Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 212
Filter
1.
Adv Exp Med Biol ; 1395: 243-248, 2022.
Article in English | MEDLINE | ID: mdl-36527644

ABSTRACT

Extracellular acidosis is a characteristic of solid tumours, resulting from hypoxia-induced glycolytic metabolism as well as from the "Warburg effect" (aerobic glycolysis). The acidic environment has shown to affect functional tumour properties (proliferation, migration, invasion) and thus the aim of the study was to identify signalling mechanisms, mediating these pH-dependent effects. Therefore, the serum response factor (Srf) and the activation of the serum response element (SRE) by acidosis were analysed in AT-1 prostate carcinoma cells. Furthermore, the expression of downstream targets of this cascade, namely the early growth response 1 (Egr1), which seems to be involved in tumour proliferation, and the cellular communication network factor 1 (Ccn1), which both contain SRE in their promotor region were examined in two tumour cell lines. Extracellular acidification led to an upregulation of Srf and a functional activation of the SRE. Egr1 expression was increased by acidosis in AT-1 cells whereas hypoxia had a suppressive effect. In experimental tumours, in vivo Egr1 and Ccn1 were also found to be acidosis-dependent. Also, it turned out that pH regulated expression of Egr1 was followed by comparable changes of p21, which is an important regulator of the cell cycle.This study identifies the Srf-SRE signalling cascade and downstream Egr1 and Ccn1 to be acidosis-regulated in vitro and in vivo, potentially affecting tumour progression. Especially linked expression changes of Egr1 and p21 may mediate acidosis-induced effects on cell proliferation.


Subject(s)
Acidosis , Hypoxia , Prostatic Neoplasms , Animals , Humans , Male , Acidosis/genetics , Acidosis/metabolism , Cell Line, Tumor , Cell Proliferation , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/pharmacology , Hypoxia/genetics , Hypoxia/metabolism , Neoplasms, Experimental , Transcriptional Activation , Rats , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Serum Response Element/genetics , Serum Response Element/physiology
2.
Gene ; 821: 146284, 2022 May 05.
Article in English | MEDLINE | ID: mdl-35143939

ABSTRACT

The basic region leucin zipper (bZIP) protein c-Fos constitutes together with other bZIP proteins the AP-1 transcription factor complex. Expression of the c-Fos gene is regulated by numerous extracellular signaling molecules including mitogens, metabolites, and ligands for receptor tyrosine kinases, G protein-coupled receptors, and cytokine receptors. Here, we analyzed the effects of the stimulus-responsive MAP kinases ERK1/2 (extracellular signal-regulated protein kinase), JNK (c-Jun N-terminal protein kinase) and p38 protein kinase on transcription of the c-Fos gene. We used chromatin-integrated c-Fos promoter-luciferase reporter genes containing inactivating point mutations of DNA binding sites for distinct transcription factors. ERK1/2, JNK, and p38 protein kinases were specifically activated following expression of either a mutant of B-Raf, a truncated version of mitogen-activated/extracellular signal responsive kinase kinase kinase-1 (MEKK1), or a mutant of MAP kinase kinase-6 (MKK6), respectively. The results show that the DNA binding sites for serum response factor (SRF) and for the ternary complex factor (TCF) are of major importance for stimulating c-Fos promoter activity by MAP kinases. ERK1/2 and p38-induced stimulation of the c-Fos promoter additionally required the DNA binding site for the transcription factor AP-1. Mutation of the DNA binding site for STAT had no or only a small effect on c-Fos promoter activity. We conclude that MAP kinases do not activate distinct transcription factors involving distinct genetic elements. Rather, these kinases mainly target SRF and TCF proteins, leading to an activation of transcription of the c-Fos gene via the serum response element.


Subject(s)
Proto-Oncogene Proteins c-fos/genetics , Serum Response Factor/metabolism , TCF Transcription Factors/metabolism , Tamoxifen/analogs & derivatives , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Mutation , Promoter Regions, Genetic , Serum Response Element , Tamoxifen/pharmacology , Transcription, Genetic
3.
PLoS One ; 16(3): e0247402, 2021.
Article in English | MEDLINE | ID: mdl-33661949

ABSTRACT

SREBP1 and 2, are cholesterol sensors able to modulate cholesterol-related gene expression responses. SREBPs binding sites are characterized by the presence of multiple target sequences as SRE, NFY and SP1, that can be arranged differently in different genes, so that it is not easy to identify the binding site on the basis of direct DNA sequence analysis. This paper presents a complete workflow based on a one-dimensional Convolutional Neural Network (CNN) model able to detect putative SREBPs binding sites irrespective of target elements arrangements. The strategy is based on the recognition of SRE linked (less than 250 bp) to NFY sequences according to chromosomal localization derived from TF Immunoprecipitation (TF ChIP) experiments. The CNN is trained with several 100 bp sequences containing both SRE and NF-Y. Once trained, the model is used to predict the presence of SRE-NFY in the first 500 bp of all the known gene promoters. Finally, genes are grouped according to biological process and the processes enriched in genes containing SRE-NFY in their promoters are analyzed in details. This workflow allowed to identify biological processes enriched in SRE containing genes not directly linked to cholesterol metabolism and possible novel DNA patterns able to fill in for missing classical SRE sequences.


