Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 13035, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35906355

ABSTRACT

Somatic mutations and changes in expression of RAD21 are common in many types of cancer. Moreover, sub-optimal levels of RAD21 expression in early development can result in cohesinopathies. Altered RAD21 levels can result directly from mutations in the RAD21 gene. However, whether DNA variants outside of the RAD21 gene could control its expression and thereby contribute to cancer and developmental disease is unknown. In this study, we searched for genomic variants that modify RAD21expression to determine their potential to contribute to development or cancer by RAD21 dysregulation. We searched 42,953,834 genomic variants for a spatial-eQTL association with the transcription of RAD21. We identified 123 significant associations (FDR < 0.05), which are local (cis) or long-distance (trans) regulators of RAD21 expression. The 123 variants co-regulate a further seven genes (AARD, AKAP11, GRID1, KCNIP4, RCN1, TRIOBP, and USP32), enriched for having Sp2 transcription factor binding sites in their promoter regions. The Sp2 transcription factor and six of the seven genes had previously been associated with cancer onset, progression, and metastasis. Our results suggest that genome-wide variation in non-coding regions impacts on RAD21 transcript levels in addition to other genes, which then could impact on oncogenesis and the process of ubiquitination. This identification of distant co-regulation of oncogenes represents a strategy for discovery of novel genetic regions influencing cancer onset and a potential for diagnostics.


Subject(s)
Neoplasms , Sp2 Transcription Factor , Binding Sites , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Genetic Variation , Genome , Humans , Neoplasms/genetics , Sp2 Transcription Factor/genetics
2.
Exp Cell Res ; 411(1): 112972, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34914964

ABSTRACT

Calcification of the bicuspid aortic valve (BAV) involves differential expression of various RNA genes, which is achieved through complex regulatory networks that are controlled in part by transcription factors and microRNAs. We previously found that miR-195-5p regulates the osteogenic differentiation of valvular interstitial cells (VICs) by targeting the TGF-ß pathway. However, the transcriptional regulation of miR-195-5p in calcified BAV patients is not yet clear. In this study, stenotic aortic valve tissues from patients with BAVs and tricuspid aortic valves (TAVs) were collected. Candidate transcription factors of miR-195-5p were predicted by bioinformatics analysis and tested in diseased valves and in male porcine VICs. SP2 gene expression and the corresponding protein levels in BAV were significantly lower than those in TAV, and a low SP2 expression level environment in VICs resulted in remarkable increases in RNA expression levels of RUNX2, BMP2, collagen 1, MMP2, and MMP9 and the corresponding proteins. ChIP assays revealed that SP2 directly bound to the transcription promoter region of miR-195-5p. Cotransfection of SP2 shRNA and a miR-195-5p mimic in porcine VICs demonstrated that SP2 repressed SMAD7 expression via miR-195-5p, while knockdown of SP2 increased the mRNA expression of SMAD7 and the corresponding protein and attenuated Smad 2/3 expression. Immunofluorescence staining of diseased valves confirmed that the functional proteins of osteogenesis differentiation, including RUNX2, BMP2, collagen 1, and osteocalcin, were overexpressed in BAVs. In Conclusion, the transcription factor Sp2 is expressed at low levels in VICs from BAV patients, which has a negative impact on miR-195-5p expression by binding its promoter region and partially promotes calcification through a SMAD-dependent pathway.


Subject(s)
Bicuspid Aortic Valve Disease/pathology , Calcinosis/pathology , Osteoblasts/pathology , Smad7 Protein/metabolism , Sp2 Transcription Factor/metabolism , Transforming Growth Factor beta1/metabolism , Tricuspid Valve/pathology , Animals , Bicuspid Aortic Valve Disease/genetics , Bicuspid Aortic Valve Disease/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Calcinosis/genetics , Calcinosis/metabolism , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Humans , Male , MicroRNAs , Middle Aged , Osteoblasts/metabolism , Osteogenesis , Smad7 Protein/genetics , Sp2 Transcription Factor/genetics , Swine , Transforming Growth Factor beta1/genetics , Tricuspid Valve/metabolism
3.
Neuroscience ; 479: 22-34, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34687796

ABSTRACT

Transcription factors bind specific sequences upstream of the 5' end of their target genes to ensure proper spatiotemporal expression of the target gene. This study aims to demonstrate that the transcription factor SP2 regulates expression of the Ski gene, which has specific binding sites for SP2, and thus enables Ski to regulate astrocyte proliferation. The upstream regulation mechanism of astrocyte proliferation was explored to further regulate the formation of glial scar in specific time and space after spinal cord injury. JASPAR and UCSC databases were used to predict transcription factor binding and the threshold was gradually reduced to screen transcription factors upstream of Ski, leading to the identification of SP2. Next, we analyzed the correlation between the expression of SP2 and Ski in normal astrocytes and reactive astrocytes, as well as the changes in astrocyte proliferation. To confirm that SP2 regulates Ski during astrocyte proliferation, astrocytes were transfected siRNA targeting SP2 and then astrocyte proliferation were analyzed. Finally, a dual luciferase reporter assay and Chromatin immunoprecipitation (ChIP) assay confirmed that the promoter region of Ski contained a specific SP2 binding site. This is the first that SP2 has been identified and confirmed to play an important role in astrocyte proliferation by regulating Ski expression. These results may help identify novel targets for the treatment of spinal cord injury.


Subject(s)
Astrocytes , Spinal Cord Injuries , Cell Proliferation , Cells, Cultured , Gliosis , Humans , Sp2 Transcription Factor
4.
Cell Death Dis ; 12(2): 153, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542193

ABSTRACT

Studies have found that RNA-binding proteins (RBPs) are dysfunctional and play a significant regulatory role in the development of glioma. Based on The Cancer Genome Atlas database and the previous studies, we selected heterogeneous nuclear ribonucleoprotein (HNRNPD) as the research candidate and sought its downstream targeted genes. In the present study, HNRNPD, linc00707, and specific protein 2 (SP2) were highly expressed, while zinc fingers and homeboxes 2 (ZHX2) and miR-651-3p were remarkedly downregulated in glioma tissues and cells. HNRNPD, linc00707, and SP2 knockdown or ZHX2 and miR-651-3p overexpression suppressed glioma cells proliferation, migration, and invasion and vasculogenic mimicry (VM) formation. Knockdown of HNRNPD increased the stability of ZHX2 mRNA. ZHX2 bound to the promoter region of linc00707 and negatively regulate its expression. Linc00707 could bind with miR-651-3p, while miR-651-3p bound to the 3' untranslated region (3'UTR) of SP2 mRNA to negatively regulate its expression. The transcription factor SP2 directly bound to the promoter regions of the VM formation-related proteins MMP2, MMP9, and VE-cadherin, playing a role in promoting transcription in order to regulate the VM formation ability of glioma cells.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Molecular Mimicry , Neovascularization, Pathologic , RNA, Long Noncoding/metabolism , Sp2 Transcription Factor/metabolism , Transcription Factors/metabolism , 3' Untranslated Regions , Animals , Binding Sites , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , HEK293 Cells , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Homeodomain Proteins/genetics , Humans , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , RNA, Long Noncoding/genetics , Signal Transduction , Sp2 Transcription Factor/genetics , Transcription Factors/genetics
5.
Dokl Biochem Biophys ; 491(1): 67-69, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32483753

ABSTRACT

The transcriptomic profile associated with osteo- and adipogenic differentiation in growth-arrested multipotent mesenchymal stromal cells (MSCs) from human adipose tissue was analyzed in vitro at 20% (standard laboratory) and 5% (tissue-related) O2 levels. Compared with day 7, at 5% O2 on day 14 spontaneous upregulation of osteo- (RUNX2, SP7, BGLAP, and SPP1) and adipogenic differentiation (CEBPA, PPARG, and ADIPOQ) genes in MSCs was observed (p < 0.05). Thus, upon expansion under tissue-related O2, MSCs demonstrated a bipotent transcriptomic profile, which may contribute to the improvement of their hematopoiesis-supportive function.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Mesenchymal Stem Cells/metabolism , Oxygen/metabolism , Adipocytes/metabolism , Adipogenesis , Adiponectin/metabolism , Adipose Tissue/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Cell Hypoxia , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Osteocalcin/metabolism , Osteogenesis , Osteopontin/metabolism , PPAR gamma/metabolism , Sp2 Transcription Factor/metabolism , Transcriptome
6.
Cancer Med ; 9(10): 3592-3603, 2020 05.
Article in English | MEDLINE | ID: mdl-32160655

ABSTRACT

OBJECTIVE: To explore the biological function and molecular mechanism of Sp2 in hepatocellular carcinoma (HCC). METHODS: Tissue microarray immunohistochemistry and western blot were used to study the expression of Sp2 in hepatocellular tissue and adjacent non-neoplastic tissues (ANT). In HCC cell lines, the role of Sp2 was determined by in vitro experiments such as CCK8, clone formation test, Transwell assay, wound-healing assay, and flow cytometry apoptotic analysis, and its possible mechanism was analyzed. RESULTS: Compared with ANT, Sp2 expression in HCC tissues was significantly up-regulated, which was strongly associated with stage of tumor and poor prognosis of patients. TCGA database were further confirmed these results. Besides, functional studies had shown that Sp2 knockdown not only leads to a decrease in cell proliferation and an increase in cell apoptosis but also inhibits the cells' abilities of migration and invasion. Sp2 silencing could inhibit the expression of TRIB3 protein and down-regulate the endoplasmic reticulum stress (ERS) level of HCC. CONCLUSION: Sp2 may play a part in promoting cancer by regulating TRIB3 protein, which may be a factor of prognostic and a potential new therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins/genetics , Liver Neoplasms/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Repressor Proteins/genetics , Sp2 Transcription Factor/genetics , Apoptosis/genetics , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Endoplasmic Reticulum Stress/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hep G2 Cells , Humans , Immunohistochemistry , In Vitro Techniques , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/metabolism , Sp2 Transcription Factor/metabolism
7.
Development ; 147(4)2020 02 21.
Article in English | MEDLINE | ID: mdl-32086334

ABSTRACT

Cortical development involves a switch from the self-amplification of stem cells to the generation of neuron and glia by progenitors. A new paper in Development investigates the molecular control of mitosis in these two stages, using simultaneous labelling and gene knockout in clones in the developing mouse brain. We caught up the paper's two authors Caroline Johnson and her supervisor Troy Ghashghaei, Professor of Neurobiology at the College of Veterinary Medicine at North Carolina State University, to find out more.


Subject(s)
Developmental Biology/methods , Animals , Brain/embryology , Cell Proliferation , Developmental Biology/history , History, 21st Century , Mice , Mitosis , Sp2 Transcription Factor/physiology
8.
Development ; 147(4)2020 02 21.
Article in English | MEDLINE | ID: mdl-32001437

ABSTRACT

Cellular and molecular mechanisms underlying the switch from self-amplification of cortical stem cells to neuronal and glial generation are incompletely understood, despite their importance for neural development. Here, we have investigated the role of the transcription factor specificity protein 2 (Sp2) in expansive and neurogenic divisions of the developing cerebral cortex by combining conditional genetic deletion with the mosaic analysis with double markers (MADM) system in mice. We find that loss of Sp2 in progenitors undergoing neurogenic divisions results in prolonged mitosis due to extension of early mitotic stages. This disruption is correlated with depletion of the populations of upper layer neurons in the cortex. In contrast, early cortical neural stem cells proliferate and expand normally in the absence of Sp2. These results indicate a stage-specific requirement for Sp2 in neural stem and progenitor cells, and reveal mechanistic differences between the early expansive and later neurogenic periods of cortical development.This article has an associated 'The people behind the papers' interview.


Subject(s)
Cerebral Cortex/embryology , Neural Stem Cells/cytology , Sp2 Transcription Factor/genetics , Sp2 Transcription Factor/physiology , Alleles , Animals , Cell Differentiation , Cell Division , Cell Lineage , Cell Proliferation , Female , Gene Deletion , Genetic Markers , Male , Mice , Mice, Transgenic , Mitosis , Mutation , Phenotype
9.
Cell Mol Life Sci ; 77(9): 1793-1810, 2020 May.
Article in English | MEDLINE | ID: mdl-31375868

ABSTRACT

The universal nine-amino-acid transactivation domains (9aaTADs) have been identified in numerous transcription activators. Here, we identified the conserved 9aaTAD motif in all nine members of the specificity protein (SP) family. Previously, the Sp1 transcription factor has been defined as a glutamine-rich activator. We showed by amino acid substitutions that the glutamine residues are completely dispensable for 9aaTAD function and are not conserved in the SP family. We described the origin and evolutionary history of 9aaTADs. The 9aaTADs of the ancestral Sp2 gene became inactivated in early chordates. We next discovered that an accumulation of valines in 9aaTADs inactivated their transactivation function and enabled their strict conservation during evolution. Subsequently, in chordates, Sp2 has duplicated and created new paralogs, Sp1, Sp3, and Sp4 (the SP1-4 clade). During chordate evolution, the dormancy of the Sp2 activation domain lasted over 100 million years. The dormant but still intact ancestral Sp2 activation domains allowed diversification of the SP1-4 clade into activators and repressors. By valine substitution in the 9aaTADs, Sp1 and Sp3 regained their original activator function found in ancestral lower metazoan sea sponges. Therefore, the vertebrate SP1-4 clade could include both repressors and activators. Furthermore, we identified secondary 9aaTADs in Sp2 introns present from fish to primates, including humans. In the gibbon genome, introns containing 9aaTADs were used as exons, which turned the Sp2 gene into an activator. Similarly, we identified introns containing 9aaTADs used conditionally as exons in the (SP family-unrelated) transcription factor SREBP1, suggesting that the intron-9aaTAD reservoir is a general phenomenon.


Subject(s)
Evolution, Molecular , Gene Expression Regulation , Introns/genetics , Sp2 Transcription Factor/antagonists & inhibitors , Sp2 Transcription Factor/genetics , Valine/metabolism , Amino Acid Sequence , Animals , Gene Duplication , Humans , Phylogeny , Sequence Homology , Sp2 Transcription Factor/metabolism , Transcriptional Activation , Valine/genetics
10.
BMC Cancer ; 19(1): 746, 2019 Jul 30.
Article in English | MEDLINE | ID: mdl-31362717

ABSTRACT

PURPOSE: Expression of human chorionic gonadotropin beta subunit by cancers is extensively documented, yet regulation of the multiple genes that can code for this protein is poorly understood. The aim of the study was to examine the mechanisms regulating CGB gene expression in ovarian cancer. METHODS: Expression of CGB genes and SP1, SP3, TFAP2A transcription factor genes was evaluated by RT-qPCR. The methylation status of CGB genes promoter regions was examined by methylation-specific PCR. RESULTS: mRNA arising from multiple CGB genes was detected in both ovarian control and malignant tissues. However, expression of CGB3-9 genes was shown to be significantly higher in malignant than healthy ovarian tissues. CGB1 and CGB2 transcripts were shown to be present in 20% of ovarian cancers, but were not detected in any of the control samples. Malignant tissues were characterized by DNA demethylation of CGB promoter regions. In ovarian cancer CGB expression positively correlated with TFAP2A transcripts level and expression of TFAP2A transcription factor was significantly higher in cancer than in control tissues. In contrast SP3 expression level was significantly lower in ovarian tumours than in control ovarian tissue. CONCLUSIONS: In ovarian cancers increased expression of human chorionic gonadotropin beta subunit is associated with demethylation of CGB promoter regions. CGB3-9 expression level strongly correlates with expression of the TFAP2A transcription factor. Presence of mRNA arising from CGB1 and CGB2 genes appears to be a unique feature of a subset of ovarian cancers.


Subject(s)
Chorionic Gonadotropin, beta Subunit, Human/genetics , Gene Expression Regulation, Neoplastic/genetics , Ovarian Neoplasms/genetics , DNA Methylation/genetics , Demethylation , Female , Humans , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Sp1 Transcription Factor/genetics , Sp2 Transcription Factor/genetics , Transcription Factor AP-2/genetics , Transcription, Genetic
11.
J Biol Chem ; 293(50): 19250-19262, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30337366

ABSTRACT

Different transcription factors operate together at promoters and enhancers to regulate gene expression. Transcription factors either bind directly to their target DNA or are tethered to it by other proteins. The transcription factor Sp2 serves as a paradigm for indirect genomic binding. It does not require its DNA-binding domain for genomic DNA binding and occupies target promoters independently of whether they contain a cognate DNA-binding motif. Hence, Sp2 is strikingly different from its closely related paralogs Sp1 and Sp3, but how Sp2 recognizes its targets is unknown. Here, we sought to gain more detailed insights into the genomic targeting mechanism of Sp2. ChIP-exo sequencing in mouse embryonic fibroblasts revealed genomic binding of Sp2 to a composite motif where a recognition sequence for TALE homeoproteins and a recognition sequence for the trimeric histone-fold domain protein nuclear transcription factor Y (Nf-y) are separated by 11 bp. We identified a complex consisting of the TALE homeobox protein Prep1, its partner PBX homeobox 1 (Pbx1), and Nf-y as the major partners in Sp2-promoter interactions. We found that the Pbx1:Prep1 complex together with Nf-y recruits Sp2 to co-occupied regulatory elements. In turn, Sp2 potentiates binding of Pbx1:Prep1 and Nf-y. We also found that the Sp-box, a short sequence motif close to the Sp2 N terminus, is crucial for Sp2's cofactor function. Our findings reveal a mechanism by which the DNA binding-independent activity of Sp2 potentiates genomic loading of Pbx1:Prep1 and Nf-y to composite motifs present in many promoters of highly expressed genes.


Subject(s)
CCAAT-Binding Factor/metabolism , Genomics , Homeodomain Proteins/metabolism , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Sp2 Transcription Factor/metabolism , Animals , CCAAT-Binding Factor/chemistry , Cell Line , Histones/metabolism , Mice , Nucleotide Motifs , Protein Binding , Protein Transport , Sp2 Transcription Factor/chemistry , Zinc Fingers
13.
Sci Rep ; 7: 41204, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28145470

ABSTRACT

Although the search for quantitative trait loci for behaviour remains a considerable challenge, the complicated genetic architecture of quantitative traits is beginning to be understood. The current project utilised heterogeneous stock (HS) male mice (n = 580) to investigate the genetic basis for brain weights, activity, anxiety and cognitive phenotypes. We identified 126 single nucleotide polymorphisms (SNPs) in genes involved in regulation of neurotransmitter systems, nerve growth/death and gene expression, and subsequently investigated their associations with changes in behaviour and/or brain weights in our sample. We found significant associations between four SNP-phenotype pairs, after controlling for multiple testing. Specificity protein 2 (Sp2, rs3708840), tryptophan hydroxylase 1 (Tph1, rs262731280) and serotonin receptor 3A (Htr3a, rs50670893) were associated with activity/anxiety behaviours, and microtubule-associated protein 2 (Map2, rs13475902) was associated with cognitive performance. All these genes except for Tph1 were expressed in the brain above the array median, and remained significantly associated with relevant behaviours after controlling for the family structure. Additionally, we found evidence for a correlation between Htr3a expression and activity. We discuss our findings in the light of the advantages and limitations of currently available mouse genetic tools, suggesting further directions for association studies in rodents.


Subject(s)
Behavior, Animal , Brain/metabolism , Genetic Association Studies/methods , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Animals , Gene Expression , Genetic Heterogeneity , Male , Mice , Microtubule-Associated Proteins/genetics , Receptors, Serotonin, 5-HT3/genetics , Sp2 Transcription Factor/genetics , Tryptophan Hydroxylase/genetics
14.
J Neurochem ; 140(2): 245-256, 2017 01.
Article in English | MEDLINE | ID: mdl-27889927

ABSTRACT

Oligodendrocytes and Schwann cells are the myelinating glia of the vertebrate nervous system and by generation of myelin sheaths allow rapid saltatory conduction. Previous in vitro work had pointed to a role of the zinc finger containing specificity proteins Sp1 and Sp3 as major regulators of glial differentiation and myelination. Here, we asked whether such a role is also evident in vivo using mice with specific deletions of Sp1 or Sp3 in myelinating glia. We also studied glia-specific conditional Sp2- and constitutive Sp4-deficient mice to include all related glutamine-rich Sp factors into our analysis. Surprisingly, we did not detect developmental Schwann cell abnormalities in any of the mutant mice. Oligodendrocyte development and differentiation was also not fundamentally affected as oligodendrocytes were present in all mouse mutants and retained their ability to differentiate and initiate myelin gene expression. The most severe defect we observed was a 50% reduction in Mbp- and proteolipid protein 1 (Plp1)-positive differentiating oligodendrocytes in Sp2 mutants at birth. Unexpectedly, glial development appeared undisturbed even in the joint absence of Sp1 and Sp3. We conclude that Sp2 has a minor effect on the differentiation of myelinating glia, and that glutamine-rich Sp proteins are not essential regulators of the process.


Subject(s)
Cell Differentiation/physiology , Glutamine/metabolism , Myelin Sheath/metabolism , Neuroglia/metabolism , Oligodendroglia/metabolism , Sp2 Transcription Factor/metabolism , Animals , Cells, Cultured , Gene Expression Regulation, Developmental/physiology , Myelin Basic Protein/metabolism , Rats , Schwann Cells/drug effects , Schwann Cells/metabolism
16.
J Leukoc Biol ; 100(5): 1213-1223, 2016 11.
Article in English | MEDLINE | ID: mdl-27256574

ABSTRACT

Th17 cells are involved in the immune response against pathogens, autoimmunity, and tumor progression. The differentiation of human Th17 cells requires the upregulation of RORγT, which in human cells is still not well understood. We identified 2 putative binding motifs for specificity protein transcription factors from the specificity protein/Kruppel-like factor family in the promoter of human RORγT and investigated the involvement of specificity proteins in the transcriptional regulation of this gene. To this end, a human lymphocytic cell line and in vitro-differentiated Th17 cells were used in promoter activity assays, in situ mutagenesis, chromatin immunoprecipitation, and real-time RT-PCR assays. In some experiments, specificity protein expression and activity was inhibited by siRNA and mithramycin A. The results showed that the transcription factor specificity protein 2 recognized binding motifs in the human RORγT promoter, which was critical for maintaining expression. Furthermore, specificity protein 2 was necessary for maximum IL-17 expression in in vitro-differentiated Th17 cells. These observations demonstrate the significant role of specificity protein 2 in the regulation of the Th17 signature transcription factor RORγT and the maintenance of the Th17 phenotype. The findings also suggest that specificity protein 2 plays a role in Th17-dependent physiologic and pathologic immune responses and might serve as a potential novel target for their modulation.


Subject(s)
Gene Expression Regulation , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Promoter Regions, Genetic/genetics , Sp2 Transcription Factor/physiology , Th17 Cells/metabolism , Animals , Base Sequence , Binding Sites , Chromatin Immunoprecipitation , Conserved Sequence , Gene Expression Regulation/drug effects , Humans , Interleukin-17/biosynthesis , Interleukin-17/genetics , Jurkat Cells , Mammals/genetics , Mutagenesis, Site-Directed , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Promoter Regions, Genetic/drug effects , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Nucleic Acid , Th17 Cells/cytology
17.
PLoS One ; 11(5): e0155821, 2016.
Article in English | MEDLINE | ID: mdl-27191968

ABSTRACT

Genetic analysis in the IL10-deficient mouse model revealed a modifier locus of experimental inflammatory bowel disease (IBD) on chromosome 18, with the allele of the strain C3H/HeJBir (C3Bir) conferring resistance and the allele of C57BL/6J (B6) conferring susceptibility. Differential Cd14 expression was associated with this background specific susceptibility to intestinal inflammation. Polymorphisms of the Cd14 promoter were found to be likely causative for strain specific expression, and Cd14-knockout mice revealed a protective role of this gene-product in experimental IBD. In this study, luciferase reporter assays confirmed an increased activity of the C3Bir derived Cd14 promoter compared to the one of B6. Promoter truncation experiments and site-directed mutagenesis in both strains resulted in reduced Cd14 promoter activity and confirmed that a central AP1 and the proximal SP1 transcription factor binding sites mediated the basal activity of the Cd14 promoter in the mouse. Moreover, a T to C exchange at position -259 replaced putative STAT1 and CDX1 sites in the Cd14 promoter from B6 by a SP2 site in C3Bir. Ablation of the Sp2 site through truncation was associated with a decreased promoter activity. Site-directed mutagenesis also demonstrated that the inactivation of SP2 led to a substantial loss of promoter activity in C3Bir. Performing electrophoretic mobility shift and supershift assays demonstrated interaction of SP2 with its potential binding site. In addition, retroviral-mediated overexpression of the SP2 transcription factor in primary bone marrow macrophages derived from C3Bir mice caused a significant increase in Cd14 transcription. These data characterized SP2 as important factor responsible for higher Cd14 expression and reduced IBD susceptibility mediated by the C3Bir allele.


Subject(s)
Colitis, Ulcerative/metabolism , Lipopolysaccharide Receptors/metabolism , Sp2 Transcription Factor/metabolism , Alleles , Animals , Cell Line , Colitis, Ulcerative/genetics , Lipopolysaccharide Receptors/genetics , Mice , Promoter Regions, Genetic , Protein Binding , Sp2 Transcription Factor/genetics
18.
Anal Bioanal Chem ; 408(13): 3453-74, 2016 May.
Article in English | MEDLINE | ID: mdl-26942738

ABSTRACT

Quantitative methods for detection of biological molecules are needed more than ever before in the emerging age of "omics" and "big data." Here, we provide an integrated approach for systematic analysis of the "lipidome" in tissue. To test our approach in a biological context, we utilized brain tissue selectively deficient for the transcription factor Specificity Protein 2 (Sp2). Conditional deletion of Sp2 in the mouse cerebral cortex results in developmental deficiencies including disruption of lipid metabolism. Silver (Ag) cationization was implemented for infrared matrix-assisted laser desorption electrospray ionization (IR-MALDESI) to enhance the ion abundances for olefinic lipids, as these have been linked to regulation by Sp2. Combining Ag-doped and conventional IR-MALDESI imaging, this approach was extended to IR-MALDESI imaging of embryonic mouse brains. Further, our imaging technique was combined with bottom-up shotgun proteomic LC-MS/MS analysis and western blot for comparing Sp2 conditional knockout (Sp2-cKO) and wild-type (WT) cortices of tissue sections. This provided an integrated omics dataset which revealed many specific changes to fundamental cellular processes and biosynthetic pathways. In particular, step-specific altered abundances of nucleotides, lipids, and associated proteins were observed in the cerebral cortices of Sp2-cKO embryos.


Subject(s)
Chromatography, Liquid/methods , Lipids/analysis , Mass Spectrometry/methods , Prosencephalon/metabolism , Spectrophotometry, Infrared/methods , Transcriptome , Animals , Mice , Mice, Knockout , Prosencephalon/embryology , Sp2 Transcription Factor/genetics
19.
Cell Physiol Biochem ; 38(3): 1040-54, 2016.
Article in English | MEDLINE | ID: mdl-26937945

ABSTRACT

BACKGROUND/AIMS: Overexpression of cytosolic sulfotransferase 2B1b (SULT2B1b) has been commonly found in colorectal and hepatocellular carcinoma, suggesting that SULT2B1b might act as a potential oncogenic protein. However, its clinical significance and biological role in gastric cancer progression remain largely unknown. METHODS: Expressions of SULT2B1b in clinical gastric cancer (GC) samples were examined using qRT-PCR and Western blot. RESULTS: SULT2B1b was markedly overexpressed in human GC samples, and positively correlated with vessel density and associated with poor clinical features. We also demonstrated that overexpression of SULT2B1b resulted in increased tumor angiogenesis and tumor growth in mouse GC models. In addition, ablation of SULT2B1b in human GC cells lines BGC823 and MKN45 decreased the capability of the cells to recruit endothelial cells. Moreover, depletion of SULT2B1b in GC cells reduced VEGF-A expression by downregulating SP1 and AP2. CONCLUSION: Our results suggested that the SULT2B1b-mediated angiogenic pathway could serve as biomarkers for GC diagnosis and prognosis, and suppressing SULT2B1b-mediated angiogenic signaling might be a promising strategy for developing novel GC treatment.


Subject(s)
Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Stomach Neoplasms/pathology , Sulfotransferases/genetics , Sulfotransferases/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , Neoplasm Transplantation , Prognosis , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Sp2 Transcription Factor/genetics , Sp2 Transcription Factor/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
20.
Biochem Biophys Res Commun ; 467(2): 341-7, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26431879

ABSTRACT

The addition of O-linked N-acetylglucosamine (O-GlcNAc) on serine or threonine modifies a myriad of proteins and regulates their function, stability and localization. O-GlcNAc modification is common among chromosome-associated proteins, such as transcription factors, suggesting its extensive involvement in gene expression regulation. In this study, we demonstrate the O-GlcNAc status of the Sp family members of transcription factors and the functional impact on their transcriptional activities. We highlight the presence of O-GlcNAc residues in Sp3 and Sp4, but not Sp2, as demonstrated by their enrichment in GlcNAc positive protein fractions and by detection of O-GlcNAc residues on Sp3 and Sp4 co-expressed in Escherichia coli together with O-GlcNAc transferase (OGT) using an O-GlcNAc-specific antibody. Deletion mutants of Sp3 and Sp4 indicate that the majority of O-GlcNAc sites reside in their N-terminal transactivation domain. Overall, using reporter gene assays and co-immunoprecipitations, we demonstrate a functional inhibitory role of O-GlcNAc modifications in Sp3 and Sp4 transcription factors. Thereby, our study strengthens the current notion that O-GlcNAc modification is an important regulator of protein interactome.


Subject(s)
Acetylglucosamine/metabolism , Protein Processing, Post-Translational , Sp3 Transcription Factor/metabolism , Sp4 Transcription Factor/metabolism , Transcription, Genetic , Escherichia coli , Genes, Reporter , HEK293 Cells , HeLa Cells , Humans , Luciferases/genetics , Luciferases/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Plasmids/chemistry , Plasmids/metabolism , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/metabolism , Signal Transduction , Sp2 Transcription Factor/genetics , Sp2 Transcription Factor/metabolism , Sp3 Transcription Factor/genetics , Sp4 Transcription Factor/genetics , Threonine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...