Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters








Publication year range
1.
Endocrine ; 62(2): 477-486, 2018 11.
Article in English | MEDLINE | ID: mdl-30027434

ABSTRACT

PURPOSE: The importance of androgen receptor variants (AR-Vs) is recognized in prostate cancer. AR-Vs have been the focus of many studies. Expression of AR-Vs has been proposed as a biomarker for resistance to androgen deprivation therapy for metastatic disease. Herein, we show dynamic changes in AR-Vs expression in response to androgen modulation. METHODS: The C4-2B cell line was exposed to low (10-13 M) and high (10-8 M) androgen (dihydrotestosterone, DHT) levels, with or without flutamide. mRNA and protein expression levels were assessed by qPCR and immunohistochemistry, respectively. RESULTS: We demonstrated that high levels of DHT downregulate AR-FL and AR-Vs. Even though AR-Vs did not present ligand-binding domain, thus were not capable of binding to DHT, they present dynamic changes under androgen treatment. Treatment with flutamide alone or in association with low levels of DHT stimulates growth of prostatic cells. CONCLUSIONS: Importantly, we provide evidence that AR-Vs respond differently to androgenic modulation. These findings have implications for a better understanding of the role of AR-Vs in prostate carcinogenesis.


Subject(s)
Androgens/pharmacology , Mutant Proteins , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Male , Mutant Proteins/agonists , Mutant Proteins/genetics , Mutant Proteins/metabolism , Polymorphism, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Isoforms/agonists , Protein Isoforms/metabolism
2.
Mol Endocrinol ; 30(2): 173-88, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26745667

ABSTRACT

Glucocorticoid receptor (GR) gene mutations may cause familial or sporadic generalized glucocorticoid resistance syndrome. Most of the missense forms distribute in the ligand-binding domain and impair its ligand-binding activity and formation of the activation function (AF)-2 that binds LXXLL motif-containing coactivators. We performed molecular dynamics simulations to ligand-binding domain of pathologic GR mutants to reveal their structural defects. Several calculated parameters including interaction energy for dexamethasone or the LXXLL peptide indicate that destruction of ligand-binding pocket (LBP) is a primary character. Their LBP defects are driven primarily by loss/reduction of the electrostatic interaction formed by R611 and T739 of the receptor to dexamethasone and a subsequent conformational mismatch, which deacylcortivazol resolves with its large phenylpyrazole moiety and efficiently stimulates transcriptional activity of the mutant receptors with LBP defect. Reduced affinity of the LXXLL peptide to AF-2 is caused mainly by disruption of the electrostatic bonds to the noncore leucine residues of this peptide that determine the peptide's specificity to GR, as well as by reduced noncovalent interaction against core leucines and subsequent exposure of the AF-2 surface to solvent. The results reveal molecular defects of pathologic mutant receptors and provide important insights to the actions of wild-type GR.


Subject(s)
Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutation/genetics , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/genetics , Amino Acid Motifs , Amino Acid Sequence , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Dexamethasone/pharmacology , HCT116 Cells , Humans , Leucine/chemistry , Ligands , Models, Molecular , Molecular Sequence Data , Mutant Proteins/agonists , Mutant Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Peptides/chemistry , Peptides/metabolism , Pregnatrienes/pharmacology , Protein Binding/drug effects , Protein Structure, Tertiary , Receptors, Glucocorticoid/agonists , Static Electricity , Thermodynamics , Transcription, Genetic/drug effects
3.
J Biol Chem ; 291(7): 3371-84, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26679999

ABSTRACT

Type I interferons serve as the first line of defense against pathogen invasion. Binding of IFNs to its receptors, IFNAR1 and IFNAR2, is leading to activation of the IFN response. To determine whether structural perturbations observed during binding are propagated to the cytoplasmic domain, multiple mutations were introduced into the transmembrane helix and its surroundings. Insertion of one to five alanine residues near either the N or C terminus of the transmembrane domain (TMD) likely promotes a rotation of 100° and a translation of 1.5 Å per added residue. Surprisingly, the added alanines had little effect on the binding affinity of IFN to the cell surface receptors, STAT phosphorylation, or gene induction. Similarly, substitution of the juxtamembrane residues of the TMD with alanines, or replacement of the TMD of IFNAR1 with that of IFNAR2, did not affect IFN binding or activity. Finally, only the addition of 10 serine residues (but not 2 or 4) between the extracellular domain of IFNAR1 and the TMD had some effect on signaling. Bioinformatic analysis shows a correlation between high sequence conservation of TMDs of cytokine receptors and the ability to transmit structural signals. Sequence conservation near the TMD of IFNAR1 is low, suggesting limited functional importance for this region. Our results suggest that IFN binding to the extracellular domains of IFNAR1 and IFNAR2 promotes proximity between the intracellular domains and that differential signaling is a function of duration of activation and affinity of binding rather than specific conformational changes transmitted from the outside to the inside of the cell.