Subject(s)
CCAAT-Binding Factor , DNA-Binding Proteins , DNA , Models, Genetic , Sequence Analysis, DNA , Serum Response Element , Sp1 Transcription Factor , Transcription Factors , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
J Cell Physiol ; 236(5): 3565-3578, 2021 05.
Article in English | MEDLINE | ID: mdl-33044017

ABSTRACT

A need for new antidepressants is necessary since traditional antidepressants have several flaws. Neuropeptide Y(NPY) Y1 receptor (NPYY1R) and galanin (GAL) receptor 2 (GALR2) interact in several regions of the limbic system, including the hippocampus. The current study assesses the antidepressant effects induced by GALR2 and NPYY1R coactivation, together with the evaluation of cell proliferation through 5-Bromo-2'-deoxyuridine expression within the dentate gyrus of the ventral hippocampus (vDG). We employed in situ proximity ligation assay to manifest GALR2/NPYY1R heteroreceptor complexes. Additionally, the expression pattern of GALR2 and the activation of the extracellular-regulated kinases (ERK) pathway after GALR2 and NPYY1R costimulation in cell cultures were examined. GALR2 and NPYY1R coactivation resulted in sustained antidepressant behaviors in the FST after 24 h, linked to increased cell proliferation in the vDG. Moreover, an increased density of GALR2/NPYY1R heteroreceptor complexes was observed in vDG, on doublecortin-expressing neuroblasts. Recruitment of the GALR2 expression to the plasma membrane was observed upon the coactivation of GALR2 and NPYY1R in cell cultures, presumably associated to the enhanced effects on the activation of ERK pathway. GALR2 may promote the GALR2/NPYY1R heteroreceptor complexes formation in the ventral hippocampus. It may induce a transformation of cell proliferation toward a neuronal lineage by enhancement of ERK pathway. Thus, it may give the mechanism for the antidepressant behavior observed. These results may provide the basis for the development of heterobivalent agonist pharmacophores, targeting GALR2/NPYY1R heteromers, especially in the neuronal precursor cells of the dentate gyrus in the ventral hippocampus for the novel treatment of depression.


Subject(s)
Antidepressive Agents/metabolism , Dentate Gyrus/metabolism , Galanin/metabolism , Neural Stem Cells/metabolism , Neuropeptide Y/metabolism , Animals , Behavior, Animal , Bromodeoxyuridine/metabolism , Cell Membrane/metabolism , Cell Proliferation , Cells, Cultured , Doublecortin Domain Proteins , Doublecortin Protein , Endocytosis , MAP Kinase Signaling System , Male , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Rats, Sprague-Dawley , Receptor, Galanin, Type 2/metabolism , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/metabolism , Serum Response Element/genetics , Swimming
5.
FASEB J ; 34(5): 6271-6283, 2020 05.
Article in English | MEDLINE | ID: mdl-32162409

ABSTRACT

Vascular smooth muscle cells (VSMCs) in the normal arterial media continually express contractile phenotypic markers which are reduced dramatically in response to injury. Tripartite motif-containing proteins are a family of scaffold proteins shown to regulate gene silencing, cell growth, and differentiation. We here investigated the biological role of tripartite motif-containing 28 (TRIM28) and tripartite motif-containing 27 (TRIM27) in VSMCs. We observed that siRNA-mediated knockdown of TRIM28 and TRIM27 inhibited platelet-derived growth factor (PDGF)-induced migration in human VSMCs. Both TRIM28 and TRIM27 can regulate serum response element activity and were required for maintaining the contractile gene expression in human VSMCs. At the same time, TRIM28 and TRIM27 knockdown reduced the expression of PDGF receptor-ß (PDGFRß) and the phosphorylation of its downstream signaling components. Immunoprecipitation showed that TRIM28 formed complexes with TRIM27 through its N-terminal RING-B boxes-Coiled-Coil domain. Furthermore, TRIM28 and TRIM27 were shown to be upregulated and mediate the VSMC contractile marker gene and PDGFRß expression in differentiating human bone marrow mesenchymal stem cells. In conclusion, we identified that TRIM28 and TRIM27 cooperatively maintain the endogenous expression of PDGFRß and contractile phenotype of human VSMCs.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Mesenchymal Stem Cells/cytology , Muscle Contraction , Muscle, Smooth, Vascular/physiology , Nuclear Proteins/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , DNA-Binding Proteins/genetics , Humans , Mesenchymal Stem Cells/metabolism , Muscle, Smooth, Vascular/cytology , Nuclear Proteins/genetics , Phenotype , Receptor, Platelet-Derived Growth Factor beta/genetics , Serum Response Element , Signal Transduction , Tripartite Motif-Containing Protein 28/genetics
6.
Protein Expr Purif ; 168: 105552, 2020 04.
Article in English | MEDLINE | ID: mdl-31866372

