Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 57(1): 261-277, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31317491

ABSTRACT

In this study, we found that Sp1 was highly expressed in astrocytes, implying that Sp1 might be important for the function of astrocytes. Sp1/GFAP-Cre-ERT2 conditional knockout mice were constructed to study the role of Sp1 in astrocytes. Knockout of Sp1 in astrocytes altered astrocytic morphology and decreased GFAP expression in the cortex and hippocampus but did not affect cell viability. Loss of Sp1 in astrocytes decreased the number of neurons in the cortex and hippocampus. Conditioned medium from primary astrocytes with Sp1 knockout disrupted neuronal dendritic outgrowth and synapse formation, resulting in abnormal learning, memory, and motor behavior. Sp1 knockout in astrocytes altered gene expression, including decreasing the expression of Toll-like receptor 2 and Cfb and increasing the expression of C1q and C4Bp, thereby affecting neurite outgrowth and synapse formation, resulting in disordered neuron function. Studying these gene regulations might be beneficial to understanding neuronal development and brain injury prevention.


Subject(s)
Astrocytes/metabolism , Neurogenesis , Neuronal Outgrowth , Sp1 Transcription Factor/metabolism , Synapses/metabolism , Animals , Astrocytes/cytology , Behavior, Animal , Cell Shape , Cells, Cultured , Cerebral Cortex/pathology , Gene Expression Regulation , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/pathology , Mice, Knockout , Neurites/metabolism , Sp1 Transcription Factor/deficiency
2.
Cell Death Dis ; 8(12): 3200, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29233979

ABSTRACT

Renal ischemia-reperfusion is a main cause of acute kidney injury (AKI), which is associated with high mortality. Here we show that extracellular vesicles (EVs) secreted from hiPSC-MSCs play a critical role in protection against renal I/R injury. hiPSC-MSCs-EVs can fuse with renal cells and deliver SP1 into target cells, subsequently active SK1 expression and increase S1P formation. Chromatin immunoprecipitation (ChIP) analyses and luciferase assay were used to confirm SP1 binds directly to the SK1 promoter region and promote promoter activity. Moreover, SP1 inhibition (MIT) or SK1 inhibition (SKI-II) completely abolished the renal protective effect of hiPSC-MSCs-EVs in rat I/R injury mode. However, pre-treatment of necroptosis inhibitor Nec-1 showed no difference with the administration of hiPSC-MSCs-EVs only. We then generated an SP1 knockout hiPSC-MSC cell line by CRISPR/Cas9 system and found that SP1 knockout failed to show the protective effect of hiPSC-MSCs-EVs unless restoring the level of SP1 by Ad-SP1 in vitro and in vivo. In conclusion, this study describes an anti-necroptosis effect of hiPSC-MSCs-EVs against renal I/R injury via delivering SP1 into target renal cells and intracellular activating the expression of SK1 and the generation of S1P. These findings suggest a novel mechanism for renal protection against I/R injury, and indicate a potential therapeutic approach for a variety of renal diseases and renal transplantation.


Subject(s)
Acute Kidney Injury/prevention & control , Extracellular Vesicles/chemistry , Mesenchymal Stem Cells/metabolism , Necrosis/prevention & control , Phosphotransferases (Alcohol Group Acceptor)/genetics , Reperfusion Injury/prevention & control , Sp1 Transcription Factor/genetics , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Apoptosis/genetics , Cell Differentiation , Cell Line, Transformed , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kidney/metabolism , Kidney/pathology , Lysophospholipids/metabolism , Male , Mesenchymal Stem Cells/cytology , Necrosis/genetics , Necrosis/metabolism , Necrosis/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction , Sp1 Transcription Factor/deficiency , Sphingosine/analogs & derivatives , Sphingosine/metabolism
3.
J Biol Chem ; 287(14): 10799-811, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22337869