Subject(s)
Interferon-alpha/metabolism , Models, Molecular , Receptor, Interferon alpha-beta/agonists , Signal Transduction , Amino Acid Sequence , Cell Line , Computational Biology , Conserved Sequence , Gene Knockout Techniques , Humans , Kinetics , Mutagenesis, Insertional , Mutant Proteins/agonists , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptide Fragments/agonists , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Receptor, Interferon alpha-beta/chemistry , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
4.
J Biol Chem ; 291(9): 4589-602, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26710849

ABSTRACT

The receptor deleted in colorectal cancer (DCC) mediates the attraction of growing axons to netrin-1 during brain development. In response to netrin-1 stimulation, DCC becomes a signaling platform to recruit proteins that promote axon outgrowth and guidance. The Ras GTPase-activating protein (GAP) p120RasGAP inhibits Ras activity and mediates neurite retraction and growth cone collapse in response to repulsive guidance cues. Here we show an interaction between p120RasGAP and DCC that positively regulates netrin-1-mediated axon outgrowth and guidance in embryonic cortical neurons. In response to netrin-1, p120RasGAP is recruited to DCC in growth cones and forms a multiprotein complex with focal adhesion kinase and ERK. We found that Ras/ERK activities are elevated aberrantly in p120RasGAP-deficient neurons. Moreover, the expression of p120RasGAP Src homology 2 (SH2)-SH3-SH2 domains, which interact with the C-terminal tail of DCC, is sufficient to restore netrin-1-dependent axon outgrowth in p120RasGAP-deficient neurons. We provide a novel mechanism that exploits the scaffolding properties of the N terminus of p120RasGAP to tightly regulate netrin-1/DCC-dependent axon outgrowth and guidance.


Subject(s)
Axons/metabolism , Cerebral Cortex/metabolism , Nerve Growth Factors/metabolism , Neurons/metabolism , Receptors, Cell Surface/agonists , Signal Transduction , Tumor Suppressor Proteins/agonists , Tumor Suppressor Proteins/metabolism , p120 GTPase Activating Protein/metabolism , Amino Acid Substitution , Animals , Cells, Cultured , Cerebral Cortex/cytology , Chickens , DCC Receptor , Embryo, Mammalian/cytology , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , HEK293 Cells , Humans , Mutant Proteins/agonists , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/chemistry , Nerve Growth Factors/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Netrin-1 , Neurons/cytology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Transport , RNA Interference , Rats , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , p120 GTPase Activating Protein/antagonists & inhibitors , p120 GTPase Activating Protein/chemistry , p120 GTPase Activating Protein/genetics
5.
PLoS Biol ; 13(12): e1002318, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26674493

ABSTRACT

Detecting danger is one of the foremost tasks for a neural system. Larval parasitoids constitute clear danger to Drosophila, as up to 80% of fly larvae become parasitized in nature. We show that Drosophila melanogaster larvae and adults avoid sites smelling of the main parasitoid enemies, Leptopilina wasps. This avoidance is mediated via a highly specific olfactory sensory neuron (OSN) type. While the larval OSN expresses the olfactory receptor Or49a and is tuned to the Leptopilina odor iridomyrmecin, the adult expresses both Or49a and Or85f and in addition detects the wasp odors actinidine and nepetalactol. The information is transferred via projection neurons to a specific part of the lateral horn known to be involved in mediating avoidance. Drosophila has thus developed a dedicated circuit to detect a life-threatening enemy based on the smell of its semiochemicals. Such an enemy-detecting olfactory circuit has earlier only been characterized in mice and nematodes.


Subject(s)
Drosophila Proteins/agonists , Drosophila melanogaster/drug effects , Nerve Tissue Proteins/agonists , Olfactory Bulb/drug effects , Receptors, Odorant/agonists , Sensory Receptor Cells/drug effects , Wasps/metabolism , Alkaloids/pharmacology , Animals , Animals, Genetically Modified , Behavior, Animal/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/parasitology , Drosophila melanogaster/physiology , Female , Iridoids/pharmacology , Larva/drug effects , Larva/genetics , Larva/parasitology , Larva/physiology , Mutant Proteins/agonists , Mutant Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Odorants , Olfactory Bulb/metabolism , Oviposition , Protein Isoforms/agonists , Protein Isoforms/metabolism , Pyridines/pharmacology , Receptors, Odorant/genetics , Receptors, Odorant/metabolism , Sensory Receptor Cells/metabolism , Signal Transduction , Terpenes/pharmacology
6.
Crit Rev Biochem Mol Biol ; 50(5): 440-52, 2015.
Article in English | MEDLINE | ID: mdl-26453028

ABSTRACT

Most proteins have multiple functions. Obviously, conventional methods of manipulating the level of the protein of interest in the cell, such as over-expression, knockout or knockdown, affect all of its functions simultaneously. The key advantage of these methods is that over-expression, knockout or knockdown does not require any knowledge of the molecular mechanisms of the function(s) of the protein of interest. The disadvantage is that these approaches are inadequate to elucidate the role of an individual function of the protein in a particular cellular process. An alternative is the use of re-engineered proteins, in which a single function is eliminated or enhanced. The use of mono-functional elements of a multi-functional protein can also yield cleaner answers. This approach requires detailed knowledge of the structural basis of each function of the protein in question. Thus, a lot of preliminary structure-function work is necessary to make it possible. However, when this information is available, replacing the protein of interest with a mutant in which individual functions are modified can shed light on the biological role of those particular functions. Here, we illustrate this point using the example of protein kinases, most of which have additional non-enzymatic functions, as well as arrestins, known multi-functional signaling regulators in the cell.