ABSTRACT

Neurturin is a potent neurotrophic factor that has been investigated as a potential therapeutic agent for the treatment of neurodegenerative diseases, including Parkinson's disease, and, more recently, for the treatment of type II diabetes. However, purification of neurturin for clinical applications has been hampered by its low solubility in aqueous solutions. Here we describe the development of a scalable manufacturing process for recombinant neurturin from E. coli. inclusion bodies. Neurturin was refolded from solubilized inclusion bodies by fed-batch dilution refolding with a titer of 90 mg per liter refold and a refold yield of 89%. A two-step purification process using cation exchange and hydrophobic interaction chromatography, followed by formulation using tangential flow filtration resulted in an overall process yield of about 56 mg purified neurturin per liter refold. Solubility of neurturin during the purification process was maintained by the addition of 15% (w/v) glycerol to all buffers. For clinical applications and parenteral administration glycerol was replaced by 15% (w/v) sulfobutyl ether-beta-cyclodextrin (i.e. Captisol) in the drug substance formulation buffer. The final purified product had low or undetectable levels of product-related impurities and concentrations of process-related contaminants such as host cell proteins, host cell DNA, endotoxins and Triton X-100 were reduced more than 10,000-fold or below the limit of detection. Bioactivity of purified recombinant neurturin was demonstrated in a cell-based assay by activation of the MAPK signaling pathway.


Subject(s)
Escherichia coli/genetics , Inclusion Bodies/chemistry , Neurturin/genetics , Xylans/chemistry , Cloning, Molecular , Enzyme Stability , Escherichia coli/metabolism , Gene Expression , Genes, Reporter , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Hydrogen-Ion Concentration , Luciferases/genetics , Luciferases/metabolism , Neurturin/chemistry , Neurturin/metabolism , Protein Refolding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serum Response Element/genetics , Temperature , Xylans/metabolism , beta-Cyclodextrins/chemistry
7.
Genes (Basel) ; 10(12)2019 12 02.
Article in English | MEDLINE | ID: mdl-31810288

ABSTRACT

The EphB4 gene encodes for a transmembrane tyrosine kinase receptor involved in embryonic blood vessel differentiation and cancer development. Although EphB4 is known to be regulated at the post-translational level, little is known about its gene regulation. The present study describes the core promoter elements' identification and cloning, the cis-regulatory elements' mapping and the serum regulation of the human EphB4 gene promoter region. Using bioinformatic analysis, Sanger sequencing and recombinant DNA technology, we analyzed the EphB4 gene upstream region spanning +40/-1509 from the actual transcription start site (TSS) and proved it to be a TATA-less gene promoter with dispersed regulatory elements characterized by a novel motif-of-ten element (MTE) at positions +18/+28, and a DPE-like motif and a DPE-like-repeated motif (DRM) spanning nt +27/+30 and +32 +35, respectively. We also mapped both proximal (multiple Sp1) and distal (HoxA9) trans-activating/dispersed cis-acting transcription factor (TF)-binding elements on the region we studied and used a transient transfection reporter assay to characterize its regulation by serum and IGF-II using EphB4 promoter deletion constructs with or without the identified new DNA-binding elements. Altogether, these findings shed new light on the human EphB4 promoter structure and regulation, suggesting mechanistic features conserved among Pol-II TATA-less genes phylogenetically shared from Drosophila to Human genomes.


Subject(s)
Gene Expression Regulation , Nucleotide Motifs , Receptor, EphB4 , Serum Response Element , Trans-Activators , Transcription, Genetic , Animals , Cell Line , Cloning, Molecular , Drosophila , Humans , Mice , Phylogeny , Receptor, EphB4/biosynthesis , Receptor, EphB4/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
8.
Chin J Physiol ; 62(2): 63-69, 2019.
Article in English | MEDLINE | ID: mdl-31243176

ABSTRACT

One of the principal signaling pathway outcomes from brain-derived neurotrophic factor (BDNF) is the activation of antiapoptotic pathways. In addition to the role of extracellular signal-regulated kinase 1/2 and phosphatidylinositol-3 kinase, BDNF activates protein kinase CK2 to mediate its neuroprotective effect. The inhibition of CK2 activity has been shown to induce apoptosis. Although serum response element (SRE)-mediated transcription has been reported to be activated by BDNF and that the phosphorylation of serum response factor (SRF) by CK2 has been shown to enhance its DNA binding activity, the biological relevance of these interactions remains largely unclear. In the present study, we found that SRE-mediated transcription, CK2 activity, and SRF phosphorylation increased in PC12 cells under BDNF treatment. The transfection of CK2α siRNA blocked the enhancing effect of BDNF on SRE-mediated transcription, SRF phosphorylation, and Mcl-1 gene expression. Moreover, the blockade of CK2 diminished the antiapoptotic effects of BDNF on SRE-mediated transcription, Mcl-1 gene expression, and cell viability under rotenone-induced cytotoxicity. Our data may assist in the development of therapeutic strategies for inhibiting apoptosis during neurodegeneration.


Subject(s)
Casein Kinase II/metabolism , Animals , Brain-Derived Neurotrophic Factor , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Rats , Serum Response Element , Signal Transduction
9.
Cell Signal ; 61: 93-107, 2019 09.
Article in English | MEDLINE | ID: mdl-31100317

ABSTRACT

The Rho family small GTPases mediate cell responses through actin cytoskeletal rearrangement. We previously reported that PLEKHG2, a Rho-specific guanine nucleotide exchange factor, is regulated via interaction with several proteins. We found that PLEKHG2 interacted with non-receptor tyrosine kinase ABL1, but the cellular function remains unclear. Here, we show that the interaction between PLEKHG2 and ABL1 attenuated the PLEKHG2-induced serum response element-dependent gene transcription in a tyrosine phosphorylation-independent manner. PLEKHG2 and ABL1 were co-localized and accumulated within cells co-expressing PLEKHG2 and ABL1. The cellular fractionation analysis suggested that the accumulation involved actin cytoskeletal reorganization. We also revealed that the co-expression of PLEKHG2 with ABL1, but not BCR-ABL, suppressed cell growth and synergistically enhanced NF-κB-dependent gene transcription. The cell growth suppression was canceled by co-expression with IκBα, a member of the NF-κB inhibitor protein family. This study suggests that the interaction between PLEKHG2 and ABL1 suppresses cell growth through intracellular protein accumulation via the NF-κB signaling pathway.


Subject(s)
Cell Proliferation/genetics , Guanine Nucleotide Exchange Factors/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Signal Transduction/genetics , Actin Cytoskeleton/metabolism , Actins/metabolism , Fusion Proteins, bcr-abl/metabolism , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation/genetics , Protein Aggregates/genetics , Protein Binding/genetics , Proto-Oncogene Proteins c-abl/genetics , Serum Response Element/genetics , Transcription, Genetic/genetics , Transfection
10.
Small GTPases ; 10(5): 361-366, 2019 09.
Article in English | MEDLINE | ID: mdl-28489964

ABSTRACT

PLEKHG2 is a Gßγ- and Gαs-dependent guanine nucleotide exchange factor for Rac1 and Cdc42 small GTPases and has been shown to mediate signaling pathways such as those for actin cytoskeletal reorganization and serum response element (SRE)-dependent gene transcription. We have shown that the four-and-a-half LIM domains (FHL) 1 acts as a positive regulator of PLEKHG2. Here, we evaluated the other FHL family members and found that the FHL1A specifically regulate the PLEKHG2 activity. Moreover, FHL1A further enhanced Gßγ- and PLEKHG2-induced SRE-dependent gene transcription, whereas FHL1A partially restored the attenuated PLEKHG2-induced SRE-dependent gene transcription by Gαs. Our results suggest that FHL1A specifically interacts with PLEKHG2 to regulate a function of PLEKHG2 that is modified by the interaction of Gßγ and Gαs.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Serum Response Element , Transcription, Genetic , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Muscle Proteins/genetics , Protein Domains , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Front Immunol ; 9: 1906, 2018.
Article in English | MEDLINE | ID: mdl-30197642

ABSTRACT

Arachidonate 15-lipoxygenase (ALOX15) and arachidonate 15-lipoxygenase, type B (ALOX15B) catalyze the dioxygenation of polyunsaturated fatty acids and are upregulated in human alternatively activated macrophages (AAMs) induced by Th2 cytokine interleukin-4 (IL-4) and/or interleukin-13. Known primarily for roles in bioactive lipid mediator synthesis, 15-lipoxygenases (15-LOXs) have been implicated in various macrophage functions including efferocytosis and ferroptosis. Using a combination of inhibitors and siRNAs to suppress 15-LOX isoforms, we studied the role of 15-LOXs in cellular cholesterol homeostasis and immune function in naïve and AAMs. Silencing or inhibiting the 15-LOX isoforms impaired sterol regulatory element binding protein (SREBP)-2 signaling by inhibiting SREBP-2 processing into mature transcription factor and reduced SREBP-2 binding to sterol regulatory elements and subsequent target gene expression. Silencing ALOX15B reduced cellular cholesterol and the cholesterol intermediates desmosterol, lanosterol, 24,25-dihydrolanosterol, and lathosterol as well as oxysterols in IL-4-stimulated macrophages. In addition, attenuating both 15-LOX isoforms did not generally affect IL-4 gene expression but rather uniquely impacted IL-4-induced CCL17 production in an SREBP-2-dependent manner resulting in reduced T cell migration to macrophage conditioned media. In conclusion, we identified a novel role for ALOX15B, and to a lesser extent ALOX15, in cholesterol homeostasis and CCL17 production in human macrophages.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Chemokine CCL17/biosynthesis , Cholesterol/metabolism , Homeostasis , Macrophages/immunology , Macrophages/metabolism , Arachidonate 15-Lipoxygenase/genetics , Cell Movement/genetics , Cytokines/genetics , Cytokines/metabolism , Gene Expression , Humans , Lipid Metabolism , Protein Binding , RNA, Small Interfering/genetics , Serum Response Element , Sterol Regulatory Element Binding Protein 2/metabolism
12.
Zoolog Sci ; 35(2): 109-114, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29623784

ABSTRACT

Human, mouse, and zebrafish ovarian cancer G protein-coupled receptors (OGR1s) are activated by both metals and extracellular protons. In the present study, we examined whether pig, rat, chicken, and Xenopus OGR1 homologs could sense and be activated by protons and metals. We found that all homologs stimulated serum response element (SRE)-driven promoter activities when they are stimulated by protons. On the other hand, metals differentially activated the homologs. The results using chimeric receptors of human and zebrafish OGR1s indicate that the specificity of the metal-induced activation lies in the extracellular region. These results suggest that protons are an evolutionally conserved agonist of OGR1. However, the types of metals that activated the receptor differed among the homologs.


Subject(s)
Chickens/genetics , Metals/administration & dosage , Protons , Rats/genetics , Receptors, G-Protein-Coupled/genetics , Sus scrofa/genetics , Xenopus/genetics , Animals , Chickens/metabolism , Female , HEK293 Cells , Humans , Ovarian Neoplasms/genetics , Rats/metabolism , Receptors, G-Protein-Coupled/metabolism , Serum Response Element/drug effects , Sus scrofa/metabolism , Xenopus/metabolism
13.
J Cell Physiol ; 233(2): 1005-1016, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28407230

ABSTRACT

Sterol regulatory element binding protein 1 (SREBP-1) is well-known as the master regulator of lipogenesis in rodents. Acyl-CoA synthetase short-chain family member 2 (ACSS2) plays a key role in lipogenesis by synthesizing acetyl-CoA from acetate for lipogenesis. ATP citrate lyase (ACLY) catalyzes the conversion of citrate and coenzyme A to acetyl-CoA, hence, it is also important for lipogenesis. Although ACSS2 function in cancer cells has been elucidated, its essentiality in ruminant mammary lipogenesis is unknown. Furthermore, ACSS2 gene promoter and its regulatory mechanisms have not known. Expression of ACSS2 was high in lipid synthesizing tissues, and its expression increased during lactation compared with non-lactating period. Simultaneous knockdown of both ACSS2 and ACLY by siRNA in primary goat mammary epithelial cells decreased (p < 0.05) the mRNA abundance of genes associated with de novo fatty acid synthesis (FASN, ACACA, SCD1) and triacylglycerol (TAG) synthesis (DGAT1, DGAT2, GPAM, and AGPAT6). Genes responsible for lipid droplet formation and secretion (PLIN2 and PLIN3) and fatty acid oxidation (ATGL, HSL, ACOX, and CPT1A) all decreased (p < 0.05) after ACSS2 and ACLY knockdown. Total cellular TAG content and lipid droplet formation also decreased. Use of a luciferase reporter assay revealed a direct regulation of ACSS2 by SREBP-1. Furthermore, SREBP-1 interacted with an SRE (SREBP response element) spanning at -475 to -483 bp on the ACSS2 promoter. Taken together, our results revealed a novel pathway that SREBP-1 may regulate fatty acid and TAG synthesis by regulating the expression of ACSS2.


Subject(s)
Acetate-CoA Ligase/metabolism , Epithelial Cells/enzymology , Fatty Acids/biosynthesis , Lactation , Mammary Glands, Animal/enzymology , Sterol Regulatory Element Binding Protein 1/metabolism , ATP Citrate (pro-S)-Lyase/genetics , ATP Citrate (pro-S)-Lyase/metabolism , Acetate-CoA Ligase/genetics , Animals , Cells, Cultured , Female , Gene Expression Regulation, Enzymologic , Goats , Lipid Droplets/metabolism , Lipogenesis/genetics , Mammary Glands, Animal/cytology , Mutagenesis, Site-Directed , Mutation , Promoter Regions, Genetic , RNA Interference , Serum Response Element , Sterol Regulatory Element Binding Protein 1/genetics , Transfection , Triglycerides/biosynthesis
14.
Mol Cell Endocrinol ; 472: 126-139, 2018 09 05.
Article in English | MEDLINE | ID: mdl-29225069

ABSTRACT

Stimulation of pancreatic ß-cells with glucose activates the protein kinases B-Raf and extracellular signal-regulated protein kinase that participate in glucose sensing. Inhibition of both kinases results in impairment of glucose-regulated gene transcription. To analyze the signaling pathway controlled by B-Raf, we expressed a conditionally active form of B-Raf in INS-1 insulinoma cells. Here, we show that stimulation of B-Raf strongly activated the transcription factor AP-1 which is accompanied by increased c-Jun and c-Fos promoter activities, an upregulation of c-Jun and c-Fos biosynthesis, and elevated transcriptional activation potentials of c-Jun and c-Fos. Mutational analysis identified the AP-1 sites within the c-Jun promoter and the serum response element (SRE) within the c-Fos promoter as the essential genetic elements connecting B-Raf stimulation with AP-1 activation. In line with this, the transcriptional activation potential of the SRE-binding protein Elk-1 was increased following B-Raf activation. The signal pathway from B-Raf to AP-1 required the activation of c-Jun. We identified the cyclin D1 gene as a delayed response gene for AP-1 following stimulation of B-Raf in insulinoma cells. Moreover, MAP kinase phosphatase-1 and the Ca2+/calmodulin-dependent protein phosphatase calcineurin were identified to function as shut-off-devices for the signaling cascade connecting B-Raf stimulation with the activation of AP-1. The fact that stimulation with glucose, activation of L-type voltage-gated Ca2+ channels, and stimulation of B-Raf all trigger an activation of AP-1 indicates that AP-1 is a point of convergence of signaling pathways in ß-cell.


Subject(s)
Insulinoma/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , Transcription Factor AP-1/genetics , Transcription, Genetic , Up-Regulation/genetics , Animals , Calcineurin/metabolism , Calcium Channels/metabolism , Cell Line, Tumor , Cyclin D1/genetics , Dual Specificity Phosphatase 1/metabolism , Gene Expression Regulation, Neoplastic , Models, Biological , Mutation/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Rats , Serum Response Element/genetics , Transcription Factor AP-1/metabolism , Transcriptional Activation/genetics , ets-Domain Protein Elk-1/metabolism
15.
Biol Reprod ; 96(5): 1043-1051, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28863434

ABSTRACT

We examined direct effect of kisspeptin on pituitary gonadotrophs. Kisspeptin-10 (KP10) significantly increased the promoter activities of the gonadotropin subunits, common alpha-glycoprotein (Cga), luteinizing hormone beta (Lhb), and follicle-stimulatinghormone beta (Fshb) in LbetaT2 cells overexpressing kisspeptin receptor (Kiss1r). KP10 and gonadotropin-releasing hormone (GnRH) increased gonadotropin subunit levels to similar degrees and combined treatment with GnRH and KP10 did not potentiate their individual effects. Adenylate cyclase-activating polypeptide 1 (ADCYAP1) also stimulates all three gonadotropin subunits. When cells were stimulated with both KP10 and ADCYAP1, expression of gonadotropin subunits was further increased compared to KP10 or ADCYAP1 alone. KP10 and GnRH dramatically increased serum response element (Sre) promoter levels but only slightly increased cAMP response element (Cre) promoter levels. Combined stimulation with KP10 and GnRH further increased Sre promoter levels. In contrast, ADCYAP1 slightly increased Sre promoter expression but did not modify the effect of KP10. However, ADCYAP1 increased Cre promoter to greater levels than KP10 alone, and combined treatment with KP10 and ADCYAP1 further increased Cre promoter expression. KP10 increased the expression of ADCYAP1 type I receptor (Adcyap1r) and the basal activity of the Cga promoter was increased at a higher Adcyap1r transfection level. The KP10-induced fold increase in all three gonadotropin subunit promoters was not altered by transfection with a higher amount of Adcyap1r vector. Our findings using model cells show that distinct signaling activation by ADCYAP1 potentiates the action of KP10. We also found that KP10 increases Adcyap1r expression.


Subject(s)
Kisspeptins/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Follicle Stimulating Hormone, beta Subunit/metabolism , Gene Expression Regulation/genetics , Gonadotrophs/metabolism , Gonadotropin-Releasing Hormone/metabolism , Humans , Kisspeptins/genetics , Luteinizing Hormone, beta Subunit/metabolism , Mice , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Plasmids , Promoter Regions, Genetic , Receptors, Kisspeptin-1/genetics , Serum Response Element/genetics
16.
Bioorg Med Chem Lett ; 27(8): 1744-1749, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28285914

ABSTRACT

We recently reported the development of a novel inhibitor of Rho-mediated gene transcription (1, CCG-203971) that is efficacious in multiple animal models of acute fibrosis, including scleroderma, when given intraperitoneally. The modest in vivo potency and poor pharmacokinetics (PK) of this lead, however, make it unsuitable for long term efficacy studies. We therefore undertook a systematic medicinal chemistry effort to improve both the metabolic stability and the solubility of 1, resulting in the identification of two analogs achieving over 10-fold increases in plasma exposures in mice. We subsequently showed that one of these analogs (8f, CCG-232601) could inhibit the development of bleomycin-induced dermal fibrosis in mice when administered orally at 50mg/kg, an effect that was comparable to what we had observed earlier with 1 at a 4-fold higher IP dose.


Subject(s)
Nipecotic Acids/pharmacokinetics , Nipecotic Acids/therapeutic use , Rho Factor/antagonists & inhibitors , Scleroderma, Systemic/drug therapy , Skin/drug effects , Transcriptional Activation/drug effects , Administration, Oral , Animals , Disease Models, Animal , Fibrosis , HEK293 Cells , Humans , Mice , Nipecotic Acids/administration & dosage , Nipecotic Acids/chemistry , Rho Factor/metabolism , Scleroderma, Systemic/genetics , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Serum Response Element/drug effects , Skin/metabolism , Skin/pathology , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism
17.
Can J Physiol Pharmacol ; 95(3): 275-280, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28157379

ABSTRACT

Lysophosphatidic acid (LPA), one component of oxidized low-density lipoprotein (ox-LDL), is a potent bioactive phospholipid. Our recent data reveal that LPA induces matricellular protein CCN1 (also known as Cyr61) expression in aortic smooth muscle cells (SMCs) and that CCN1 bridges LPA and integrin signaling pathways leading to SMC migration. Whether and how LPA regulates the transcriptional machinery of the CCN1 gene are unknown. In this study, we found that LPA markedly induces CCN1 mRNA expression in SMCs. Using deleting mutation and reporter gene strategies, we demonstrated regions from -2038 to -1787 and from -101 to +63 of the CCN1 promoter contain the essential regulatory elements. The serum response element (SRE) and cyclic AMP-response element (CRE) are located in these regions. LPA induced time-dependent phosphorylation of serum response factor (SRF) and CRE-binding protein (CREB) in mouse SMCs. Luciferase assays of a series of deleted, mutated CCN1 promoter-reporter gene constructs and dominant negative construct revealed the distal SRE and the proximal CRE in the CCN1 promoter are required for LPA-induced CCN1 gene expression. Our results imply that elevated LPA levels may trigger SMC migration and exacerbate restenosis and atherosclerotic lesions through the induced CCN1, which communicates with a set of plasma membrane proteins and intracellular kinases.


Subject(s)
Cysteine-Rich Protein 61/genetics , Lysophospholipids/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Serum Response Element/drug effects , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Animals , Aorta/drug effects , Aorta/metabolism , Binding Sites , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Cysteine-Rich Protein 61/metabolism , Genes, Reporter , Mice , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serum Response Factor/metabolism , Time Factors , Transfection , Up-Regulation
18.
Cell Signal ; 32: 115-123, 2017 04.
Article in English | MEDLINE | ID: mdl-28108261

ABSTRACT

PLEKHG2 is a Gßγ-dependent guanine nucleotide exchange factor (GEF) for the small GTPases Rac and Cdc42, and has been shown to mediate signalling pathways such as actin cytoskeleton reorganization and serum response element (SRE)-dependent gene transcription. Here we show that the constitutively active mutant of the Gαs subunit significantly attenuated PLEKHG2-induced SRE-mediated gene transcription. Strikingly, we observed that the constitutive activation of endogenous Gαs by treatment with CTx caused a similar inhibitory effect on PLEKHG2-induced activation of SRE. However, both the enforced expression of the catalytic subunit ß of protein kinase A and the treatment with dibutyl-cyclic AMP failed to mimic the inhibitory effect of Gαs on PLEKHG2. Furthermore, the dominant negative mutant of protein kinase A had no effect on PLEKHG2-mediated SRE activation. Performing immunoprecipitation and an in vitro pulldown assay, we found that PLEKHG2 directly interacted with the active form of the Gαs subunit in cells. The interaction between PLEKHG2 and Gαs required the N-terminal region of PLEKHG2, which includes the DH domain, a functional domain of GEF, suggesting that Gαs directly masks the DH domain of PLEKHG2. In a previous study, we reported that Gßγ accelerates PLEKHG2-mediated SRE-dependent gene transcription. Interestingly, Gαs also inhibited the hyperactivation of SRE induced by the co-expression of Gßγ and PLEKHG2; however, Gαs and Gßγ bind to different regions of PLEKHG2. This is the first report to show that PLEKHG2 is a novel effector of Gαs, and is negatively regulated by the Gαs subunit through direct interaction.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , HEK293 Cells , Humans , Models, Biological , Protein Binding , Serum Response Element/genetics , Transcription, Genetic
19.
Biochemistry ; 56(3): 473-486, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28005346

ABSTRACT

GPR55 is a newly deorphanized class A G-protein-coupled receptor that has been implicated in inflammatory pain, neuropathic pain, metabolic disorder, bone development, and cancer. Few potent GPR55 ligands have been identified to date. This is largely due to an absence of information about salient features of GPR55, such as residues important for signaling and residues implicated in the GPR55 signaling cascade. The goal of this work was to identify residues that are key for the signaling of the GPR55 endogenous ligand, l-α-lysophosphatidylinositol (LPI), as well as the signaling of the GPR55 agonist, ML184 {CID 2440433, 3-[4-(2,3-dimethylphenyl)piperazine-1-carbonyl]-N,N-dimethyl-4-pyrrolidin-1-ylbenzenesulfonamide}. Serum response element (SRE) and serum response factor (SRF) luciferase assays were used as readouts for studying LPI and ML184 signaling at the GPR55 mutants. A GPR55 R* model based on the recent δ-opioid receptor (DOR) crystal structure was used to interpret the resultant mutation data. Two residues were found to be crucial for agonist signaling at GPR55, K2.60 and E3.29, suggesting that these residues form the primary interaction site for ML184 and LPI at GPR55. Y3.32F, H(170)F, and F6.55A/L mutation results suggested that these residues are part of the orthosteric binding site for ML184, while Y3.32F and H(170)F mutation results suggest that these two residues are part of the LPI binding pocket. Y3.32L, M3.36A, and F6.48A mutation results suggest the importance of a Y3.32/M3.36/F6.48 cluster in the GPR55 signaling cascade. C(10)A and C(260)A mutations suggest that these residues form a second disulfide bridge in the extracellular domain of GPR55, occluding ligand extracellular entry in the TMH1-TMH7 region of GPR55. Taken together, these results provide the first set of discrete information about GPR55 residues important for LPI and ML184 signaling and for GPR55 activation. This information should aid in the rational design of next-generation GPR55 ligands and the creation of the first high-affinity GPR55 radioligand, a tool that is sorely needed in the field.


Subject(s)
Lysophospholipids/chemistry , Piperazines/chemistry , Pyrrolidines/chemistry , Receptors, G-Protein-Coupled/chemistry , Recombinant Fusion Proteins/chemistry , Serum Response Element , Amino Acid Motifs , Binding Sites , Crystallography, X-Ray , Gene Expression , HEK293 Cells , Humans , Kinetics , Ligands , Lysophospholipids/pharmacology , Molecular Docking Simulation , Mutation , Piperazines/pharmacology , Protein Binding , Pyrrolidines/pharmacology , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid, delta/chemistry , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serum Response Factor/chemistry , Serum Response Factor/genetics , Serum Response Factor/metabolism , Signal Transduction , Glycine max , Structural Homology, Protein , Thermodynamics
20.
J Biol Chem ; 291(48): 25227-25238, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27765816

ABSTRACT

PLEKHG2/FLJ00018 is a Gßγ-dependent guanine nucleotide exchange factor for the small GTPases Rac and Cdc42 and has been shown to mediate the signaling pathways leading to actin cytoskeleton reorganization. Here we showed that the zinc finger domain-containing protein four-and-a-half LIM domains 1 (FHL1) acts as a novel interaction partner of PLEKHG2 by the yeast two-hybrid system. Among the isoforms of FHL1 (i.e. FHL1A, FHL1B, and FHL1C), FHL1A and FHL1B interacted with PLEKHG2. We found that there was an FHL1-binding region at amino acids 58-150 of PLEKHG2. The overexpression of FHL1A but not FHL1B enhanced the PLEKHG2-induced serum response element-dependent gene transcription. The co-expression of FHL1A and Gßγ synergistically enhanced the PLEKHG2-induced serum response element-dependent gene transcription. Increased transcription activity was decreased by FHL1A knock-out with the CRISPR/Cas9 system. Compared with PLEKHG2-expressing cells, the number and length of finger-like protrusions were increased in PLEKHG2-, Gßγ-, and FHL1A-expressing cells. Our results provide evidence that FHL1A interacts with PLEKHG2 and regulates cell morphological change through the activity of PLEKHG2.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Serum Response Element/physiology , Transcription, Genetic/physiology , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Muscle Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL