Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biomed Res ; 41(3): 119-129, 2020.
Article in English | MEDLINE | ID: mdl-32522929

ABSTRACT

Sodium trisulfide (Na2S3) releases hydrogen polysulfide (H2Sn) and is useful for the investigation of the effects of H2Sn on the cell functions. In the present study, we first examined the effects of Na2S3 on the gene expression of IEC-6 cells, a rat intestinal epithelial cell line. Microarray analysis and reverse transcription-polymerase chain reaction analysis revealed that Na2S3 increased the gene expression of early growth response 1 (EGR1) and Kruppel-like transcription factor 4 (KLF4). It was interesting that U0126, an inhibitor of the activation of extracellular signal-regulated kinase 1 (ERK1), ERK2, and ERK5, inhibited the Na2S3-induced gene expression of EGR1 and KLF4. Na2S3 activated ERK1 and ERK2 (ERK1/2) within 15 min. In addition to ERK1/2, Na2S3 activated ERK5. We noticed that the electrophoretic mobility of ERK5 was decreased after Na2S3 treatment. Phos-tag analysis and in vitro dephosphorylation of the cell extracts indicated that the gel-shift of ERK5 was due to its phosphorylation. The gel-shift of ERK5 was inhibited completely by both U0126 and ERK5-IN-1, a specific inhibitor of ERK5. From these results, we concluded that the gel-shift of ERK5 was induced through autophosphorylation by activated ERK5 after Na2S3 treatment. The present study suggested that H2Sn affected various functions of intestinal epithelial cells through the activation of the ERK1/2 and ERK5 pathways.


Subject(s)
Early Growth Response Protein 1/genetics , Epithelial Cells/drug effects , Hydrogen Sulfide/pharmacology , Kruppel-Like Transcription Factors/genetics , Signal Transduction/drug effects , Animals , Butadienes/pharmacology , Cell Line , Early Growth Response Protein 1/agonists , Early Growth Response Protein 1/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/agonists , Kruppel-Like Transcription Factors/metabolism , Microarray Analysis , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Nitriles/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Rats , Signal Transduction/genetics
2.
Biochem Pharmacol ; 164: 115-128, 2019 06.
Article in English | MEDLINE | ID: mdl-30954486

ABSTRACT

Uric acid (UA) is a promising protective treatment in ischaemic stroke, but the precise molecular targets underlying its in vivo beneficial actions remain unclear. High concentrations of UA inhibit angiogenesis of cultured endothelial cells via Krüppel-like factor 2 (KLF)-induced downregulation of vascular endothelial growth factor (VEGF), a pro-angiogenic mediator that is able to increase blood-brain barrier (BBB) permeability in acute stroke. Here, we investigated whether UA treatment after ischaemic stroke protects brain endothelial cell functions and modulates the KLF2-VEGF-A axis. Transient intraluminal middle cerebral artery (MCA) occlusion/reperfusion was induced in adult male spontaneously hypertensive (SHR) rats and corresponding normotensive Wistar-Kyoto (WKY) rats. Animals received UA (16 mg/kg) or vehicle (Locke's buffer) i.v. at reperfusion. BBB permeability was evaluated by Evans blue extravasation to the brain and in human cerebral endothelial hCMEC/D3 cells under oxygen-glucose deprivation/re-oxygenation. Circulating VEGF-A levels were measured in rats and acute ischaemic stroke patients from the URICO-ICTUS trial. Angiogenesis progression was assessed in Matrigel-cultured MCA. Worse post-stroke brain damage in SHR than WKY rats was associated with higher hyperaemia at reperfusion, increased Evans blue extravasation, exacerbated MCA angiogenic sprouting, and higher VEGF-A levels. UA treatment reduced infarct volume and Evans blue leakage in both rat strains, improved endothelial cell barrier integrity and KLF2 expression, and lowered VEGF-A levels in SHR rats. Hypertensive stroke patients treated with UA showed lower levels of VEGF-A than patients receiving vehicle. Consistently, UA prevented the enhanced MCA angiogenesis in SHR rats by a mechanism involving KLF2 activation. We conclude that UA treatment after ischaemic stroke upregulates KLF2, reduces VEGF-A signalling, and attenuates brain endothelial cell dysfunctions leading to neuroprotection.


Subject(s)
Blood-Brain Barrier/metabolism , Hypertension/blood , Kruppel-Like Transcription Factors/blood , Stroke/blood , Uric Acid/therapeutic use , Vascular Endothelial Growth Factor A/blood , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Biomarkers/blood , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/metabolism , Cell Line , Double-Blind Method , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Hypertension/drug therapy , Hypertension/pathology , Kruppel-Like Transcription Factors/agonists , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Stroke/drug therapy , Stroke/pathology , Treatment Outcome , Uric Acid/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors
3.
Mol Med Rep ; 19(2): 967-973, 2019 02.
Article in English | MEDLINE | ID: mdl-30569122

ABSTRACT

Uncontrolled proliferation and defective apoptosis are two major factors responsible for maintaining the malignant properties of melanoma cells. Our previous study demonstrated that induced expression of four reprogramming factors remodeled the phenotype of B16­F10 mouse melanoma cells into melanoma stem cells. The present study was conducted to investigate the effect of the four Yamanaka reprogramming factors, namely Oct4, Sox2, Klf4 and c­Myc (OSKM), on the proliferation and apoptosis of melanoma cells, and to identify the responsible molecular signals. The results identified that expression of the four reprogramming factors was highly induced by doxycycline treatment in the stable melanoma cell clone that was transfected with a plasmid expressing these factors, driven by the Tet­On element. It was further confirmed that induced expression of these factors enhanced the proliferation and suppressed the apoptosis of the melanoma cells. In addition, induced OSKM expression increased cell proliferation, accelerated the progression of the cell cycle, and upregulated the mRNA expression levels of Janus kinase 2 (JAK2) and Cyclin­B1. Induced expression of these factors also decreased the apoptosis, as well as upregulated B­cell lymphoma 2 (BCL­2) and downregulated BCL­2­associated X (BAX) mRNA expression levels. Taken together, the results suggested that upregulated JAK2 and Cyclin­B1 may be responsible for the enhanced proliferation of melanoma cells, and that BCL­2 upregulation and BAX downregulation may account for the suppressed apoptosis of these cells.


Subject(s)
Cellular Reprogramming , Doxycycline/pharmacology , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/genetics , Octamer Transcription Factor-3/genetics , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin B1/genetics , Cyclin B1/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/agonists , Kruppel-Like Transcription Factors/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/agonists , Octamer Transcription Factor-3/metabolism , Plasmids/chemistry , Plasmids/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/agonists , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/agonists , SOXB1 Transcription Factors/metabolism , Transfection , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
4.
Free Radic Biol Med ; 129: 394-406, 2018 12.
Article in English | MEDLINE | ID: mdl-30315936

ABSTRACT

Nuclear factor-E2-related factor 2 (NRF2) is a transcription factor that activates the antioxidant cellular defense in response to oxidative stress, leading to neuroprotective effects in Parkinson's disease (PD) models. We have previously shown that Angiotensin II (AngII) induces an increase in reactive oxygen species (ROS) via AngII receptor type 1 and NADPH oxidase (NOX), which may activate the NRF2 pathway. However, controversial data suggest that AngII induces a decrease in NRF2 signaling leading to an increase in oxidative stress. We analyzed the effect of AngII and the dopaminergic neurotoxin 6-hydroxydopamine (6-OHDA) in culture and in vivo, and examined the effects on the expression of NRF2-related genes. Treatment of neuronal cell lines Mes23.5, N27 and SH-SY5Y with AngII, 6-OHDA or a combination of both increased ROS production and reduced cell viability. Simultaneously, these treatments induced an increase in expression in the NRF2-regulated genes heme oxygenase 1 (Hmox1), NAD(P)H quinone dehydrogenase 1 (Nqo1) and Kruppel like factor 9 (Klf9). Moreover, overexpression of KLF9 transcription factor caused a reduction in the production of ROS induced by treatment with AngII or 6-OHDA and improved the survival of these neuronal cells. Rats treated with AngII, 6-OHDA or a combination of both also showed an increased expression of NRF2 related genes and KLF9. In conclusion, our data indicate that AngII induces a damaging effect in neuronal cells, but also acts as a signaling molecule to activate NRF2 and KLF9 neuroprotective pathways in cellular and animal models of PD.


Subject(s)
Angiotensin II/pharmacology , Dopaminergic Neurons/drug effects , Kruppel-Like Transcription Factors/genetics , NF-E2-Related Factor 2/genetics , Oxidopamine/pharmacology , Animals , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Gene Expression Regulation , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Humans , Injections, Intraventricular , Kruppel-Like Transcription Factors/agonists , Kruppel-Like Transcription Factors/metabolism , Male , Mice , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Signal Transduction , Stereotaxic Techniques
5.
J Am Heart Assoc ; 6(12)2017 Nov 30.
Article in English | MEDLINE | ID: mdl-29191808

ABSTRACT

BACKGROUND: Kruppel-like factor 2 (KLF2) is an important zinc-finger transcription factor that maintains endothelial homeostasis by its anti-inflammatory, -thrombotic, -oxidative, and -proliferative effects in endothelial cells. In light of the potent vasoprotective effects of KLF2, modulating KLF2 expression or function could give rise to new therapeutic strategies to treat cardiovascular diseases. METHODS AND RESULTS: High-throughput drug screening based on KLF2 promoter luciferase reporter assay was performed to screen KLF2 activators. Real-time PCR and western blot were used to detect gene and protein expression. Identified KLF2 activator was orally administered to ApoE-/- mice to evaluate anti-atherosclerotic efficacy. By screening 2400 compounds in the Spectrum library, we identified suberanilohydroxamic (SAHA) acid, also known as vorinostat as a pharmacological KLF2 activator through myocyte enhancer factor 2. We found that SAHA exhibited anti-inflammatory effects and attenuated monocyte adhesion to endothelial cells inflamed with tumor necrosis factor alpha. We further showed that the inhibitory effect of SAHA on endothelial inflammation and ensuing monocyte adhesion was KLF2 dependent using KLF2-deficient mouse lung endothelial cells or KLF2 small interfering RNA- depleted human endothelial cells. Importantly, we observed that oral administration of SAHA reduced diet-induced atherosclerotic lesion development in ApoE-/- mice without significant effect on serum lipid levels. CONCLUSIONS: These results demonstrate that SAHA has KLF2-dependent anti-inflammatory effects in endothelial cells and provide the proof of concept that KLF2 activation could be a promising therapeutic strategy for treating atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Atherosclerosis/prevention & control , Hydroxamic Acids/administration & dosage , Kruppel-Like Transcription Factors/agonists , Vasculitis/prevention & control , Administration, Oral , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , COS Cells , Cell Adhesion/drug effects , Chlorocebus aethiops , Coculture Techniques , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , High-Throughput Screening Assays , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice, Knockout, ApoE , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Promoter Regions, Genetic/drug effects , RNA Interference , Signal Transduction/drug effects , THP-1 Cells , Transfection , Vasculitis/genetics , Vasculitis/metabolism , Vasculitis/pathology , Vorinostat
6.
J Biol Chem ; 292(37): 15192-15204, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28667171

ABSTRACT

Hedgehog (HH) signaling critically regulates embryonic and postnatal development as well as adult tissue homeostasis, and its perturbation can lead to developmental disorders, birth defects, and cancers. Neuropilins (NRPs), which have well-defined roles in Semaphorin and VEGF signaling, positively regulate HH pathway function, although their mechanism of action in HH signaling remains unclear. Here, using luciferase-based reporter assays, we provide evidence that NRP1 regulates HH signaling specifically at the level of GLI transcriptional activator function. Moreover, we show that NRP1 localization to the primary cilium, a key platform for HH signal transduction, does not correlate with HH signal promotion. Rather, a structure-function analysis suggests that the NRP1 cytoplasmic and transmembrane domains are necessary and sufficient to regulate HH pathway activity. Furthermore, we identify a previously uncharacterized, 12-amino acid region within the NRP1 cytoplasmic domain that mediates HH signal promotion. Overall, our results provide mechanistic insight into NRP1 function within and potentially beyond the HH signaling pathway. These insights have implications for the development of novel modulators of HH-driven developmental disorders and diseases.


Subject(s)
Hedgehog Proteins/metabolism , Kruppel-Like Transcription Factors/agonists , Models, Biological , Neuropilin-1/metabolism , Nuclear Proteins/agonists , Signal Transduction , Amino Acid Motifs , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Embryo, Mammalian/cytology , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Mutant Strains , Mutation , NIH 3T3 Cells , Neuropilin-1/chemistry , Neuropilin-1/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Transport , Recombinant Fusion Proteins/metabolism , Zinc Finger Protein Gli2
7.
Biochem Biophys Res Commun ; 484(3): 486-492, 2017 03 11.
Article in English | MEDLINE | ID: mdl-28108288

ABSTRACT

KLF4 is a transcriptional factor that can function either as a tumor suppressor or oncogene in cancer based on its cellular context. We recently demonstrated that KLF4 was a tumor suppressor in ovarian cancer cells by inhibiting the epithelial to mesenchymal transition. Here we report that KLF4 expression was downregulated in ovarian cancer tissue compared to normal ovarian tissue, and low KLF4 expression correlated with high risk ovarian carcinoma and poor patient survival. Enforced KLF4 expression by lentiviral transduction sensitized ovarian cancer cells to the effects of the chemotherapy drugs, paclitaxel and cisplatin. Treatment of ovarian cancer cells with APTO-253, a small molecule inducer of KLF4, enhanced the efficacy of both chemotherapy drugs. KLF4 expression mediated by lentiviral vector or induced by APTO-253 resulted in G1 phase arrest in ovarian cancer cells. Our results demonstrate that for the first time that inducing KLF4 expression with APTO-253 is a novel therapeutic strategy for treating ovarian cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/metabolism , Imidazoles/administration & dosage , Kruppel-Like Transcription Factors/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Phenanthrolines/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/agonists , Ovarian Neoplasms/pathology , Up-Regulation/drug effects
8.
Stem Cells ; 34(11): 2721-2732, 2016 11.
Article in English | MEDLINE | ID: mdl-27300003

ABSTRACT

Prenatal folic acid (FA) supplementation prevents neural tube defects. Folate receptor alpha (FRα) is critical for embryonic development, including neural crest (NC) development. Previously we showed that FRα translocates to the nucleus in response to FA, where it acts as a transcription factor. In this study, we examined if FA through interaction with FRα regulates stem cell characteristics of cranial neural crest cells (CNCCs)-critical for normal development. We hypothesized that FRα upregulates coding genes and simultaneously downregulates non-coding miRNA which targets coding genes in CNCCs. Quantitative RT-PCR and chromatin immunoprecipitation showed that FRα upregulates Oct4, Sox2, and Klf4 by binding to their cis-regulator elements-5' enhancer/promoters defined by H3K27Ac and p300 occupancy. FA via FRα downregulates miRNAs, miR-138 and miR-let-7, which target Oct4 and Trim71 (an Oct4 downstream effector), respectively. Co-immunoprecipitation data suggests that FRα interacts with the Drosha-DGCR8 complex to affect pre-miRNA processing. Transfecting anti-miR-138 or anti-miR-let-7 into non-proliferating neural crest cells (NCCs) derived from Splotch (Sp-/- ), restored their proliferation potential. In summary, these results suggest a novel pleiotropic role of FRα: (a) direct activation of Oct4, Sox2, and Klf4 genes; and (b) repression of biogenesis of miRNAs that target these genes or their effector molecules. Stem Cells 2016;34:2721-2732.


Subject(s)
Folate Receptor 1/genetics , Kruppel-Like Transcription Factors/genetics , MicroRNAs/genetics , Neural Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , SOXB1 Transcription Factors/genetics , Animals , Antagomirs/genetics , Antagomirs/metabolism , Female , Folate Receptor 1/antagonists & inhibitors , Folate Receptor 1/metabolism , Folic Acid/metabolism , Folic Acid/pharmacology , Gene Expression Regulation, Developmental , Histones/genetics , Histones/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/agonists , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neural Crest/cytology , Neural Crest/drug effects , Neural Crest/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Octamer Transcription Factor-3/agonists , Octamer Transcription Factor-3/metabolism , PAX3 Transcription Factor/deficiency , PAX3 Transcription Factor/genetics , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , SOXB1 Transcription Factors/agonists , SOXB1 Transcription Factors/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
9.
PLoS One ; 11(3): e0150936, 2016.
Article in English | MEDLINE | ID: mdl-26938105

ABSTRACT

Understanding the mechanisms that regulate pluripotency of embryonic stem cells (ESCs) is important to ensure their safe clinical use. CHIR99021 (CHIR)-induced activation of Wnt/ß-catenin signaling promotes self-renewal in mouse ESCs (mESCs). ß-catenin functions individually or cooperates with transcription factors to activate stemness factors such as c-Myc, Esrrb, Pou5f1, and Nanog. However the relationship between the core pluripotent factor, Kruppel-like factor 4 (also known as GKLF or EZF) and Wnt/ß-catenin signaling, remains ambiguous in J1 mESCs. DNA microarray analysis revealed that CHIR-treatment promoted pluripotency-maintaining transcription factors and repressed germ layer specification markers. CHIR also promoted genes related to the development of extracellular regions and the plasma membrane to maintain pluripotency of J1 mESCs. Among the CHIR-regulated genes, Klf4 has not been reported previously. We identified a novel cis element in the Klf4 gene that was activated by ß-catenin in J1 mESCs. We determined that ß-catenin interacted with this cis element, identifying Klf4 as a ß-catenin target gene in this context. Moreover, several microRNAs that targeted the 3'-UTR of Klf4 mRNA were identified, with miR-7a being down-regulated by CHIR in a ß-catenin-independent manner in J1 mESCs. These data collectively suggest that CHIR enhances Klf4 expression by repressing miR-7a expression or canonical Wnt pathway activation.


Subject(s)
Kruppel-Like Transcription Factors/genetics , MicroRNAs/genetics , Mouse Embryonic Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , beta Catenin/genetics , 3' Untranslated Regions , Animals , Base Sequence , Cell Line , Gene Expression Regulation , Gene Ontology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/agonists , Kruppel-Like Transcription Factors/metabolism , Mice , MicroRNAs/metabolism , Molecular Sequence Annotation , Molecular Sequence Data , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
10.
J Clin Invest ; 125(10): 3819-30, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26368306

ABSTRACT

Recent genome-wide association studies have revealed that variations near the gene locus encoding the transcription factor Krüppel-like factor 14 (KLF14) are strongly associated with HDL cholesterol (HDL-C) levels, metabolic syndrome, and coronary heart disease. However, the precise mechanisms by which KLF14 regulates lipid metabolism and affects atherosclerosis remain largely unexplored. Here, we report that KLF14 is dysregulated in the liver of 2 dyslipidemia mouse models. We evaluated the effects of both KLF14 overexpression and genetic inactivation and determined that KLF14 regulates plasma HDL-C levels and cholesterol efflux capacity by modulating hepatic ApoA-I production. Hepatic-specific Klf14 deletion in mice resulted in decreased circulating HDL-C levels. In an attempt to pharmacologically target KLF14 as an experimental therapeutic approach, we identified perhexiline, an approved therapeutic small molecule presently in clinical use to treat angina and heart failure, as a KLF14 activator. Indeed, in WT mice, treatment with perhexiline increased HDL-C levels and cholesterol efflux capacity via KLF14-mediated upregulation of ApoA-I expression. Moreover, perhexiline administration reduced atherosclerotic lesion development in apolipoprotein E-deficient mice. Together, these results provide comprehensive insight into the KLF14-dependent regulation of HDL-C and subsequent atherosclerosis and indicate that interventions that target the KLF14 pathway should be further explored for the treatment of atherosclerosis.


Subject(s)
Apolipoprotein A-I/biosynthesis , Atherosclerosis/prevention & control , Cholesterol, HDL/blood , Cholesterol/metabolism , Hyperlipoproteinemia Type II/drug therapy , Kruppel-Like Transcription Factors/physiology , Liver/metabolism , Perhexiline/pharmacology , Animals , Apolipoprotein A-I/genetics , Apolipoproteins E/deficiency , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/therapy , Diet, Atherogenic , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Genetic Therapy , Genetic Vectors/therapeutic use , Genome-Wide Association Study , Hep G2 Cells , Humans , Hyperlipoproteinemia Type II/metabolism , Kruppel-Like Transcription Factors/agonists , Leptin/deficiency , Liver/drug effects , Mice , Mice, Inbred C57BL , Mice, Obese , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Fusion Proteins/metabolism , Sp Transcription Factors/genetics , Sp Transcription Factors/metabolism , Sterol Regulatory Element Binding Proteins/biosynthesis , Sterol Regulatory Element Binding Proteins/genetics
11.
J Nutr Biochem ; 26(3): 277-84, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25542418

ABSTRACT

Physiological concentrations (1 µM) of 15 flavonoids were evaluated in human umbilical vein endothelial cells in the presence of hydrogen peroxide (H2O2) for their ability to affect endothelial nitric oxide synthase (eNOS) and endothelin-1 (ET-1) expression in order to establish the structural basis of their bioactivity. Flavonoid effects on eNOS transcription factor Krüpple like factor-2 (KLF-2) expression were also evaluated. All studied flavonoids appeared to be effective compounds for counteracting the oxidative stress-induced effects on vascular gene expression, indicating that flavonoids are an excellent source of functional endothelial regulator products. Notably, the more effective flavonoids for KLF-2 up-regulation resulted in the highest values for eNOS expression, showing that the increment of eNOS expression would take place through KLF-2 induction. Structure-activity relationship studies showed that the combinations of substructures on flavonoid skeleton that regulate eNOS expression are made up of the following elements: glycosylation and hydroxylation of C-ring, double bond C2=C3 at C-ring, methoxylation and hydroxylation of B-ring, ketone group in C4 at C-ring and glycosylation in C7 of A-ring, while flavonoid features involved in the reduction of vasoconstrictor ET-1 expression are as follows: double bond C2=C3 at C-ring glycosylation in C7 of A-ring and ketone group in C4 of C-ring.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Antioxidants/metabolism , Endothelium, Vascular/metabolism , Flavonoids/metabolism , Gene Expression Regulation , Kruppel-Like Transcription Factors/agonists , Oxidative Stress , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antioxidants/chemistry , Cell Survival/drug effects , Cells, Cultured , Diet , Endothelin-1/agonists , Endothelin-1/genetics , Endothelin-1/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Flavonoids/chemistry , Gene Expression Profiling , Gene Expression Regulation/drug effects , Glycosylation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrogen Peroxide/toxicity , Hydroxylation , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Molecular Structure , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidants/antagonists & inhibitors , Oxidants/toxicity , Oxidative Stress/drug effects
12.
Development ; 138(22): 4921-30, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22007132

ABSTRACT

Protein kinase A (PKA) is an evolutionarily conserved negative regulator of the hedgehog (Hh) signal transduction pathway. PKA is known to be required for the proteolytic processing event that generates the repressor forms of the Ci and Gli transcription factors that keep target genes off in the absence of Hh. Here, we show that complete loss of PKA activity in the mouse leads to midgestation lethality and a completely ventralized neural tube, demonstrating that PKA is as strong a negative regulator of the sonic hedgehog (Shh) pathway as patched 1 (Ptch1) or suppressor of fused (Sufu). Genetic analysis shows that although PKA is important for production of the repressor form of Gli3, the principal function of PKA in the Shh pathway in neural development is to restrain activation of Gli2. Activation of the Hh pathway in PKA mutants depends on cilia, and the catalytic and regulatory subunits of PKA are localized to a compartment at the base of the primary cilia, just proximal to the basal body. The data show that PKA does not affect cilia length or trafficking of smoothened (Smo) in the cilium. Instead, we find that there is a significant increase in the level of Gli2 at the tips of cilia of PKA-null cells. The data suggest a model in which PKA acts at the base of the cilium after Gli proteins have transited the primary cilium; in this model the sequential movement of Gli proteins between compartments in the cilium and at its base controls accessibility of Gli proteins to PKA, which determines the fates of Gli proteins and the activity of the Shh pathway.


Subject(s)
Body Patterning/genetics , Cilia/genetics , Cyclic AMP-Dependent Protein Kinases/physiology , Kruppel-Like Transcription Factors/antagonists & inhibitors , Neural Tube/embryology , Animals , Body Patterning/physiology , Catalysis , Cells, Cultured , Cilia/metabolism , Cilia/physiology , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Embryo, Mammalian , Hedgehog Proteins/metabolism , Kruppel-Like Transcription Factors/agonists , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C3H , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neural Tube/metabolism , Neural Tube/physiology , Protein Processing, Post-Translational , Signal Transduction , Tissue Distribution , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3
13.
J Biol Chem ; 282(7): 4782-4793, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17178721

ABSTRACT

Krüppel-like factor 5 (KLF5) is a zinc finger-containing transcription factor that regulates proliferation of various cell types, including fibroblasts, smooth muscle cells, and intestinal epithelial cells. To identify proteins that interact with KLF5, we performed a yeast two-hybrid screen of a 17-day mouse embryo cDNA library with KLF5 as bait. The screen revealed 21 preys clustered in four groups as follows: proteins mediating gene expression, metabolism, trafficking, and signaling. Among them was protein inhibitor of activated STAT1 (PIAS1), a small ubiquitin-like modifier (SUMO) ligase that regulates transcription factors through SUMOylation or physical interaction. Association between PIAS1 and KLF5 was verified by co-immunoprecipitation. Structural determination showed that the acidic domain of PIAS1 bound to both the amino- and carboxyl-terminal regions of KLF5 and that this interaction was inhibited by the amino terminus of PIAS1. Indirect immunofluorescence demonstrated that PIAS1 and KLF5 co-localized to the nucleus. Furthermore, the PIAS1-KLF5 complex was co-localized with the TATA-binding protein and was enriched in RNA polymerase II foci. Transient transfection of COS-7 cells by PIAS1 and KLF5 significantly increased the steady-state protein levels of each other. Luciferase reporter and chromatin immunoprecipitation assays showed that PIAS1 significantly activated the promoters of KLF5 and PIAS1 and synergistically increased the transcriptional activity of KLF5 in activating the cyclin D1 and Cdc2 promoters. Importantly, PIAS1 increased the ability of KLF5 to enhance cell proliferation in transfected cells. These results indicate that PIAS1 is a functional partner of KLF5 and enhances the ability of KLF5 to promote proliferation.


Subject(s)
Cell Nucleus/metabolism , Cell Proliferation , Kruppel-Like Transcription Factors/metabolism , Protein Inhibitors of Activated STAT/metabolism , Up-Regulation/physiology , Active Transport, Cell Nucleus/genetics , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , COS Cells , Cell Nucleus/genetics , Chlorocebus aethiops , Cyclin D1/genetics , Cyclin D1/metabolism , Embryo, Mammalian/metabolism , Enzyme Activation/physiology , Kruppel-Like Transcription Factors/agonists , Kruppel-Like Transcription Factors/genetics , Mice , Promoter Regions, Genetic/physiology , Protein Binding/physiology , Protein Inhibitors of Activated STAT/agonists , Protein Inhibitors of Activated STAT/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/metabolism , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , TATA-Box Binding Protein/genetics , TATA-Box Binding Protein/metabolism , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...