Subject(s)
Arrestins/metabolism , G-Protein-Coupled Receptor Kinases/metabolism , Models, Molecular , Animals , Arrestins/agonists , Arrestins/chemistry , Arrestins/genetics , Enzyme Activation , G-Protein-Coupled Receptor Kinases/chemistry , G-Protein-Coupled Receptor Kinases/genetics , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Ligands , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction
7.
Int J Toxicol ; 34(3): 242-9, 2015.
Article in English | MEDLINE | ID: mdl-25878193

ABSTRACT

Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates multiple cellular processes. The anticancer drug doxorubicin (DOX) can activate AhR-mediated transcription of target genes. Because DOX in cells activates a DNA damage response involving ataxia telangiectasia-mutated (ATM)-mediated activation of p53, we investigated whether the activation of the p53 in cells by DNA-damaging agents such as DOX or bleomycin could regulate the AhR levels. Here we report that activation of p53 by DNA-damaging agents in human cells increased levels of AhR through a posttranscriptional mechanism. Accordingly, fibroblasts from ATM patients, which are defective in p53 activation, expressed reduced constitutive levels of AhR and treatment of cells with bleomycin did not appreciably increase the AhR levels. Further, activation of p53 in cells stimulated the expression of AhR target genes. In murine cells, activation of p53 reduced the levels of AhR messenger RNA and protein and reduced the expression of AhR target genes. Our observations revealed that activation of p53 in human and murine cells differentially regulates AhR levels.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Mutagens/toxicity , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Teratogens/toxicity , Tumor Suppressor Protein p53/agonists , Animals , Antibiotics, Antineoplastic/adverse effects , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Humans , Ligands , Macrophages/drug effects , Macrophages/metabolism , Mice , Mutant Proteins/agonists , Mutant Proteins/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Protein Stability/drug effects , Receptors, Aryl Hydrocarbon/chemistry , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Species Specificity , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
Chem Commun (Camb) ; 51(17): 3522-5, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25633558

ABSTRACT

A general computational scheme to evaluate the effects of single point mutations on ligand binding is reported. This scheme is applied to characterize agonist binding to the A2A adenosine receptor, and is found to accurately explain how point mutations of different nature affect the binding affinity of a potent agonist.


Subject(s)
Adenosine A2 Receptor Agonists/chemistry , Molecular Dynamics Simulation , Mutant Proteins/agonists , Mutant Proteins/chemistry , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/genetics , Thermodynamics , Adenosine A2 Receptor Agonists/pharmacology , Binding Sites , Humans , Molecular Structure , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation/genetics , Receptor, Adenosine A2A/metabolism
9.
Biochem J ; 466(2): 347-58, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25494937

ABSTRACT

Trans-aconitate methyltransferase regulator (TamR) is a member of the ligand-responsive multiple antibiotic resistance regulator (MarR) family of transcription factors. In Streptomyces coelicolor, TamR regulates transcription of tamR (encoding TamR), tam (encoding trans-aconitate methyltransferase) and sacA (encoding aconitase); up-regulation of these genes promotes metabolic flux through the citric acid cycle. DNA binding by TamR is attenuated and transcriptional derepression is achieved on binding of ligands such as citrate and trans-aconitate to TamR. In the present study, we show that three additional genes are regulated by S. coelicolor TamR. Genes encoding malate synthase (aceB1; SCO6243), malate dehydrogenase (mdh; SCO4827) and isocitrate dehydrogenase (idh; SCO7000) are up-regulated in vivo when citrate and trans-aconitate accumulate, and TamR binds the corresponding gene promoters in vitro, a DNA binding that is attenuated by cognate ligands. Mutations to the TamR binding site attenuate DNA binding in vitro and result in constitutive promoter activity in vivo. The predicted TamR binding sites are highly conserved in the promoters of these genes in Streptomyces species that encode divergent tam-tamR gene pairs, suggesting evolutionary conservation. Like aconitase and trans-aconitate methyltransferase, malate dehydrogenase, isocitrate dehydrogenase and malate synthase are closely related to the citric acid cycle, either catalysing individual reaction steps or, in the case of malate synthase, participating in the glyoxylate cycle to produce malate that enters the citric acid cycle to replenish the intermediate pool. Taken together, our data suggest that TamR plays an important and conserved role in promoting metabolic flux through the citric acid cycle.


Subject(s)
Bacterial Proteins/metabolism , Citric Acid Cycle , Gene Expression Regulation, Bacterial , Methyltransferases/metabolism , Repressor Proteins/metabolism , Streptomyces coelicolor/metabolism , Aconitic Acid/metabolism , Bacterial Proteins/agonists , Bacterial Proteins/genetics , Citric Acid/metabolism , Enzyme Induction , Genes, Reporter , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Ligands , Malate Dehydrogenase/chemistry , Malate Dehydrogenase/genetics , Malate Dehydrogenase/metabolism , Malate Synthase/chemistry , Malate Synthase/genetics , Malate Synthase/metabolism , Methyltransferases/chemistry , Methyltransferases/genetics , Mutant Proteins/agonists , Mutant Proteins/metabolism , Promoter Regions, Genetic , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Response Elements , Species Specificity , Streptomyces coelicolor/enzymology
10.
Biochemistry ; 53(49): 7846-53, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25453233

