854213

ABSTRACT

The link between inflammation and cancer was first proposed by R. Virchow. It was later realized that it is chronic inflammation that may promote cancer, whereas acute inflammation can actually block tumor development or even result in cure. Many molecular mediators of these diverse processes have been characterized only during the past 3 decades thanks to the advances in molecular and cellular techniques, as well as due to technologies of reverse genetics. In this chapter we discuss the role of Toll-like receptor (TLR) 4 signaling in cancer and contributions of proinflammatory cytokine signaling (whose expression may be driven by TLR-mediated signals) to tumor-promoting microenvironment. We also discuss recent clinical advances to target these pro-tumorigenic pathways at distinct stages of tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Tumor Microenvironment/immunology , Animals , Cell Transformation, Neoplastic/pathology , Cytokines/immunology , Humans , Neoplasms/pathology
20.
Gene ; 602: 50-56, 2017 Feb 20.
Article in English | MEDLINE | ID: mdl-27876533

ABSTRACT

IL2RA gene encodes the alpha subunit of a high-affinity receptor for interleukin-2 which is expressed by several distinct populations of lymphocytes involved in autoimmune processes. A large number of polymorphic alleles of the IL2RA locus are associated with the development of various autoimmune diseases. With bioinformatics analysis we the dissected the first intron of the IL2RA gene and selected several single nucleotide polymorphisms (SNPs) that may influence the regulation of the IL2RA gene in cell types relevant to autoimmune pathology. We described five enhancers containing the selected SNPs that stimulated activity of the IL2RA promoter in a cell-type specific manner, and tested the effect of specific SNP alleles on activity of the respective enhancers (E1 to E5, labeled according to the distance to the promoter). The E4 enhancer with minor T variant of rs61839660 SNP demonstrated reduced activity due to disrupted binding of MEF2A/C transcription factors (TFs). Neither rs706778 nor rs706779 SNPs, both associated with a number of autoimmune diseases, had any effect on the activity of the enhancer E2. However, rare variants of several SNPs (rs139767239, rs115133228, rs12722502, rs12722635) genetically linked to either rs706778 and/or rs706779 significantly influenced the activity of E1, E3 and E5 enhancers, presumably by disrupting EBF1, GABPA and ELF1 binding sites.


Subject(s)
Interleukin-2 Receptor alpha Subunit/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Cell Line , Enhancer Elements, Genetic , Genetic Predisposition to Disease , Humans , Introns , Jurkat Cells , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...