ABSTRACT

The transcription factor Krüppel-like factor 4 (KLF4) plays a critical role in vascular smooth muscle cell (VSMC) differentiation induced by all-trans-retinoic acid (ATRA). Although it has been demonstrated that ATRA stimulation augments both KLF4 protein and mRNA levels in VSMCs, the molecular mechanisms by which ATRA regulates Klf4 transcription are unknown. In this study, we examined the roles of ATRA-selective nuclear retinoic acid receptors (RARs) in the transcriptional regulation of Klf4. The introduction of small interfering RNA and an RAR antagonist demonstrated that RARα, but not RARß or RARγ, mediated ATRA-induced Klf4 expression. A luciferase assay for the Klf4 promoter showed that three GC boxes in the proximal Klf4 promoter were indispensible for ATRA-induced Klf4 transcription and that RARα enhanced Klf4 promoter activity in a GC box-dependent manner. Furthermore, chromatin immunoprecipitation and oligonucleotide pulldown assays demonstrated that the transcription factors KLF4, Sp1, and YB1 directly bound to the GC boxes of the proximal Klf4 promoter. Upon RARα agonist stimulation, RARα was recruited to the Klf4 promoter through its interaction with KLF4, Sp1, and YB1 to form a transcriptional activation complex on the three GC boxes of the Klf4 promoter. These results suggest that RARα serves as an essential co-activator for ATRA signaling and that the recruitment of RARα to the KLF4-Sp1-YB1 complex, which leads to Klf4 expression in VSMCs, is independent of a retinoic acid response element.


Subject(s)
Kruppel-Like Transcription Factors/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Promoter Regions, Genetic/drug effects , Receptors, Retinoic Acid/metabolism , Transcriptional Activation/drug effects , Tretinoin/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Gene Knockdown Techniques , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/deficiency , Male , Rats , Response Elements/drug effects , Retinoic Acid Receptor alpha , Signal Transduction/drug effects , Sp1 Transcription Factor/deficiency , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Tretinoin/metabolism , Y-Box-Binding Protein 1/deficiency , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism
4.
J Invest Dermatol ; 131(11): 2213-22, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21753780

ABSTRACT

Transcription factor specificity protein 1 (Sp1) is involved in diverse cellular functions. We recently found that Sp1 was significantly decreased in skin biopsy samples obtained from patients with atopic dermatitis (AD) and had an even greater reduction in AD patients with a history of eczema herpeticum. In the current study, we sought to better understand the role of Sp1 in skin biological processes by using a small-interfering RNA (siRNA) technique to knock down Sp1 gene expression in normal human keratinocytes (NHKs) and investigated the genome-wide gene expression profiling of Sp1-silenced NHKs. The gene arrays revealed that 53 genes had greater than 3-fold changes in the expression in Sp1-silenced NHKs as compared with scrambled siRNA-silenced cells. Strikingly, six kallikrein (KLK)-related peptidase genes, namely KLK5, KLK6, KLK7, KLK8, KLK10, and KLK12, were upregulated in NHKs following Sp1 silencing. Functionally, protease activity was significantly enhanced in Sp1-silenced keratinocytes as compared with scrambled siRNA-silenced keratinocytes. Moreover, thymic stromal lymphopoietin (TSLP), an epithelial-derived T(H)2-promoting cytokine, was induced in Sp1-silenced keratinocytes because of elevated KLK activity. These results indicate that Sp1 expression deficiency leads to abnormally increased KLK protease activity in keratinocytes and may contribute to T(H)2 immune responses in the skin by inducing TSLP.


Subject(s)
Cytokines/metabolism , Gene Expression Profiling , Kallikreins/metabolism , Keratinocytes/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Up-Regulation/physiology , Cells, Cultured , Gene Silencing/physiology , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Peptide Hydrolases/metabolism , RNA, Small Interfering/pharmacology , Sp1 Transcription Factor/deficiency , Sp1 Transcription Factor/drug effects , Thymic Stromal Lymphopoietin
5.
FEBS Lett ; 585(7): 1103-11, 2011 Apr 06.
Article in English | MEDLINE | ID: mdl-21402072

ABSTRACT

Pharmacological manipulations to purge human immunodeficiency virus (HIV) from latent reservoirs have been considered as an adjuvant therapeutic approach to highly-active antiretroviral therapy for the eradication of HIV. Our novel histone deacetylase inhibitor NCH-51 induced expression of latent HIV-1 with minimal cytotoxicity. Using chromatin immunoprecipitation assays, we observed a reduction of HDAC1 occupancy, histone hyperacetylation and the recruitment of positive transcription factors at the HIV-1 promoter in latently infected-cells under the treatment with NCH-51. Mutation studies of the long terminal repeat (LTR) revealed NCH-51 mediated gene expression through the Sp1 sites. When Sp1 expression was knocked-down by small interfering RNA, the NCH-51-mediated activation of a stably integrated HIV-1 LTR was attenuated. Moreover, the Sp1 inhibitor mithramycin A abolished the effects of NCH-51.


Subject(s)
Gene Expression Regulation, Viral/drug effects , HIV-1/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Sulfhydryl Compounds/pharmacology , Virus Latency/drug effects , Virus Latency/genetics , Acetylation/drug effects , Chromatin Assembly and Disassembly/drug effects , Gene Knockdown Techniques , HIV-1/genetics , HIV-1/physiology , HL-60 Cells , Histones/metabolism , Humans , Nucleosomes/drug effects , Nucleosomes/metabolism , Promoter Regions, Genetic/genetics , Sp1 Transcription Factor/deficiency , Sp1 Transcription Factor/genetics , Terminal Repeat Sequences/genetics , Transcriptional Activation/drug effects , Virus Replication/drug effects
6.
Genes Chromosomes Cancer ; 49(3): 282-97, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20013896

ABSTRACT

Specificity protein 1 (SP1) is an essential transcription factor implicated in the regulation of genes that control multiple cellular processes, including cell cycle, apoptosis, and DNA damage. Very few nontranscriptional roles for SP1 have been reported thus far. Using confocal microscopy and centrosome fractionation, we identified SP1 as a centrosomal protein. Sp1-deficient mouse embryonic fibroblasts and cells depleted of SP1 by RNAi have increased centrosome number associated with centriole splitting, decreased microtubule nucleation, chromosome misalignment, formation of multipolar mitotic spindles and micronuclei, and increased incidence of aneuploidy. Using mass spectrometry, we identified P70S6K, an effector of the mTOR/raptor (mTORC1) kinase complex, as a novel interacting protein of SP1. We found that SP1-deficient cells have increased phosphorylation of the P70S6K effector ribosomal protein S6, suggesting that SP1 participates in the regulation of the mTORC1/P70S6K/S6 signaling pathway. We previously reported that aberrant mTORC1 activation leads to supernumerary centrosomes, a phenotype rescued by the mTORC1 inhibitor rapamycin. Similarly, treatment with rapamycin rescued the multiple centrosome phenotype of SP1-deficient cells. Taken together, these data strongly support the hypothesis that SP1 is involved in the control of centrosome number via regulation of the mTORC1 pathway, and predict that loss of SP1 function can lead to aberrant centriole splitting, deregulated mTORC1 signaling, and aneuploidy, thereby contributing to malignant transformation.


Subject(s)
Centrioles/physiology , Chromosomal Instability , Gene Silencing , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Sp1 Transcription Factor/metabolism , 3T3 Cells , Animals , Apoptosis , Cell Cycle , Cell Line , Centrosome/physiology , Centrosome/ultrastructure , DNA Damage , Gene Expression Regulation , HeLa Cells , Humans , Mice , RNA Interference/physiology , Sp1 Transcription Factor/deficiency , Sp1 Transcription Factor/genetics , TOR Serine-Threonine Kinases
7.
Dev Dyn ; 236(8): 2235-44, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17584888

ABSTRACT

The ubiquitously expressed zinc finger transcription factors Sp1 and Sp3 play critical roles in embryonic development. Sp1 knockout mice die around embryonic day 10.5. Mice lacking Sp3 are postnatal lethal. Mice heterozygous for either Sp1 or Sp3 are apparently normal, although slightly smaller. Here, we show that compound heterozygosity of Sp1 and Sp3 results in embryonic lethality accompanied by a spectrum of developmental abnormalities, including growth retardation, morphological alterations of the lung, impaired ossification, anemia, and placental defects. Anemia in Sp1/Sp3 compound heterozygous mutant embryos is associated with impaired maturation of erythrocytes. Analyses of the placenta revealed a markedly reduced spongiotrophoblast layer and a severe disorganization of the labyrinth layer in Sp1/Sp3 compound heterozygous as well as in Sp3-deficient mutant embryos. Our findings demonstrate that a threshold of Sp1 and Sp3 activity is required for normal embryonic development, suggesting that Sp1 and Sp3 act cooperatively to regulate downstream targets.


Subject(s)
Erythropoiesis/genetics , Placenta/abnormalities , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/genetics , Animals , Heterozygote , Mice , Phenotype , Placenta/pathology , Sp1 Transcription Factor/deficiency , Sp3 Transcription Factor/deficiency , Survival Rate
8.
Mol Cell Biol ; 24(2): 865-74, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14701757

ABSTRACT

Proteolytic processing of the beta-amyloid precursor protein (APP) at the beta site is essential to generate Abeta. BACE1, the major beta-secretase involved in cleaving APP, has been identified as a type 1 membrane-associated aspartyl protease. We have cloned a 2.1-kb fragment upstream of the human BACE1 gene and identified key regions necessary for promoter activity. BACE1 gene expression is controlled by a TATA-less promoter. The region of bp -619 to +46 is the minimal promoter to control the transcription of the BACE1 gene. Several putative cis-acting elements, such as a GC box, HSF-1, a PU box, AP1, AP2, and lymphokine response element, are found in the 5' flanking region of the BACE1 gene. Transcriptional activation and gel shift assays demonstrated that the BACE1 promoter contains a functional Sp1 response element, and overexpression of the transcription factor Sp1 potentiates BACE gene expression and APP processing to generate Abeta. Furthermore, Sp1 knockout reduced BACE1 expression. These results suggest that BACE1 gene expression is tightly regulated at the transcriptional level and that the transcription factor Sp1 plays an important role in regulation of BACE1 to process APP generating Abeta in Alzheimer's disease.


Subject(s)
Aspartic Acid Endopeptidases/genetics , Plicamycin/analogs & derivatives , Sp1 Transcription Factor/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/metabolism , Animals , Base Sequence , Cell Line , Chromosome Mapping , Cloning, Molecular , DNA/genetics , Endopeptidases , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Plicamycin/pharmacology , Promoter Regions, Genetic/drug effects , Protein Processing, Post-Translational , Rats , Sequence Deletion , Sp1 Transcription Factor/deficiency , Sp1 Transcription Factor/genetics , Transcription, Genetic , Up-Regulation
9.
Endocrinology ; 142(4): 1635-42, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11250945

ABSTRACT

Members of the family of Sp transcription factors include Sp1, Sp3, and Sp4 and are important regulators of eukaryotic gene expression. We previously reported that Sp1 mediated stimulation of rat calmodulin I gene expression in response to insulin. To test whether other members of the Sp family are direct targets of insulin action, we compared the levels of Sp1 and Sp3 proteins from nuclear extracts obtained from both insulin-treated and untreated rat hepatoma (H-411E) cells. We demonstrated by Western blot analysis that levels of Sp1 and Sp3 proteins were increased more than 2-fold in the insulin-treated group. Additionally, the up-regulation of both Sp1 and Sp3 transcription factors by insulin was antagonized by tumor necrosis factor-alpha, a known inhibitor of insulin action. Immunohistochemical analysis demonstrated that H-411E cells treated with insulin (10,000 microU/ml) had a marked increase in demonstrable Sp1 in the nucleus compared with cells incubated in insulin-free medium. We extended these in vitro observations to in vivo studies in the streptozotocin-diabetic rat model. We demonstrated in rat liver tissue by both Western blot and immunohistochemical staining with anti-Sp1 antibody that 1) livers of fully diabetic streptozotocin rats have low levels of Sp1 transcription factor; and 2) insulin treatment of the diabetic rat rapidly reversed this process by markedly stimulating accumulation of Sp1 in rat liver. Studies of the signal transduction mechanisms involved in insulin's effect on Sp1 demonstrate a facilitating role for phosphoinositol 3-kinase and an inhibitory role for cyclic nucleotides. In summary, insulin stimulates Sp1 protein, a transcription factor that is shown to regulate calmodulin gene expression and most likely other, as yet untested, genes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Ketoacidosis/metabolism , Insulin/physiology , Liver Neoplasms, Experimental/metabolism , Sp1 Transcription Factor/deficiency , Animals , Blotting, Western , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Sp1 Transcription Factor/genetics , Tumor Cells, Cultured
10.
Oncogene ; 19(40): 4660-7, 2000 Sep 21.
Article in English | MEDLINE | ID: mdl-11030155

ABSTRACT

In this report, we describe the mechanism of TGF-beta receptor type I (RI) repression in the GEO human colon carcinoma cells. Treatment of GEO cells with the DNA methyltransferase inhibitor, 5 azacytidine induced RI expression and restored TGF-beta response. A stably transfected RI promoter-reporter construct (RI-Luc) expressed higher activity in the 5 aza C treated GEO cells, suggesting the activation of a transactivator for RI transcription. Gel shift analysis indicated enhanced binding of proteins from the 5 aza C treated nuclear extracts to radiolabeled Sp1 oligonucleotides specifically contained in the RI promoter. Protein stability studies after cyclohexamide treatment suggested an increase in the Sp1 protein stability from the 5 aza C treated GEO cells. Further, transfection of Sp1 cDNA into untreated GEO control cells increased RI promoter activity and thus induced RI expression. 5 aza C mediated Sp1 expression in Sp1 deficient GEO colon and MCF-7 breast cancer cells also enhanced the activity of several other Sp1 dependent promoters such as TGF-beta receptor type II (RII), Cyclin A and p21/waf1/cip1. These results indicate that restoration of Sp1 in several different types of Sp1 deficient cells leads to enhanced activation of a wide range of Sp1 dependent promoters.


Subject(s)
Activin Receptors, Type I , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Sp1 Transcription Factor/deficiency , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclin A/biosynthesis , Cyclin A/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA Methylation/drug effects , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/deficiency , Protein Serine-Threonine Kinases/biosynthesis , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/biosynthesis , Recombinant Fusion Proteins/physiology , Sp1 Transcription Factor/physiology , Transcriptional Activation , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
11.
EMBO J ; 19(4): 655-61, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10675334

ABSTRACT

Sp3 is a ubiquitously expressed transcription factor closely related to Sp1 (specificity protein 1). We have disrupted the mouse Sp3 gene by homologous recombination. Sp3-deficient embryos are growth retarded and invariably die at birth of respiratory failure. The cause for the observed breathing defect remains obscure since only minor morphological alterations were observed in the lung, and surfactant protein expression is indistinguishable from that in wild-type mice. Histological examinations of individual organs in Sp3(-/-) mice show a pronounced defect in late tooth formation. In Sp3 null mice, the dentin/enamel layer of the developing teeth is impaired due to the lack of ameloblast-specific gene products. Comparison of the Sp1 and Sp3 knockout phenotype shows that Sp1 and Sp3 have distinct functions in vivo, but also suggests a degree of functional redundancy.


Subject(s)
DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Odontogenesis/genetics , Odontogenesis/physiology , Transcription Factors/deficiency , Transcription Factors/genetics , Ameloblasts/metabolism , Animals , Animals, Newborn , Base Sequence , DNA Primers/genetics , DNA-Binding Proteins/physiology , Female , Gene Expression , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pregnancy , Respiratory Insufficiency/genetics , Respiratory Insufficiency/metabolism , Respiratory Insufficiency/pathology , Sp1 Transcription Factor/deficiency , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/physiology , Sp3 Transcription Factor , Tooth/growth & development , Tooth/metabolism , Tooth/pathology , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...