ABSTRACT

Phenylalanine hydroxylase (PheH), a liver enzyme that catalyzes the hydroxylation of excess phenylalanine in the diet to tyrosine, is activated by phenylalanine. The lack of activity at low levels of phenylalanine has been attributed to the N-terminus of the protein's regulatory domain acting as an inhibitory peptide by blocking substrate access to the active site. The location of the site at which phenylalanine binds to activate the enzyme is unknown, and both the active site in the catalytic domain and a separate site in the N-terminal regulatory domain have been proposed. Binding of catecholamines to the active-site iron was used to probe the accessibility of the active site. Removal of the regulatory domain increases the rate constants for association of several catecholamines with the wild-type enzyme by ∼2-fold. Binding of phenylalanine in the active site is effectively abolished by mutating the active-site residue Arg270 to lysine. The k(cat)/K(phe) value is down 104 for the mutant enzyme, and the K(m) value for phenylalanine for the mutant enzyme is >0.5 M. Incubation of the R270K enzyme with phenylalanine also results in a 2-fold increase in the rate constants for catecholamine binding. The change in the tryptophan fluorescence emission spectrum seen in the wild-type enzyme upon activation by phenylalanine is also seen with the R270K mutant enzyme in the presence of phenylalanine. Both results establish that activation of PheH by phenylalanine does not require binding of the amino acid in the active site. This is consistent with a separate allosteric site, likely in the regulatory domain.


Subject(s)
Models, Molecular , Phenylalanine Hydroxylase/metabolism , Phenylalanine/metabolism , Allosteric Regulation , Allosteric Site , Amino Acid Substitution , Animals , Arginine/chemistry , Catalytic Domain , Enzyme Activation , Kinetics , Mutagenesis, Site-Directed , Mutant Proteins/agonists , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phenylalanine Hydroxylase/chemistry , Phenylalanine Hydroxylase/genetics , Protein Conformation , Protein Interaction Domains and Motifs , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrometry, Fluorescence
11.
Biochemistry ; 53(35): 5613-8, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25148434

ABSTRACT

The G551D cystic fibrosis transmembrane conductance regulator (CFTR) mutation is associated with severe disease in ∼5% of cystic fibrosis patients worldwide. This amino acid substitution in NBD1 results in a CFTR chloride channel characterized by a severe gating defect that can be at least partially overcome in vitro by exposure to a CFTR potentiator. In contrast, the more common ΔF508 mutation is associated with a severe protein trafficking defect, as well as impaired channel function. Recent clinical trials demonstrated a beneficial effect of the CFTR potentiator, Ivacaftor (VX-770), on lung function of patients bearing at least one copy of G551D CFTR, but no comparable effect on ΔF508 homozygotes. This difference in efficacy was not surprising in view of the established difference in the molecular phenotypes of the two mutant channels. Recently, however, it was shown that the structural defect introduced by the deletion of F508 is associated with the thermal instability of ΔF508 CFTR channel function in vitro. This additional mutant phenotype raised the possibility that the differences in the behavior of ΔF508 and G551D CFTR, as well as the disparate efficacy of Ivacaftor, might be a reflection of the differing thermal stabilities of the two channels at 37 °C. We compared the thermal stability of G551D and ΔF508 CFTR in Xenopus oocytes in the presence and absence of CTFR potentiators. G551D CFTR exhibited a thermal instability that was comparable to that of ΔF508 CFTR. G551D CFTR, however, was protected from thermal instability by CFTR potentiators, whereas ΔF508 CFTR was not. These results suggest that the efficacy of VX-770 in patients bearing the G551D mutation is due, at least in part, to the ability of the small molecule to protect the mutant channel from thermal instability at human body temperature.


Subject(s)
Aminophenols/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/agonists , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Mutant Proteins/agonists , Mutant Proteins/chemistry , Quinolones/pharmacology , Amino Acid Substitution , Animals , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Female , Humans , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Oocytes/metabolism , Protein Stability/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Temperature , Xenopus laevis
12.
Br J Pharmacol ; 171(3): 676-87, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24490858

ABSTRACT

BACKGROUND AND PURPOSE: Dysregulation of the thromboxane A2 (TP) receptor, resulting in agonist hypersensitivity and hyper-responsiveness, contributes to exaggerated vasoconstriction in the hypoxic pulmonary artery in neonatal persistent pulmonary hypertension. We previously reported that hypoxia inhibits TP receptor phosphorylation, causing desensitization. Hence, we examined the role of PKA-accessible serine residues in determining TP receptor affinity, using site-directed mutational analysis. EXPERIMENTAL APPROACH: Vasoconstriction to a thromboxane mimetic and phosphorylation of TP receptor serine was examined in pulmonary arteries from neonatal swine with persistent pulmonary hypertension and controls. Effects of hypoxia were determined in porcine and human TP receptors. Human TPα serines at positions 324, 329 and 331 (C-terminal tail) were mutated to alanine and transiently expressed in HEK293T cells. Saturation binding and displacement kinetics of a TP antagonist and agonist were determined in porcine TP, wild-type human TPα and all TP mutants. Agonist-elicited calcium mobilization was determined for each TP mutant, in the presence of a PKA activator or inhibitor, and in hypoxic and normoxic conditions. KEY RESULTS: The Ser324A mutant was insensitive to PKA activation and hypoxia, had a high affinity for agonist and increased agonist-induced calcium mobilization. Ser329A was no different from wild-type TP receptors. Ser331A was insensitive to hypoxia and PKA with a decreased agonist-mediated response. CONCLUSIONS AND IMPLICATIONS: In hypoxic pulmonary hypertension, loss of site-specific phosphorylation of the TP receptor causes agonist hyper-responsiveness. Ser324 is the primary residue phosphorylated by PKA, which regulates TP receptor-agonist interactions. Ser331 mutation confers loss of TP receptor-agonist interaction, regardless of PKA activity.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Persistent Fetal Circulation Syndrome/metabolism , Protein Processing, Post-Translational , Pulmonary Artery/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/agonists , Serine/metabolism , Amino Acid Substitution , Animals , Animals, Newborn , Calcium Signaling/drug effects , Cell Hypoxia , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/chemistry , Enzyme Activators/pharmacology , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Mutant Proteins/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Persistent Fetal Circulation Syndrome/enzymology , Persistent Fetal Circulation Syndrome/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Specific Pathogen-Free Organisms , Sus scrofa , Thromboxane A2/analogs & derivatives , Thromboxane A2/metabolism , Thromboxane A2/pharmacology
13.
J Pharmacol Exp Ther ; 348(2): 346-58, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24307198

ABSTRACT

Ibogaine is a psychoactive indole alkaloid. Its use as an antiaddictive agent has been accompanied by QT prolongation and cardiac arrhythmias, which are most likely caused by human ether a go-go-related gene (hERG) potassium channel inhibition. Therefore, we studied in detail the interaction of ibogaine with hERG channels heterologously expressed in mammalian kidney tsA-201 cells. Currents through hERG channels were blocked regardless of whether ibogaine was applied via the extracellular or intracellular solution. The extent of inhibition was determined by the relative pH values. Block occurred during activation of the channels and was not observed for resting channels. With increasing depolarizations, ibogaine block grew and developed faster. Steady-state activation and inactivation of the channel were shifted to more negative potentials. Deactivation was slowed, whereas inactivation was accelerated. Mutations in the binding site reported for other hERG channel blockers (Y652A and F656A) reduced the potency of ibogaine, whereas an inactivation-deficient double mutant (G628C/S631C) was as sensitive as wild-type channels. Molecular drug docking indicated binding within the inner cavity of the channel independently of the protonation of ibogaine. Experimental current traces were fit to a kinetic model of hERG channel gating, revealing preferential binding of ibogaine to the open and inactivated state. Taken together, these findings show that ibogaine blocks hERG channels from the cytosolic side either in its charged form alone or in company with its uncharged form and alters the currents by changing the relative contribution of channel states over time.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Excitatory Amino Acid Antagonists/pharmacology , Hallucinogens/pharmacology , Ibogaine/pharmacology , Narcotic Antagonists/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Amino Acid Substitution , Binding Sites/drug effects , Cell Line , Cytosol/metabolism , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Excitatory Amino Acid Antagonists/adverse effects , Excitatory Amino Acid Antagonists/chemistry , Hallucinogens/adverse effects , Hallucinogens/chemistry , Humans , Hydrogen-Ion Concentration , Ibogaine/adverse effects , Ibogaine/chemistry , Ion Channel Gating/drug effects , Kinetics , Membrane Potentials/drug effects , Membrane Transport Modulators/pharmacology , Molecular Conformation , Molecular Docking Simulation , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Narcotic Antagonists/adverse effects , Narcotic Antagonists/chemistry , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
14.
Endocr J ; 60(12): 1309-19, 2013.
Article in English | MEDLINE | ID: mdl-24077220

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is a skeletal disorder with progressive heterotopic ossification in skeletal muscle. A mutation causing constitutive activation in a bone morphogenetic protein (BMP) type 1 receptor [ALK2(R206H)] is found in most patients with FOP. However, the details in the heterotopic ossification of muscle in FOP and the role of matrix metalloproteinase-10 (MMP-10) in bone remain to be fully elucidated. In the present study, we investigated the role of MMP-10 in the differentiation of mouse myoblastic C2C12 cells into osteoblasts. MMP-10 was extracted as a factor, whose expression was most extensively enhanced by ALK2 (R206H) transfection in C2C12 cells. MMP-10 significantly augmented the levels of Osterix, type 1 collagen, alkaline phosphatase (ALP) and osteocalcin mRNA as well as ALP activity enhanced by BMP-2 in C2C12 cells. Moreover, a reduction in endogenous MMP-10 levels by siRNA significantly decreased the levels of Runx2, Osterix, type 1 collagen, ALP and osteocalcin mRNA enhanced by BMP-2 in these cells. In addition, MMP-10 increased the phosphorylation of Smad1/5/8 as well as enhanced the levels of Smad6 and Smad7 mRNA induced by BMP-2. In conclusion, the present study first demonstrated that MMP-10 promotes the differentiation of myoblasts into osteoblasts by interacting with the BMP signaling pathway. MMP-10 may play some important role in the heterotopic ossification of muscle in FOP.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Gene Expression Regulation, Enzymologic , Matrix Metalloproteinase 10/metabolism , Osteoblasts/cytology , Signal Transduction , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Amino Acid Substitution , Animals , Bone Morphogenetic Protein 2/agonists , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Line , Collagen Type I/agonists , Collagen Type I/genetics , Collagen Type I/metabolism , Humans , Matrix Metalloproteinase 10/chemistry , Matrix Metalloproteinase 10/genetics , Mice , Mutant Proteins/agonists , Mutant Proteins/metabolism , Myoblasts/cytology , Myoblasts/enzymology , Myoblasts/metabolism , Osteoblasts/enzymology , Osteoblasts/metabolism , Osteocalcin/agonists , Osteocalcin/genetics , Osteocalcin/metabolism , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Smad6 Protein/agonists , Smad6 Protein/genetics , Smad6 Protein/metabolism , Smad7 Protein/agonists , Smad7 Protein/genetics , Smad7 Protein/metabolism , Sp7 Transcription Factor , Transcription Factors/agonists , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Clin Endocrinol Metab ; 98(12): E1909-17, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24106286

