Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-24634666

ABSTRACT

Guanine nucleotide binding protein (G protein)-coupled receptors (GPCRs) function in complexes with a range of molecules and proteins including ligands, G proteins, arrestins, ubiquitin, and other receptors. Elements of these complexes may interact constitutively or dynamically, dependent upon factors such as ligand binding, phosphorylation, and dephosphorylation. They may also be allosterically modulated by other proteins in a manner that changes temporally and spatially within the cell. Elucidating how these complexes function has been greatly enhanced by biophysical technologies that are able to monitor proximity and/or binding, often in real time and in live cells. These include resonance energy transfer approaches such as bioluminescence resonance energy transfer (BRET) and fluorescence resonance energy transfer (FRET). Furthermore, the use of fluorescent ligands has enabled novel insights into allosteric interactions between GPCRs. Consequently, biophysical approaches are helping to unlock the amazing diversity and bias in G protein-coupled receptor signaling.

2.
Biochemistry ; 53(11): 1753-67, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24559102

ABSTRACT

A gene induction competition assay has recently uncovered new inhibitory activities of two transcriptional cofactors, NELF-A and NELF-B, in glucocorticoid-regulated transactivation. NELF-A and -B are also components of the NELF complex, which participates in RNA polymerase II pausing shortly after the initiation of gene transcription. We therefore asked if cofactors (Cdk9 and ELL) best known to affect paused polymerase could reverse the effects of NELF-A and -B. Unexpectedly, Cdk9 and ELL augmented, rather than prevented, the effects of NELF-A and -B. Furthermore, Cdk9 actions are not blocked either by Ckd9 inhibitors (DRB or flavopiridol) or by two Cdk9 mutants defective in kinase activity. The mode and site of action of NELF-A and -B mutants with an altered NELF domain are similarly affected by wild-type and kinase-dead Cdk9. We conclude that Cdk9 is a new modulator of GR action, that Ckd9 and ELL have novel activities in GR-regulated gene expression, that NELF-A and -B can act separately from the NELF complex, and that Cdk9 possesses activities that are independent of Cdk9 kinase activity. Finally, the competition assay has succeeded in ordering the site of action of several cofactors of GR transactivation. Extension of this methodology should be helpful in determining the site and mode of action of numerous additional cofactors and in reducing unwanted side effects.


Subject(s)
Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Receptors, Glucocorticoid/physiology , Transcriptional Activation/genetics , Animals , COS Cells , Chlorocebus aethiops , Humans , Mutation/genetics , Rats , Receptors, Glucocorticoid/antagonists & inhibitors , Transcription Factors/physiology
3.
J Biol Chem ; 289(11): 7873-83, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24482225

ABSTRACT

Gonadotropin-releasing hormone (GnRH) is secreted in brief pulses that stimulate synthesis and secretion of pituitary gonadotropin hormones and thereby mediate control of reproduction. It acts via G-protein-coupled receptors to stimulate effectors, including ERK. Information could be encoded in GnRH pulse frequency, width, amplitude, or other features of pulse shape, but the relative importance of these features is unknown. Here we examine this using automated fluorescence microscopy and mathematical modeling, focusing on ERK signaling. The simplest scenario is one in which the system is linear, and response dynamics are relatively fast (compared with the signal dynamics). In this case integrated system output (ERK activation or ERK-driven transcription) will be roughly proportional to integrated input, but we find that this is not the case. Notably, we find that relatively slow response kinetics lead to ERK activity beyond the GnRH pulse, and this reduces sensitivity to pulse width. More generally, we show that the slowing of response kinetics through the signaling cascade creates a system that is robust to pulse width. We, therefore, show how various levels of response kinetics synergize to dictate system sensitivity to different features of pulsatile hormone input. We reveal the mathematical and biochemical basis of a dynamic GnRH signaling system that is robust to changes in pulse amplitude and width but is sensitive to changes in receptor occupancy and frequency, precisely the features that are tightly regulated and exploited to exert physiological control in vivo.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Gonadotropin-Releasing Hormone/metabolism , Active Transport, Cell Nucleus , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , HeLa Cells , Hormones/metabolism , Humans , Kinetics , Luciferases/metabolism , Models, Theoretical , Promoter Regions, Genetic , Signal Transduction , Transcriptome
4.
PLoS One ; 8(6): e65885, 2013.
Article in English | MEDLINE | ID: mdl-23762448

ABSTRACT

Arginine vasopressin (AVP) is released from the posterior pituitary and controls water homeostasis. AVP binding to vasopressin V2 receptors (V2Rs) located on kidney collecting duct epithelial cells triggers activation of Gs proteins, leading to increased cAMP levels, trafficking of aquaporin-2 water channels, and consequent increased water permeability and antidiuresis. Typically, loss-of-function V2R mutations cause nephrogenic diabetes insipidus (NDI), whereas gain-of-function mutations cause nephrogenic syndrome of inappropriate antidiuresis (NSIAD). Here we provide further characterization of two mutant V2Rs, R181C and M311V, reported to cause complete and partial NDI respectively, together with a V266A variant, in a patient diagnosed with NSIAD. Our data in HEK293FT cells revealed that for cAMP accumulation, AVP was about 500- or 30-fold less potent at the R181C and M311V mutants than at the wild-type receptor respectively (and about 4000- and 60-fold in COS7 cells respectively). However, in contrast to wild type V2R, the R181C mutant failed to increase inositol phosphate production, while with the M311V mutant, AVP exhibited only partial agonism in addition to a 37-fold potency decrease. Similar responses were detected in a BRET assay for ß-arrestin recruitment, with the R181C receptor unresponsive to AVP, and partial agonism with a 23-fold decrease in potency observed with M311V in both HEK293FT and COS7 cells. Notably, the V266A V2R appeared functionally identical to the wild-type receptor in all assays tested, including cAMP and inositol phosphate accumulation, ß-arrestin interaction, and in a BRET assay of receptor ubiquitination. Each receptor was expressed at comparable levels. Hence, the M311V V2R retains greater activity than the R181C mutant, consistent with the milder phenotype of NDI associated with this mutant. Notably, the R181C mutant appears to be a Gs protein-biased receptor incapable of signaling to inositol phosphate or recruiting ß-arrestin. The etiology of NSIAD in the patient with V266A V2R remains unknown.


Subject(s)
Diabetes Insipidus, Nephrogenic/genetics , Genetic Diseases, X-Linked/genetics , Inappropriate ADH Syndrome/genetics , Mutation , Polymorphism, Genetic , Receptors, Vasopressin/genetics , Animals , Aquaporin 2/genetics , Aquaporin 2/metabolism , Arginine Vasopressin/metabolism , Arrestins/genetics , Arrestins/metabolism , COS Cells , Chlorocebus aethiops , Cyclic AMP/metabolism , Diabetes Insipidus, Nephrogenic/metabolism , Diabetes Insipidus, Nephrogenic/pathology , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Protein alpha Subunits, Gs/metabolism , Gene Expression Regulation , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/pathology , HEK293 Cells , Humans , Inappropriate ADH Syndrome/metabolism , Inappropriate ADH Syndrome/pathology , Inositol Phosphates/metabolism , Receptors, Vasopressin/metabolism , Signal Transduction , beta-Arrestins
5.
PLoS One ; 8(5): e64672, 2013.
Article in English | MEDLINE | ID: mdl-23700486

ABSTRACT

Heteromerization can play an important role in regulating the activation and/or signal transduction of most forms of receptors, including receptor tyrosine kinases (RTKs). The study of receptor heteromerization has evolved extensively with the emergence of resonance energy transfer based approaches such as bioluminescence resonance energy transfer (BRET). Here, we report an adaptation of our Receptor-Heteromer Investigation Technology (Receptor-HIT) that has recently been published as the G protein-coupled receptor (GPCR) Heteromer Identification Technology (GPCR-HIT). We now demonstrate the utility of this approach for investigating RTK heteromerization by examining the functional interaction between the epidermal growth factor (EGF) receptor (EGFR; also known as erbB1/HER1) and heregulin (HRG) receptor 3 (HER3; also known as erbB3) in live HEK293FT cells using recruitment of growth factor receptor-bound protein 2 (Grb2) to the activated receptors. We found that EGFR and HER3 heteromerize specifically as demonstrated by HRG inducing a BRET signal between EGFR/Rluc8 and Grb2/Venus only when HER3 was co-expressed. Similarly, EGF stimulation promoted a specific BRET signal between HER3/Rluc8 and Grb2/Venus only when EGFR was co-expressed. Both EGF and HRG effects on Grb2 interaction are dose-dependent, and specifically blocked by EGFR inhibitor AG-1478. Furthermore, truncation of HER3 to remove the putative Grb2 binding sites appears to abolish EGF-induced Grb2 recruitment to the EGFR-HER3 heteromer. Our results support the concept that EGFR interacts with Grb2 in both constitutive and EGF-dependent manners and this interaction is independent of HER3 co-expression. In contrast, HER3-Grb2 interaction requires the heteromerization between EGFR and HER3. These findings clearly indicate the importance of EGFR-HER3 heteromerization in HER3-mediated Grb2-dependent signaling pathways and supports the central role of HER3 in the diversity and regulation of HER family functioning.


Subject(s)
ErbB Receptors/metabolism , GRB2 Adaptor Protein/metabolism , Protein Multimerization , Receptor, ErbB-3/metabolism , Binding Sites , Fluorescence Resonance Energy Transfer , GRB2 Adaptor Protein/antagonists & inhibitors , HEK293 Cells , Humans , Kinetics , Protein Binding , Protein Interaction Domains and Motifs , Quinazolines/pharmacology , Signal Transduction , Tyrphostins/pharmacology
6.
J Biol Chem ; 288(21): 15167-80, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23558686

ABSTRACT

TTLL5/STAMP (tubulin tyrosine ligase-like family member 5) has multiple activities in cells. TTLL5 is one of 13 TTLLs, has polyglutamylation activity, augments the activity of p160 coactivators (SRC-1 and TIF2) in glucocorticoid receptor-regulated gene induction and repression, and displays steroid-independent growth activity with several cell types. To examine TTLL5/STAMP functions in whole animals, mice were prepared with an internal deletion that eliminated several activities of the Stamp gene. This mutation causes both reduced levels of STAMP mRNA and C-terminal truncation of STAMP protein. Homozygous targeted mutant (Stamp(tm/tm)) mice appear normal except for marked decreases in male fertility associated with defects in progressive sperm motility. Abnormal axonemal structures with loss of tubulin doublets occur in most Stamp(tm/tm) sperm tails in conjunction with substantial reduction in α-tubulin polyglutamylation, which closely correlates with the reduction in mutant STAMP mRNA. The axonemes in other structures appear unaffected. There is no obvious change in the organs for sperm development of WT versus Stamp(tm/tm) males despite the levels of WT STAMP mRNA in testes being 20-fold higher than in any other organ examined. This defect in male fertility is unrelated to other Ttll genes or 24 genes previously identified as important for sperm function. Thus, STAMP appears to participate in a unique, tissue-selective TTLL-mediated pathway for α-tubulin polyglutamylation that is required for sperm maturation and motility and may be relevant for male fertility.


Subject(s)
Carrier Proteins/metabolism , Gene Deletion , Infertility, Male/metabolism , Sperm Motility , Spermatozoa/metabolism , Testis/metabolism , Animals , Carrier Proteins/genetics , Gene Expression Regulation/genetics , Infertility, Male/genetics , Infertility, Male/pathology , Male , Mice , Mice, Mutant Strains , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/metabolism , Nuclear Receptor Coactivator 2/genetics , Nuclear Receptor Coactivator 2/metabolism , Protein Processing, Post-Translational/genetics , Spermatozoa/pathology , Testis/pathology , Tubulin/genetics , Tubulin/metabolism
7.
J Biol Chem ; 287(16): 12952-65, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22371491

ABSTRACT

We have provided the first evidence for specific heteromerization between the α(1A)-adrenoceptor (α(1A)AR) and CXC chemokine receptor 2 (CXCR2) in live cells. α(1A)AR and CXCR2 are both expressed in areas such as the stromal smooth muscle layer of the prostate. By utilizing the G protein-coupled receptor (GPCR) heteromer identification technology on the live cell-based bioluminescence resonance energy transfer (BRET) assay platform, our studies in human embryonic kidney 293 cells have identified norepinephrine-dependent ß-arrestin recruitment that was in turn dependent upon co-expression of α(1A)AR with CXCR2. These findings have been supported by co-localization observed using confocal microscopy. This norepinephrine-dependent ß-arrestin recruitment was inhibited not only by the α(1)AR antagonist Terazosin but also by the CXCR2-specific allosteric inverse agonist SB265610. Furthermore, Labetalol, which is marketed for hypertension as a nonselective ß-adrenoceptor antagonist with α(1)AR antagonist properties, was identified as a heteromer-specific-biased agonist exhibiting partial agonism for inositol phosphate production but essentially full agonism for ß-arrestin recruitment at the α(1A)AR-CXCR2 heteromer. Finally, bioluminescence resonance energy transfer studies with both receptors tagged suggest that α(1A)AR-CXCR2 heteromerization occurs constitutively and is not modulated by ligand. These findings support the concept of GPCR heteromer complexes exhibiting distinct pharmacology, thereby providing additional mechanisms through which GPCRs can potentially achieve their diverse biological functions. This has important implications for the use and future development of pharmaceuticals targeting these receptors.


Subject(s)
Prostate/metabolism , Protein Structure, Quaternary , Receptors, Adrenergic, alpha-1/chemistry , Receptors, Interleukin-8B/chemistry , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Adrenergic alpha-Agonists/pharmacology , Allosteric Regulation/physiology , Animals , Arrestins/metabolism , CHO Cells , Chemokines/metabolism , Cricetinae , HEK293 Cells , Humans , Inositol Phosphates/metabolism , Labetalol/pharmacology , Male , Mice , Mice, Inbred C57BL , Norepinephrine/pharmacology , Prazosin/analogs & derivatives , Prazosin/pharmacology , Receptors, Adrenergic, alpha-1/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, Interleukin-8B/metabolism , beta-Arrestins
8.
Biochem Soc Trans ; 40(1): 273-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22260704

ABSTRACT

GnRH (gonadotropin-releasing hormone) mediates control of reproduction. It is secreted in pulses and acts via intracellular effectors to activate gene expression. Submaximal GnRH pulse frequency can elicit maximal responses, yielding bell-shaped frequency-response curves characteristic of genuine frequency decoders. GnRH frequency decoding is therapeutically important (pulsatile GnRH can drive ovulation in assisted reproduction, whereas sustained activation can treat breast and prostate cancers), but the mechanisms are unknown. In the present paper, we review recent work in this area, placing emphasis on the regulation of transcription, and showing how mathematical modelling of GnRH effects on two effectors [ERK (extracellular-signal-regulated kinase) and NFAT (nuclear factor of activated T-cells)] reveals the potential for genuine frequency decoding as an emergent feature of the GnRH signalling network, rather than an intrinsic feature of a given protein or pathway within it.


Subject(s)
Calcium Signaling , Gonadotropin-Releasing Hormone/physiology , MAP Kinase Signaling System , Algorithms , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Gonadotropin-Releasing Hormone/metabolism , Humans , Models, Biological , NFATC Transcription Factors/metabolism , Protein Transport
9.
J R Soc Interface ; 9(66): 170-82, 2012 Jan 07.
Article in English | MEDLINE | ID: mdl-21676968

ABSTRACT

Gonadotropin-releasing hormone (GnRH) mediates control of reproduction. It is secreted in pulses and acts via intracellular effectors to activate gonadotrophin secretion and gene expression. Sub-maximal GnRH pulse frequency can elicit maximal responses, yielding bell-shaped frequency-response curves characteristic of genuine frequency decoders. GnRH frequency decoding is therapeutically important (pulsatile GnRH can drive ovulation in assisted reproduction whereas sustained activation can treat breast and prostate cancers), but the mechanisms are unknown. Here, we consider the possibility that it is due to convergence of distinct pulsatile signals at the transcriptome. We develop a model that mirrors wet-laboratory data for activation and nuclear translocation of GnRH effectors (extracellular signal regulated kinase and nuclear factors of activated T-cells) and incorporates transcription. The model predicts genuine frequency decoding when two transcription factors (TFs) converge at a cooperative gate, and shows how optimal pulse frequency could reflect TF activation kinetics and affinities. Importantly, this behaviour is revealed as an emergent feature of the network, rather than an intrinsic feature of a given protein or pathway, and since such network topology is extremely common, may well be widespread in biological systems.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Models, Biological , Signal Transduction , Active Transport, Cell Nucleus , Calcium Signaling , Extracellular Signal-Regulated MAP Kinases/metabolism , Gonadotropin-Releasing Hormone/physiology , NFATC Transcription Factors/metabolism , Time Factors
10.
Methods Mol Biol ; 661: 507-24, 2010.
Article in English | MEDLINE | ID: mdl-20812005

ABSTRACT

Gonadotrophin-releasing hormone (GnRH) is a hypothalamic peptide that acts via G(q/11)-coupled 7TM receptors on pituitary gonadotrophs and mediates the central control of reproduction. Recent evidence also indicates that GnRH can affect numerous tissues, but the molecular mechanisms of GnRH receptor stimulation are cell type-specific. Extracellular signal-regulated kinase (ERK) 1 and 2 are key regulators of GnRH function in several cell types, but they also integrate signals from a wide variety of other stimuli. This leads to the obvious question of how specific cellular responses to ERK activation occur, and it is now clear that this is, in part, achieved through strict spatiotemporal control of ERK activity. This means that, in order to infer the function of ERK regulation accurately, multiple readouts for ERK activity, localisation and downstream consequences (e.g. transcriptional activation or cell growth) must be compared simultaneously. Here, we describe some of our findings in the investigation of GnRH signalling to ERK, with particular emphasis on novel, high-content microscopy methods for studying ERK regulation.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Microscopy/methods , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , HeLa Cells , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/deficiency , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/deficiency , Mitogen-Activated Protein Kinase 3/genetics , Phosphoproteins/metabolism , RNA, Small Interfering/genetics , Staining and Labeling , Transfection
11.
J Biol Chem ; 285(32): 24360-71, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20507982

ABSTRACT

Gonadotropin-releasing hormone (GnRH) acts via G-protein-coupled receptors on gonadotrophs to stimulate synthesis and secretion of luteinizing hormone and follicle-stimulating hormone. It is secreted in pulses, and its effects depend on pulse frequency, but decoding mechanisms are unknown. Here we have used an extracellular signal regulated kinase-green fluorescent protein (ERK2-GFP) reporter to monitor GnRH signaling. GnRH caused dose-dependent ERK2-GFP translocation to the nucleus, providing a live-cell readout for activation. Pulsatile GnRH caused dose- and frequency-dependent ERK2-GFP translocation. These responses were rapid and transient, showed only digital tracking, and did not desensitize under any condition tested (dose, frequency, and receptor number varied). We also tested for the effects of cycloheximide (to prevent induction of nuclear-inducible MAPK phosphatases) and used GFP fusions containing ERK mutations (D319N, which prevents docking domain-dependent binding to MAPK phosphatases, and K52R, which prevents catalytic activity). These manipulations had little or no effect on the translocation responses, arguing against a role for MAPK phosphatases or ERK-mediated feedback in shaping ERK activation during pulsatile stimulation. GnRH also caused dose- and frequency-dependent activation of the alpha-gonadotropin subunit-, luteinizing hormone beta-, and follicle-stimulating hormone beta- luciferase reporters, and the latter response was inhibited by ERK1/2 knockdown. Moreover, GnRH caused frequency-dependent activation of an Egr1-luciferase reporter, but the response was proportional to cumulative pulse duration. Our data suggest that frequency decoding is not due to negative feedback shaping ERK signaling in this model.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, LHRH/metabolism , Catalysis , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , Gonadotropin-Releasing Hormone/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Image Processing, Computer-Assisted , Luteinizing Hormone/metabolism , MAP Kinase Signaling System , Models, Biological , Mutation , Signal Transduction
12.
Mol Endocrinol ; 24(2): 423-35, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20009083

ABSTRACT

Gonadotropin-releasing hormone acts via cell surface receptors but most human (h) GnRH receptors (GnRHRs) are intracellular. A membrane-permeant nonpeptide antagonist [(2S)-2-[5-[2-(2-axabicyclo[2.2.2]oct-2-yl)-1,1-dimethy-2-oxoethyl]-2-(3,5-dimethylphenyl)-1H-indol-3-yl]-N-(2-pyridin-4-ylethyl)propan-1-amine (IN3)] increases hGnRHR expression at the surface, apparently by facilitating its exit from the endoplasmic reticulum. Here we have quantified GnRHR by automated imaging in HeLa cells transduced with adenovirus expressing hemagglutinin-tagged GnRHR. Consistent with an intracellular site of action, IN3 increases cell surface hGnRHR, and this effect is not blocked or mimicked by membrane-impermeant peptide antagonists [Ac-D2Nal-D4Cpa-D3Pal-Ser-Tyr-d-Cit-Leu-Arg-Pro-d-Ala-NH(2) (cetrorelix) and antide]. However, when the C-terminal tail of a Xenopus (X) GnRHR was added (h.XGnRHR) to increase expression, both peptides further increased cell surface GnRHR. Cetrorelix also synergized with IN3 to increase expression of hGnRHR and a G-protein coupling-deficient mutant (A261K-hGnRHR). Cetrorelix also increased cell surface expression of hGnRHR, h.XGnRHR, and mouse GnRHR in gonadotrope-lineage LbetaT2 cells, and in HeLa cells it slowed h.XGnRHR internalization (measured by receptor-mediated antihemagglutinin uptake). Thus cetrorelix has effects other than GnRHR blockade; it acts as an inverse agonist in internalization assays, supporting the potential importance of ligand-biased efficacy at GnRHR. We also developed an imaging assay for GnRH function based on Ca(2+)-dependent nuclear translocation of a nuclear factor of activated T cells reporter. Using this in HeLa and LbetaT2 cells, IN3 and cetrorelix behaved as competitive antagonists when coincubated with GnRH, and long-term pretreatment (16 h) with IN3 reduced its effectiveness as an inhibitor whereas pretreatment with cetrorelix increased its inhibitory effect. This distinction between peptide and nonpeptide antagonists may prove important for therapeutic applications of GnRH antagonists.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Membrane/metabolism , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/pharmacology , Indoles/pharmacology , Peptides/pharmacology , Pyridines/pharmacology , Receptors, LHRH/antagonists & inhibitors , Receptors, LHRH/metabolism , Animals , Buserelin/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Gonadotrophs/metabolism , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , HeLa Cells , Humans , Image Processing, Computer-Assisted/methods , Ligands , Mice , Microscopy, Fluorescence , Oligopeptides/pharmacology , Protein Transport/drug effects , Receptors, LHRH/agonists , Receptors, LHRH/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Species Specificity
13.
J Biol Chem ; 284(51): 35746-57, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19858197

ABSTRACT

Gonadotropin-releasing hormone (GnRH) acts via 7 transmembrane region receptors on gonadotrophs to stimulate synthesis and secretion of the luteinizing hormone and follicle-stimulating hormone. It is secreted in pulses, and its effects depend on pulse frequency, but decoding mechanisms are unknown. Here we have used (nuclear factor of activated T-cells 2 (NFAT2)-emerald fluorescent protein) to monitor GnRH signaling. Increasing [Ca(2+)](i) causes calmodulin/calcineurin-dependent nuclear NFAT translocation, a response involving proteins (calmodulins and NFATs) that decode frequency in other systems. Using live cell imaging, pulsatile GnRH caused dose- and frequency-dependent increases in nuclear NFAT2-emerald fluorescent protein, and at low frequency, translocation simply tracked GnRH exposure (albeit with slower kinetics). At high frequency (30-min intervals), failure to return to basal conditions before repeat stimulation caused integrative tracking, illustrating how the relative dynamics of up- and downstream signals can increase efficiency of GnRH action. Mathematical modeling predicted desensitization of GnRH effects on [Ca(2+)](i) and that desensitization would increase with dose, frequency, and receptor number, but no such desensitization was seen in HeLa and/or LbetaT2 cells possibly because pulsatile GnRH did not reduce receptor expression (measured by immunofluorescence). GnRH also caused dose- and frequency-dependent activation of alphaGSU, luteinizing hormone beta, and follicle-stimulating hormone beta luciferase reporters, effects that were blocked by calcineurin inhibition. Pulsatile GnRH also activated an NFAT-responsive luciferase reporter, but this response was directly related to cumulative pulse duration. This together with the lack of desensitization of translocation responses suggests that NFAT may mediate GnRH action but is not a genuine decoder of GnRH pulse frequency.


Subject(s)
Biological Clocks/physiology , Calcium Signaling/physiology , Cell Nucleus/metabolism , Models, Biological , NFATC Transcription Factors/metabolism , Receptors, LHRH/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Biological Clocks/drug effects , Calcineurin/genetics , Calcineurin/metabolism , Calcium Signaling/drug effects , Calmodulin/genetics , Calmodulin/metabolism , Cell Nucleus/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , HeLa Cells , Humans , Mice , NFATC Transcription Factors/genetics , Receptors, LHRH/genetics
14.
Am J Physiol Cell Physiol ; 297(3): C591-600, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19587220

ABSTRACT

Gonadotropin-releasing hormone (GnRH) acts via seven transmembrane receptors to stimulate gonadotropin secretion. Sustained stimulation desensitizes GnRH receptor (GnRHR)-mediated gonadotropin secretion, and this underlies agonist use in hormone-dependent cancers. Since type I mammalian GnRHR do not desensitize, agonist-induced internalization and downregulation may underlie desensitization of GnRH-stimulated gonadotropin secretion; however, research focus has recently shifted to anterograde trafficking, with the finding that human (h)GnRHR are mostly intracellular. Moreover, there is little direct evidence for agonist-induced trafficking of hGnRHR, and whether or not type I mammalian GnRHR show agonist-induced internalization is controversial. Here we use automated imaging to monitor expression and internalization of hemagglutinin (HA)-tagged hGnRHRs, mouse (m) GnRHR, Xenopus (X) GnRHRs, and chimeric receptors (hGnRHR with added XGnRHR COOH tails, h.XGnRHR) expressed by adenoviral transduction in HeLa cells. We find that agonists stimulate downregulation and/or internalization of mGnRHR and XGnRHR, that GnRH stimulates trafficking of hGnRHR and can stimulate internalization or downregulation of hGnRHR when steps are taken to increase cell surface expression (addition of the XGnRHR COOH tail or pretreatment with pharmacological chaperone). Agonist effects on internalization (of h.XGnRHR) and downregulation (of hGnRHR and h.XGnRHR) were not mimicked by a peptide antagonist and were prevented by a mutation that prevents GnRHR signaling, demonstrating dependence on receptor signaling as well as agonist occupancy. Thus agonist-induced internalization and downregulation of type I mammalian GnRHR occurs in HeLa cells, and we suggest that the high throughput imaging systems described here will facilitate study of the molecular mechanisms involved.


Subject(s)
Gene Expression Regulation/drug effects , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Buserelin/pharmacology , Down-Regulation , Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/metabolism , HeLa Cells , Hemagglutinins , Humans , Indoles/pharmacology , Mice , Pyridines/pharmacology , Recombinant Proteins , Signal Transduction , Xenopus
15.
J Biol Chem ; 283(39): 26612-23, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18650424

ABSTRACT

Although many stimuli activate extracellular signal-regulated kinases 1 and 2 (ERK1/2), the kinetics and compartmentalization of ERK1/2 signals are stimulus-dependent and dictate physiological consequences. ERKs can be inactivated by dual specificity phosphatases (DUSPs), notably the MAPK phosphatases (MKPs) and atypical DUSPs, that can both dephosphorylate and scaffold ERK1/2. Using a cell imaging model (based on knockdown of endogenous ERKs and add-back of wild-type or mutated ERK2-GFP reporters), we explored possible effects of DUSPs on responses to transient or sustained ERK2 activators (epidermal growth factor and phorbol 12,13-dibutyrate, respectively). For both stimuli, a D319N mutation (which impairs DUSP binding) increased ERK2 activity and reduced nuclear accumulation. These stimuli also increased mRNA levels for eight DUSPs. In a short inhibitory RNA screen, 12 of 16 DUSPs influenced ERK2 responses. These effects were evident among nuclear inducible MKP, cytoplasmic ERK MKP, JNK/p38 MKP, and atypical DUSP subtypes and, with the exception of the nuclear inducible MKPs, were paralleled by corresponding changes in Egr-1 luciferase activation. Simultaneous removal of all JNK/p38 MKPs or nuclear inducible MKPs revealed them as positive and negative regulators of ERK2 signaling, respectively. The effects of JNK/p38 MKP short inhibitory RNAs were not dependent on protein neosynthesis but were reversed in the presence of JNK and p38 kinase inhibitors, indicating DUSP-mediated cross-talk between MAPK pathways. Overall, our data reveal that a large number of DUSPs influence ERK2 signaling. Together with the known tissue-specific expression of DUSPs and the importance of ERK1/2 in cell regulation, our data support the potential value of DUSPs as targets for drug therapy.


Subject(s)
Cell Nucleus/enzymology , Dual-Specificity Phosphatases/metabolism , MAP Kinase Signaling System/physiology , Models, Biological , Amino Acid Substitution , Dual-Specificity Phosphatases/genetics , Enzyme Activators/pharmacology , HeLa Cells , Humans , Kinetics , MAP Kinase Signaling System/drug effects , Microscopy, Fluorescence/methods , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mutation, Missense , Phosphorylation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL