Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
PLoS One ; 10(8): e0133751, 2015.
Article in English | MEDLINE | ID: mdl-26241044

ABSTRACT

Genome-scale expression data on the absolute numbers of gene isoforms offers essential clues in cellular functions and biological processes. Smooth muscle cells (SMCs) perform a unique contractile function through expression of specific genes controlled by serum response factor (SRF), a transcription factor that binds to DNA sites known as the CArG boxes. To identify SRF-regulated genes specifically expressed in SMCs, we isolated SMC populations from mouse small intestine and colon, obtained their transcriptomes, and constructed an interactive SMC genome and CArGome browser. To our knowledge, this is the first online resource that provides a comprehensive library of all genetic transcripts expressed in primary SMCs. The browser also serves as the first genome-wide map of SRF binding sites. The browser analysis revealed novel SMC-specific transcriptional variants and SRF target genes, which provided new and unique insights into the cellular and biological functions of the cells in gastrointestinal (GI) physiology. The SRF target genes in SMCs, which were discovered in silico, were confirmed by proteomic analysis of SMC-specific Srf knockout mice. Our genome browser offers a new perspective into the alternative expression of genes in the context of SRF binding sites in SMCs and provides a valuable reference for future functional studies.


Subject(s)
Muscle Proteins/genetics , Myocytes, Smooth Muscle/metabolism , RNA, Messenger/genetics , Serum Response Element/genetics , Serum Response Factor/metabolism , Web Browser , Animals , Binding Sites , Carrier Proteins/genetics , Colon/cytology , Computer Simulation , Gene Library , Genes, Reporter , Green Fluorescent Proteins , Histone Code , Histones/metabolism , Ion Channels/genetics , Jejunum/cytology , Mice , Mice, Knockout , Mice, Transgenic , Organ Specificity , Proteomics , Serum Response Factor/deficiency , Transcriptome
9.
PLoS One ; 10(4): e0124444, 2015.
Article in English | MEDLINE | ID: mdl-25923532

ABSTRACT

The transcriptional activity of the serum response factor (SRF) protein is triggered by its binding to a 10-base-pair DNA consensus sequence designated the CArG box, which is the core sequence of the serum response element (SRE). Sequence-specific recognition of the CArG box by a core domain of 100 amino acid residues of SRF (core-SRF) was asserted to depend almost exclusively on the intrinsic SRE conformation and on the degree of protein-induced SRE bending. Nevertheless, this paradigm was invalidated by a temperature-dependent Raman spectroscopy study of 20-mer oligonucleotides involved in bonding interactions with core-SRF that reproduced both wild type and mutated c-fos SREs. Indeed, the SRE moieties that are complexed with core-SRF exhibit permanent interconversion dynamics between bent and linear conformers. Thus, sequence-specific recognition of the CArG box by core-SRF cannot be explained only in terms of the three-dimensional structure of the SRE. A particular dynamic pairing process discriminates between the wild type and mutated complexes. Specific oscillations of the phosphate charge network of the SRE govern the recognition between both partners rather than an intrinsic set of conformations of the SRE.


Subject(s)
DNA/chemistry , Oligonucleotides/chemistry , Phosphates/chemistry , Serum Response Element/genetics , Serum Response Factor/chemistry , Binding Sites , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Nucleic Acid Conformation , Nucleotide Motifs , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Serum Response Factor/genetics , Spectrum Analysis, Raman , Static Electricity , Thermodynamics , Transcription, Genetic
10.
Br J Pharmacol ; 171(11): 2827-41, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24641548

ABSTRACT

BACKGROUND AND PURPOSE: Relaxin family peptide receptor 3 (RXFP3) is expressed in brain areas important for processing sensory information and feeding, suggesting that it may be a target for anti-anxiety and anti-obesity drugs. We examined the effects of H3 relaxin, the biased agonist H2 relaxin and the antagonist, R3(BΔ23-27)R/I5, on RXFP3 signalling to establish their suitability as tools to assess the physiological roles of RXFP3. EXPERIMENTAL APPROACH: The signalling profile of the RXFP3 ligands was determined using reporter gene assays, multiplexed signalling assays and direct examination of receptor-G protein and receptor-ß-arrestin interactions using BRET. KEY RESULTS: H2 relaxin activated p38MAPK and ERK1/2 with lower efficacy than H3 relaxin, but had similar efficacy for JNK1/2 phosphorylation. H2 or H3 relaxin activation of p38MAPK, JNK1/2 or ERK1/2 involved Pertussis toxin-sensitive G-proteins. R3(BΔ23-27)R/I5 blocked H3 relaxin AP-1 reporter gene activation, but not H2 relaxin AP-1 activation or H3 relaxin NF-κB activation. R3(BΔ23-27)R/I5 activated the SRE reporter, but did not inhibit either H2 or H3 relaxin SRE activation. R3(BΔ23-27)R/I5 blocked H3 relaxin-stimulated p38MAPK and ERK1/2 phosphorylation, but was a weak partial agonist for p38MAPK and ERK1/2 signalling. p38MAPK activation by R3(BΔ23-27)R/I5 was G protein-independent. H3 relaxin-activated RXFP3 interacts with Gαi2 , Gαi3 , Gαo A and Gαo B whereas H2 relaxin or R3(BΔ23-27)R/I5 induce interactions only with Gαi2 or Gαo B . Only H3 relaxin promoted RXFP3/ß-arrestin interactions that were blocked by R3(BΔ23-27)R/I5. CONCLUSION AND IMPLICATIONS: Understanding signalling profile of drugs acting at RXFP3 is essential for development of therapies targeting this receptor.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Relaxin/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CHO Cells , Cricetulus , Cyclic AMP/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , Receptors, G-Protein-Coupled/genetics , Serum Response Element/genetics , Transcription Factor AP-1/genetics
11.
FEBS J ; 280(23): 6097-113, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24102982

ABSTRACT

Mammalian muscle cell differentiation is a complex process of multiple steps for which many of the factors involved have not yet been defined. In a screen to identify the regulators of myogenic cell fusion, we found that the gene for G-protein coupled receptor 56 (GPR56) was transiently up-regulated during the early fusion of human myoblasts. Human mutations in the gene for GPR56 cause the disease bilateral frontoparietal polymicrogyria; however, the consequences of receptor dysfunction on muscle development have not been explored. Using knockout mice, we defined the role of GPR56 in skeletal muscle. GPR56(-/-) myoblasts have decreased fusion and smaller myotube sizes in culture. In addition, a loss of GPR56 expression in muscle cells results in decreases or delays in the expression of myogenic differentiation 1, myogenin and nuclear factor of activated T-cell (NFAT)c2. Our data suggest that these abnormalities result from decreased GPR56-mediated serum response element and NFAT signalling. Despite these changes, no overt differences in phenotype were identified in the muscle of GPR56 knockout mice, which presented only a mild but statistically significant elevation of serum creatine kinase compared to wild-type. In agreement with these findings, clinical data from 13 bilateral frontoparietal polymicrogyria patients revealed mild serum creatine kinase increase in only two patients. In summary, targeted disruption of GPR56 in mice results in myoblast abnormalities. The absence of a severe muscle phenotype in GPR56 knockout mice and human patients suggests that other factors may compensate for the lack of this G-protein coupled receptor during muscle development and that the motor delay observed in these patients is likely not a result of primary muscle abnormalities.


Subject(s)
Cell Fusion , Malformations of Cortical Development/pathology , Muscle Development/physiology , Myoblasts/cytology , NFATC Transcription Factors/metabolism , Receptors, G-Protein-Coupled/physiology , Serum Response Element/genetics , Animals , Blotting, Western , Cell Communication , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Immunoenzyme Techniques , Luciferases/metabolism , Male , Malformations of Cortical Development/genetics , Malformations of Cortical Development/metabolism , Mice , Mice, Knockout , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/metabolism , Myogenin/genetics , Myogenin/metabolism , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
12.
Cell Signal ; 25(6): 1486-97, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23524338

ABSTRACT

Whereas the activation of MAPKs (mitogen activated kinases) and Rho dependant pathways by GPCR (G protein coupled receptors) has been the subject of many studies, its implication in the signalling of olfactory receptors, which constitute the largest GPCR family, has been far less analysed. Using an in vitro heterologous system, we showed that odorant activated ORs activate SRE containing promoters via the ERK pathway. We also demonstrated that RhoA and Rock kinases but not Rac were involved in ORs-induced SRE/SRF activation and that AP1 was activated, via JNK and p38 MAPKinase. Using real time PCR we found that mOR23, RnI7 and CfOR12A07 induced elevated levels of transcription factors ELK-4, srf, c-fos and c-jun mRNAs whereas mOREG induced an elevated transcription levels of c-fos and c-jun mRNA only. We showed also that odorant activated ORs stimulate the downstream MAPKs and Rho pathways in primary cultures of rat olfactory sensory neurons (OSNs). Similar results were also obtained with OE (olfactory epithelium) extracts prepared from rats exposed to odorants in vivo. Finally, we showed the important role of the AKT and MAPK signalling pathways in OSNs survival. Taken together, these data provide direct evidence that the binding of odorants onto their ORs activates the MAPK and Rho signalling pathways that are involved in OSNs survival events. This suggests that these pathways could be implicated in the regulation of OSNs homeostasis.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Receptors, Odorant/metabolism , Serum Response Element/genetics , Transcription Factor AP-1/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Olfactory Receptor Neurons/metabolism , Phosphorylation , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Rats , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Exp Cell Res ; 319(5): 718-30, 2013 Mar 10.
Article in English | MEDLINE | ID: mdl-23318675

ABSTRACT

Muscle spindles are sensory receptors embedded within muscle that detect changes in muscle length. Each spindle is composed of specialized muscle fibers, known as intrafusal muscle fibers, along with the endings of axons from sensory neurons that innervate these muscle fibers. Formation of muscle spindles requires neuregulin1 (NRG1), which is released by sensory axons, activating ErbB receptors in muscle cells that are contacted. In muscle cells, the transcription factor Egr3 is transcriptionally induced by NRG1, which in turn activates various target genes involved in forming the intrafusal fibers of muscle spindles. The signaling relay within the NRG1-ErbB pathway that acts to induce Egr3 is presumably critical for muscle spindle formation but for the most part has not been determined. In the current studies, we examined, using cultured muscle cells, transcriptional regulatory mechanisms by which Egr3 responds to NRG1. We identified a composite regulatory element for the Egr3 gene, consisting adjacent sites that bind cAMP response element binding protein (CREB) and serum response factor (SRF), with a role in NRG1 responsiveness. The SRF element also influences Egr3 basal expression in unstimulated myotubes, and in the absence of the SRF element, the CREB element influences basal expression. We show that NRG1 signaling, to target SRF, acts on the SRF coactivators myocardian-related transcription factor (MRTF)-A and MRTF-B, which are known to activate SRF-mediated transcription, by stimulating their translocation from the cytoplasm to the nucleus. CREB is phosphorylated, which is known to contribute to its activation, in response to NRG1. These results suggest that NRG1 induces expression of the muscle spindle-specific gene Egr3 by stimulating the transcriptional activity of CREB and SRF.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Early Growth Response Protein 3/genetics , Gene Expression Regulation , Muscle Spindles/metabolism , Neuregulin-1/metabolism , Serum Response Factor/metabolism , Animals , Binding Sites , Blotting, Western , Cell Nucleus/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Cyclic AMP Response Element-Binding Protein/genetics , Early Growth Response Protein 3/metabolism , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Spindles/cytology , Neuregulin-1/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Serum Response Element/genetics , Serum Response Factor/genetics
14.
Biochem Biophys Res Commun ; 425(2): 450-5, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22846568

ABSTRACT

Ca(2+) and cAMP are widely used in concert by neurons to relay signals from the synapse to the nucleus, where synaptic activity modulates gene expression required for synaptic plasticity. Neurons utilize different transcriptional regulators to integrate information encoded in the spatiotemporal dynamics and magnitude of Ca(2+) and cAMP signals, including some that are Ca(2+)-responsive, some that are cAMP-responsive and some that detect coincident Ca(2+) and cAMP signals. Because Ca(2+) and cAMP can influence each other's amplitude and spatiotemporal characteristics, we investigated how cAMP acts to regulate gene expression when increases in intracellular Ca(2+) are buffered. We show here that cAMP-mobilizing stimuli are unable to induce expression of the immediate early gene c-fos in hippocampal neurons in the presence of the intracellular Ca(2+) buffer BAPTA-AM. Expression of enzymes that attenuate intracellular IP(3) levels also inhibited cAMP-dependent c-fos induction. Synaptic activity induces c-fos transcription through two cis regulatory DNA elements - the CRE and the SRE. We show here that in response to cAMP both CRE-mediated and SRE-mediated induction of a luciferase reporter gene is attenuated by IP(3) metabolizing enzymes. Furthermore, cAMP-induced nuclear translocation of the CREB coactivator TORC1 was inhibited by depletion of intracellular Ca(2+) stores. Our data indicate that Ca(2+) release from IP(3)-sensitive pools is required for cAMP-induced transcription in hippocampal neurons.


Subject(s)
Calcium/metabolism , Cyclic AMP/metabolism , Hippocampus/metabolism , Inositol/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-fos/genetics , Transcription, Genetic , Active Transport, Cell Nucleus , Animals , Cell Nucleus , Cells, Cultured , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Hippocampus/cytology , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Rats , Rats, Wistar , Serum Response Element/genetics , TOR Serine-Threonine Kinases/metabolism
15.
Endocr J ; 59(10): 867-79, 2012.
Article in English | MEDLINE | ID: mdl-22785235

ABSTRACT

We examined the effects of sex steroids on prolactin promoter activity in rat somatolactotrophic GH3 cells. Both estradiol (E2) and progesterone (P4) were found to inhibit basal prolactin promoter activity, but to potentiate Thyrotropin-releasing hormone (TRH)-induced prolactin promoter activity. P4 had a greater inhibitory effect on basal prolactin promoter activity than E2, and P4 also potentiated TRH-induced prolactin promoter more potently than E2. Combined treatment with E2 and P4 further increased TRH-induced prolactin promoter activity. E2 and P4 also both reduced basal serum response element (SRE) promoter activity, and increased TRH-induced SRE promoter activity. Combination treatment with E2 and P4 reduced basal activity of SRE promoter and increased TRH-induced SRE activity more potently than E2 or P4 alone. In contrast, basal cAMP response element (CRE) promoter activity was not influenced by either E2 or P4, although TRH-induced CRE promoter was potentiated by each of these steroids, and was further increased by E2 and P4 combination treatment. Both E2 and P4 increased TRH-induced extracellular signal-regulated kinase (ERK) phosphorylation; however, intracellular cAMP levels was not influenced by E2 or P4. TRH-induced CRE promoter was inhibited by mitogen-activated protein kinase/ERK kinase (MEK) inhibitor and was increased by overexpression of MEK kinase (MEKK). This study showed that ERK and SRE transcriptional pathways, but not the cAMP/CRE pathway, may be involved in the suppression of basal prolactin promoter activity, whereas both the ERK/SRE and MAP kinase-mediated CRE pathways appear to be involved in the increased transcriptional efficiency of the prolactin promoter induced by TRH stimulation.


Subject(s)
Estradiol/pharmacology , Lactotrophs/metabolism , Progesterone/pharmacology , Prolactin/biosynthesis , Animals , Cell Line, Tumor , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Promoter Regions, Genetic/drug effects , Rats , Serum Response Element/genetics , Serum Response Element/physiology , Thyrotropin-Releasing Hormone/pharmacology
16.
Eur J Pharmacol ; 642(1-3): 37-46, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20541544

ABSTRACT

The pentadecapeptide comprising the 104-118 amino acid sequence of the ilotropin-derived Reg3-related islet neogenesis-associated protein (INGAP-PP) has been implicated in beta cell neogenesis and enhancement of insulin secretion in pancreatic islets. The aim of this study was to investigate intracellular pathways by which INGAP-PP signals in insulin-producing cells. Treatment with INGAP-PP increased insulin secretion and intracellular calcium levels in MIN6 cells. INGAP-PP exposure activated c-Myc, serum and particularly nuclear factor-kappaB (NF-kappaB) response elements in insulin-producing cells (1.7+/-0.1, 1.8+/-0.1, 2.4+/-0.3 for RINm5F, and 1.3+/-0.1, 1.3+/-0.1 and 1.6+/-0.1 fold for MIN6 cells compared to controls, respectively). There was an increase in the proliferation rate of viable cells (162+/-17% for RINm5F and 155+/-13% for MIN6) that was accompanied by an increase in proliferating cell nuclear antigen (PCNA) protein expression (187+/-19% and 170+/-8% for RINm5F and MIN6 cells respectively) following INGAP-PP treatment. INGAP-PP increased the expression of the muscarinic M(3) receptor subtype (169+/-4% for RINm5F and 222+/-20% for MIN6 cells). Activation of multiple serum response elements by foetal calf serum also increased muscarinic M(3) receptor expression (173+/-9% for RINm5F and 140+/-7% for MIN6 cells). The blockade of NF-kappaB signalling pathway strongly decreased muscarinic M(3) receptor expression in response to both stimuli. In summary, a network of intracellular signals that includes activation of c-Myc signalling pathway and increased PCNA expression might be related to the increased proliferation rate of insulin-producing cells following incubation with INGAP-PP. NF-kappaB signalling plays an essential role in controlling the expression of the muscarinic M(3) receptor.


Subject(s)
Acetylcholine/metabolism , Cytokines/pharmacology , Gene Expression Regulation/drug effects , Insulin-Secreting Cells/cytology , Peptide Fragments/pharmacology , Receptor, Muscarinic M3/metabolism , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus/drug effects , Amino Acid Sequence , Animals , Base Sequence , Carbachol/pharmacology , Catalytic Domain , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cytokines/chemistry , Gene Knockdown Techniques , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Molecular Sequence Data , Pancreatitis-Associated Proteins , Peptide Fragments/chemistry , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/genetics , Rats , Reproducibility of Results , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Serum Response Element/genetics , Transcription Factor RelA/deficiency , Transcription Factor RelA/genetics , Up-Regulation/drug effects
17.
Exp Cell Res ; 316(3): 401-11, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19913013

ABSTRACT

It is well established that histamine modulates cell proliferation through the activation of the histamine H1 receptor (H1R), a G protein-coupled receptor (GPCR) that is known to couple to phospholipase C (PLC) activation via Gq. In the present study, we aimed to determine whether H1R activation modulates Rho GTPases, well-known effectors of Gq/G(11)-coupled receptors, and whether such modulation influences cell proliferation. Experiments were carried out in CHO cells stably expressing H1R (CHO-H1R). By using pull-down assays, we found that both histamine and a selective H1R agonist activated Rac and RhoA in a time- and dose-dependent manner without significant changes in the activation of Cdc42. Histamine response was abolished by the H1R antagonist mepyramine, RGS2 and the PLC inhibitor U73122, suggesting that Rac and RhoA activation is mediated by H1R via Gq coupling to PLC stimulation. Histamine caused a marked activation of serum response factor activity via the H1R, as determined with a serum-responsive element (SRE) luciferase reporter, and this response was inhibited by RhoA inactivation with C3 toxin. Histamine also caused a significant activation of JNK which was inhibited by expression of the Rac-GAP beta2-chimaerin. On the other hand, H1R-induced ERK1/2 activation was inhibited by U73122 but not affected by C3 or beta2-chimaerin, suggesting that ERK1/2 activation was dependent on PLC and independent of RhoA or Rac. [(3)H]-Thymidine incorporation assays showed that both histamine and the H1R agonist inhibited cell proliferation in a dose-dependent manner and that the effect was independent of RhoA but partially dependent on JNK and Rac. Our results reveal that functional coupling of the H1R to Gq-PLC leads to the activation of RhoA and Rac small GTPases and suggest distinct roles for Rho GTPases in the control of cell proliferation by histamine.


Subject(s)
Histamine/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Receptors, Histamine H1/metabolism , Signal Transduction/drug effects , Type C Phospholipases/metabolism , rac GTP-Binding Proteins/metabolism , Animals , CHO Cells , Cell Proliferation/drug effects , Clone Cells , Cricetinae , Cricetulus , Enzyme Activation/drug effects , Genes, Reporter , Humans , Luciferases/metabolism , Serum Response Element/genetics , rhoA GTP-Binding Protein/metabolism
18.
Mol Cell Biol ; 29(22): 6018-32, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19737918

ABSTRACT

Vascular fibrosis is a major complication of hypertension and atherosclerosis, yet it is largely untreatable. Natriuretic peptides (NPs) repress fibrogenic activation of vascular smooth muscle cells (VSMCs), but the intracellular mechanism mediating this effect remains undetermined. Here we show that inhibition of RhoA through phosphorylation at Ser188, the site targeted by the NP effector cyclic GMP (cGMP)-dependent protein kinase I (cGK I), is critical to fully exert antifibrotic potential. cGK I(+/-) mouse blood vessels exhibited an attenuated P-RhoA level and concurrently increased RhoA/ROCK signaling. Importantly, cGK I insufficiency caused dynamic recruitment of ROCK into the fibrogenic programs, thereby eliciting exaggerated vascular hypertrophy and fibrosis. Transgenic expression of cGK I-unphosphorylatable RhoA(A188) in VSMCs augmented ROCK activity, vascular hypertrophy, and fibrosis more prominently than did that of wild-type RhoA, consistent with the notion that RhoA(A188) escapes the intrinsic inhibition by cGK I. Additionally, VSMCs expressing RhoA(A188) became refractory to the antifibrotic effects of NPs. Our results identify cGK I-mediated Ser188 phosphorylation of RhoA as a converging node for pro- and antifibrotic signals and may explain how diminished cGMP signaling, commonly associated with vascular malfunction, predisposes individuals to vascular fibrosis.


Subject(s)
Blood Vessels/enzymology , Blood Vessels/pathology , Cyclic GMP-Dependent Protein Kinases/metabolism , Phosphoserine/metabolism , Signal Transduction , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Angiotensin II/pharmacology , Animals , Blood Vessels/drug effects , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/deficiency , Enzyme Activation/drug effects , Fibrosis , Gene Expression Regulation/drug effects , Humans , Hypertrophy , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Mutant Proteins/metabolism , Organ Specificity/drug effects , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Serum Response Element/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects
19.
J Biol Chem ; 284(34): 23125-36, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19542562

ABSTRACT

Smooth muscle-rich tissues respond to mechanical overload by an adaptive hypertrophic growth combined with activation of angiogenesis, which potentiates their mechanical overload-bearing capabilities. Neovascularization is associated with mechanical strain-dependent induction of angiogenic factors such as CCN1, an immediate-early gene-encoded matricellular molecule critical for vascular development and repair. Here we have demonstrated that mechanical strain-dependent induction of the CCN1 gene involves signaling cascades through RhoA-mediated actin remodeling and the p38 stress-activated protein kinase (SAPK). Actin signaling controls serum response factor (SRF) activity via SRF interaction with the myocardin-related transcriptional activator (MRTF)-A and tethering to a single CArG box sequence within the CCN1 promoter. Such activity was abolished in mechanically stimulated mouse MRTF-A(-/-) cells or upon inhibition of CREB-binding protein (CBP) histone acetyltransferase (HAT) either pharmacologically or by siRNAs. Mechanical strain induced CBP-mediated acetylation of histones 3 and 4 at the SRF-binding site and within the CCN1 gene coding region. Inhibition of p38 SAPK reduced CBP HAT activity and its recruitment to the SRF.MRTF-A complex, whereas enforced induction of p38 by upstream activators (e.g. MKK3 and MKK6) enhanced both CBP HAT and CCN1 promoter activities. Similarly, mechanical overload-induced CCN1 gene expression in vivo was associated with nuclear localization of MRTF-A and enrichment of the CCN1 promoter with both MRTF-A and acetylated histone H3. Taken together, these data suggest that signal-controlled activation of SRF, MRTF-A, and CBP provides a novel connection between mechanical stimuli and angiogenic gene expression.


Subject(s)
CREB-Binding Protein/physiology , Cysteine-Rich Protein 61/genetics , DNA-Binding Proteins/physiology , Histone Acetyltransferases/metabolism , Oncogene Proteins, Fusion/physiology , Stress, Mechanical , Trans-Activators/physiology , Acetylation/drug effects , Animals , Base Sequence , Blotting, Western , CREB-Binding Protein/antagonists & inhibitors , CREB-Binding Protein/genetics , Cells, Cultured , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/genetics , Histones/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Molecular Sequence Data , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/genetics , Protein Transport/drug effects , Protein Transport/genetics , Serum Response Element/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Mol Cells ; 26(5): 443-53, 2008 Nov 30.
Article in English | MEDLINE | ID: mdl-18719355

ABSTRACT

The Bric-a-brac, Tramtrack, Broad-complex (BTB) domain is a protein-protein interaction domain that is found in many zinc finger transcription factors. BTB containing proteins play important roles in a variety of cellular functions including regulation of transcription, regulation of the cytoskeleton, protein ubiquitination, angiogenesis, and apoptosis. Here, we report the cloning and characterization of a novel human gene, KLHL31, from a human embryonic heart cDNA library. The cDNA of KLHL31 is 5743 bp long, encoding a protein product of 634 amino acids containing a BTB domain. The protein is highly conserved across different species. Western blot analysis indicates that the KLHL31 protein is abundantly expressed in both embryonic skeletal and heart tissue. In COS-7 cells, KLHL31 proteins are localized to both the nucleus and the cytoplasm. In primary cultures of nascent mouse cardiomyocytes, the majority of endogenous KLHL31 proteins are localized to the cytoplasm. KLHL31 acts as a transcription repressor when fused to GAL4 DNA-binding domain and deletion analysis indicates that the BTB domain is the main region responsible for this repression. Overexpression of KLHL31 in COS-7 cells inhibits the transcriptional activities of both the TPA-response element (TRE) and serum response element (SRE). KLHL31 also significantly reduces JNK activation leading to decreased phosphorylation and protein levels of the JNK target c-Jun in both COS-7 and Hela cells. These results suggest that KLHL31 protein may act as a new transcriptional repressor in MAPK/JNK signaling pathway to regulate cellular functions.


Subject(s)
Muscles/metabolism , Myocardium/metabolism , Proteins/metabolism , Serum Response Element/genetics , Transcription, Genetic , Amino Acid Sequence , Animals , Antibodies/metabolism , Cell Nucleus/metabolism , Conserved Sequence , Embryo, Mammalian/metabolism , Enzyme Activation , Evolution, Molecular , Gene Expression Profiling , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Organ Specificity , Phosphorylation , Proteins/chemistry , Proteins/genetics , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...