ABSTRACT

CONTEXT: Shikonin, which is an active naphthoquinone isolated from traditional Chinese herbal medicine Zi Cao, has been recently developed to use as an antitumor agent in colorectal cancer, melanoma, leukemia, breast cancer, and hepatocellular cancer. However, its antitumor effect in thyroid cancer remains largely unknown. OBJECTIVES: The aim of the study was to test the therapeutic potential of shikonin for thyroid cancer and explore the mechanisms underlying antitumor effects of shikonin. EXPERIMENTAL DESIGN: We examined the effects of shikonin on proliferation, cell cycle, apoptosis, migration, invasion, and xenograft tumor growth in thyroid cancer cells and the effect of shikonin on proliferation of primary thyroid cancer cells. RESULTS: Shikonin inhibited thyroid cancer cell proliferation in a dose- and time-dependent manner and induced cell cycle arrest. Moreover, shikonin induced cell apoptosis through reactive oxygen species-mediated DNA damage and activation of the p53 signaling pathway. Our data also showed that shikonin dramatically inhibited thyroid cancer cell migration and invasion by suppressing epithelial-mesenchymal transition and downregulating expression of Slug and MMP-2, -9, and -14. Further elucidation of the mechanisms involved revealed that shikonin markedly repressed the phosphorylation of Erk and Akt and activated the p16/Retinoblastoma protein (Rb) pathway in thyroid cancer cells. Growth of xenograft tumors derived from the thyroid cancer cell line FTC133 in nude mice was significantly inhibited by shikonin. Importantly, we did not find the effect of shikonin on liver function in mice. CONCLUSION: We for the first time demonstrated that shikonin is a potentially effective antitumor agent for thyroid cancers.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Cyclin-Dependent Kinase Inhibitor p16/agonists , Naphthoquinones/therapeutic use , Retinoblastoma Protein/agonists , Signal Transduction/drug effects , Thyroid Neoplasms/drug therapy , Tumor Suppressor Protein p53/agonists , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Epithelial-Mesenchymal Transition/drug effects , Humans , Inhibitory Concentration 50 , Liver/drug effects , Liver/physiopathology , Mice , Mice, Nude , Mutant Proteins/agonists , Mutant Proteins/metabolism , Naphthoquinones/adverse effects , Naphthoquinones/pharmacology , Neoplasm Invasiveness/prevention & control , Neoplasm Proteins/agonists , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Retinoblastoma Protein/metabolism , Thyroid Neoplasms/pathology , Thyroid Neoplasms/physiopathology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
16.
PLoS Pathog ; 9(7): e1003508, 2013.
Article in English | MEDLINE | ID: mdl-23935486

ABSTRACT

Bacterial populations co-ordinate gene expression collectively through quorum sensing (QS), a cell-to-cell communication mechanism employing diffusible signal molecules. The LysR-type transcriptional regulator (LTTR) protein PqsR (MvfR) is a key component of alkyl-quinolone (AQ)-dependent QS in Pseudomonas aeruginosa. PqsR is activated by 2-alkyl-4-quinolones including the Pseudomonas quinolone signal (PQS; 2-heptyl-3-hydroxy-4(1H)-quinolone), its precursor 2-heptyl-4-hydroxyquinoline (HHQ) and their C9 congeners, 2-nonyl-3-hydroxy-4(1H)-quinolone (C9-PQS) and 2-nonyl-4-hydroxyquinoline (NHQ). These drive the autoinduction of AQ biosynthesis and the up-regulation of key virulence determinants as a function of bacterial population density. Consequently, PqsR constitutes a potential target for novel antibacterial agents which attenuate infection through the blockade of virulence. Here we present the crystal structures of the PqsR co-inducer binding domain (CBD) and a complex with the native agonist NHQ. We show that the structure of the PqsR CBD has an unusually large ligand-binding pocket in which a native AQ agonist is stabilized entirely by hydrophobic interactions. Through a ligand-based design strategy we synthesized and evaluated a series of 50 AQ and novel quinazolinone (QZN) analogues and measured the impact on AQ biosynthesis, virulence gene expression and biofilm development. The simple exchange of two isosteres (OH for NH2) switches a QZN agonist to an antagonist with a concomitant impact on the induction of bacterial virulence factor production. We also determined the complex crystal structure of a QZN antagonist bound to PqsR revealing a similar orientation in the ligand binding pocket to the native agonist NHQ. This structure represents the first description of an LTTR-antagonist complex. Overall these studies present novel insights into LTTR ligand binding and ligand-based drug design and provide a chemical scaffold for further anti-P. aeruginosa virulence drug development by targeting the AQ receptor PqsR.


Subject(s)
Bacterial Proteins/metabolism , Pseudomonas aeruginosa/physiology , Quinolones/metabolism , Quorum Sensing , Signal Transduction , Transcription Factors/metabolism , Alkylation , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/agonists , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Binding Sites , Biofilms/drug effects , Drug Design , Gene Expression Regulation, Bacterial , Ligands , Molecular Conformation , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptide Fragments/agonists , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/pathogenicity , Quinolones/chemistry , Quinolones/pharmacology , Quorum Sensing/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Virulence/drug effects
17.
PLoS Pathog ; 9(7): e1003510, 2013.
Article in English | MEDLINE | ID: mdl-23874206

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) interacts with human dermal endothelial cell surface tyrosine kinase EphrinA2 (EphA2) and integrins (α3ß1 and αVß3) in the lipid raft (LR) region, and EphA2 regulates macropinocytic virus entry by coordinating integrin-c-Cbl associated signaling. In contrast, KSHV enters human foreskin fibroblast (HFF) cells by LR-independent clathrin mediated endocytosis. The present studies conducted to identify the key molecules regulating KSHV entry in HFF cells showed that KSHV induces association with integrins (αVß5, αVß3 and α3ß1) and EphA2 in non-LR regions early during infection and activates EphA2, which in turn associates with phosphorylated c-Cbl, myosin IIA, FAK, Src, and PI3-K, as well as clathrin and its adaptor AP2 and effector Epsin-15 proteins. EphA2 knockdown significantly reduced these signal inductions, virus internalization and gene expression. c-Cbl knockdown ablated the c-Cbl mediated K63 type polyubiquitination of EphA2 and clathrin association with EphA2 and KSHV. Mutations in EphA2's tyrosine kinase domain (TKD) or sterile alpha motif (SAM) abolished its interaction with c-Cbl. Mutations in tyrosine kinase binding (TKB) or RING finger (RF) domains of c-Cbl resulted in very poor association of c-Cbl with EphA2 and decreased EphA2 polyubiquitination. These studies demonstrated the contributions of these domains in EphA2 and c-Cbl association, EphA2 polyubiquitination and virus-EphA2 internalization. Collectively, these results revealed for the first time that EphA2 influences the tyrosine phosphorylation of clathrin, the role of EphA2 in clathrin mediated endocytosis of a virus, and c-Cbl mediated EphA2 polyubiquitination directing KSHV entry in HFF cells via coordinated signal induction and progression of endocytic events, all of which suggest that targeting EphA2 and c-Cbl could block KSHV entry and infection.


Subject(s)
Clathrin-Coated Vesicles/metabolism , Endocytosis , Ephrin-A2/metabolism , Fibroblasts/virology , Herpesvirus 8, Human/physiology , Integrins/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Cells, Cultured , Ephrin-A2/agonists , Ephrin-A2/antagonists & inhibitors , Ephrin-A2/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Viral , HEK293 Cells , Humans , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phosphorylation , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering , Signal Transduction , Ubiquitination , Up-Regulation , Viral Proteins/biosynthesis , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Internalization
18.
Biochem J ; 454(2): 283-93, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23782265

ABSTRACT

The atherogenic cytokine IL-6 (interleukin-6) induces pro-inflammatory gene expression in VECs (vascular endothelial cells) by activating the JAK (Janus kinase)/STAT3 (signal transducer and activator of transcription 3) signalling pathway, which is normally down-regulated by the STAT3-dependent induction of the E3 ubiquitin ligase component SOCS3 (suppressor of cytokine signalling 3). Novel treatments based on the regulation of SOCS3 protein levels could therefore have value in the treatment of diseases with an inflammatory component, such as atherosclerosis. To this end we carried out a screen of 1031 existing medicinal compounds to identify inducers of SOCS3 gene expression and identified the flavanoids naringenin and flavone as effective inducers of SOCS3 protein, mRNA and promoter activity. This was in contrast with the action of traditional JAK/STAT3 inhibitors and the polyphenol resveratrol, which effectively suppress SOCS3 gene expression. Both naringenin and flavone also effectively suppressed IL-6-stimulated phosphorylation of STAT3 (Tyr7°5) which led to suppression of IL-6-induction of the atherogenic STAT3 target gene MCP1 (monocyte chemotactic protein-1), suggesting that their ability to induce SOCS3 gene expression is STAT3-independent. Supporting this idea was the observation that the general kinase inhibitor compound C inhibits flavone- and cAMP-dependent, but not JAK-dependent, SOCS3 induction in VECs. Indeed, the ability of flavanoids to induce SOCS3 expression requires activation of the ERK (extracellular-signal-regulated kinase)-dependent transcription factor SP3, and not STAT3. In the present paper we therefore describe novel molecular actions of flavanoids, which control SOCS3 gene induction and suppression of STAT3 signalling in VECs. These mechanisms could potentially be exploited to develop novel anti-atherogenic therapies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cytokine Receptor gp130/antagonists & inhibitors , Endothelium, Vascular/metabolism , Flavonoids/metabolism , Interleukin-6/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/agonists , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Cell Line , Cells, Cultured , Chlorocebus aethiops , Cytokine Receptor gp130/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Flavonoids/antagonists & inhibitors , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-6/metabolism , Interleukin-6 Receptor alpha Subunit/metabolism , Mice , Mutant Proteins/agonists , Mutant Proteins/metabolism , Promoter Regions, Genetic/drug effects , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/agonists , Recombinant Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
19.
Endocrinology ; 154(9): 3294-304, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23782943

ABSTRACT

Gonadal steroids are potent regulators of adult neurogenesis. We previously reported that androgens, such as testosterone (T) and dihydrotestosterone (DHT), but not estradiol, increased the survival of new neurons in the dentate gyrus of the male rat. These results suggest androgens regulate hippocampal neurogenesis via the androgen receptor (AR). To test this supposition, we examined the role of ARs in hippocampal neurogenesis using 2 different approaches. In experiment 1, we examined neurogenesis in male rats insensitive to androgens due to a naturally occurring mutation in the gene encoding the AR (termed testicular feminization mutation) compared with wild-type males. In experiment 2, we injected the AR antagonist, flutamide, into castrated male rats and compared neurogenesis levels in the dentate gyrus of DHT and oil-treated controls. In experiment 1, chronic T increased hippocampal neurogenesis in wild-type males but not in androgen-insensitive testicular feminization mutation males. In experiment 2, DHT increased hippocampal neurogenesis via cell survival, an effect that was blocked by concurrent treatment with flutamide. DHT, however, did not affect cell proliferation. Interestingly, cells expressing doublecortin, a marker of immature neurons, did not colabel with ARs in the dentate gyrus, but ARs were robustly expressed in other regions of the hippocampus. Together these studies provide complementary evidence that androgens regulate adult neurogenesis in the hippocampus via the AR but at a site other than the dentate gyrus. Understanding where in the brain androgens act to increase the survival of new neurons in the adult brain may have implications for neurodegenerative disorders.


Subject(s)
Androgens/metabolism , Dentate Gyrus/metabolism , Neurogenesis , Neurons/metabolism , Neuroprotective Agents/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Androgen Antagonists/toxicity , Androgen-Insensitivity Syndrome/chemically induced , Androgen-Insensitivity Syndrome/drug therapy , Androgen-Insensitivity Syndrome/metabolism , Androgens/chemistry , Androgens/pharmacology , Androgens/therapeutic use , Animals , Biomarkers/metabolism , Castration/adverse effects , Cell Survival/drug effects , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Doublecortin Domain Proteins , Doublecortin Protein , Drug Resistance , Hormone Replacement Therapy , Male , Microtubule-Associated Proteins/metabolism , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/pathology , Neuropeptides/metabolism , Neuroprotective Agents/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Signal Transduction/drug effects , Testosterone Propionate/antagonists & inhibitors , Testosterone Propionate/pharmacology , Testosterone Propionate/therapeutic use
20.
FEBS J ; 280(16): 3840-53, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23745637

ABSTRACT

The cyclin-dependent kinase inhibitor p21(CIP1/WAF1) is a regulatory factor of the cell cycle. Its transcriptional activation and protein stability are tightly controlled by several distinct mechanisms. S100A11 is a member of the S100 family of Ca²âº-binding proteins involved in several biological processes, including cell cycle progression and signal transduction. In the present study, we show that down-regulation of S100A11 results in the reduction of p21 protein in human HaCaT keratinocytes. It appears that a ubiquitin-independent proteasomal degradation process is involved in p21 degradation in S100A11 down-regulated cells. The application of a proteasome inhibitor stabilized p21 protein in these cells. Analysis of distinct signal transduction pathways revealed a disturbed phosphatidylinositol-3-kinase/Akt pathway after S100A11 knockdown. We determined that the glycogen synthase kinase-3, which is negatively regulated by phosphatidylinositol 3-kinase/Akt, was activated in cells possessing knocked-down S100A11 and appears to be involved in p21 protein destabilization. The application of a specific inhibitor of glycogen synthase kinase 3 resulted in an increase of the p21 protein level in S100A11 down-regulated HaCaT cells. Glycogen synthase kinase 3 is able to phosphorylate p21 at T57, which induces p21 proteasomal turnover. Mutation of the glycogen synthase kinase 3 site threonine 57 into alanine (T57A) stabilizes p21 in HaCaT cells lacking S100A11. Beside decreased p21 protein, down-regulation of S100A11 triggered the induction of apoptosis in HaCaT cells. These observations suggest that S100A11 is involved in the maintenance of p21 protein stability and appears to function as an inhibitor of apoptosis in human HaCaT keratinocyte cells. Thus, the data shed light on a novel pathway regulating p21 protein stability.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation , Keratinocytes/metabolism , S100 Proteins/metabolism , Up-Regulation , Apoptosis/drug effects , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/agonists , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , Down-Regulation/drug effects , Gene Silencing , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/chemistry , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Keratinocytes/drug effects , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Stability/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , S100 Proteins/antagonists & inhibitors , S100 Proteins/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL